51
|
Nellist M, Brouwer RWW, Kockx CEM, van Veghel-Plandsoen M, Withagen-Hermans C, Prins-Bakker L, Hoogeveen-Westerveld M, Mrsic A, van den Berg MMP, Koopmans AE, de Wit MC, Jansen FE, Maat-Kievit AJA, van den Ouweland A, Halley D, de Klein A, van IJcken WFJ. Targeted Next Generation Sequencing reveals previously unidentified TSC1 and TSC2 mutations. BMC MEDICAL GENETICS 2015; 16:10. [PMID: 25927202 PMCID: PMC4422413 DOI: 10.1186/s12881-015-0155-4] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 02/16/2015] [Indexed: 12/24/2022]
Abstract
Background Tuberous sclerosis complex (TSC) is an autosomal dominant disorder caused by mutations in TSC1 and TSC2. Conventional DNA diagnostic screens identify a TSC1 or TSC2 mutation in 75 - 90% of individuals categorised with definite TSC. The remaining individuals either have a mutation that is undetectable using conventional methods, or possibly a mutation in another as yet unidentified gene. Methods Here we apply a targeted Next Generation Sequencing (NGS) approach to screen the complete TSC1 and TSC2 genomic loci in 7 individuals fulfilling the clinical diagnostic criteria for definite TSC in whom no TSC1 or TSC2 mutations were identified using conventional screening methods. Results We identified and confirmed pathogenic mutations in 3 individuals. In the remaining individuals we identified variants of uncertain clinical significance. The identified variants included mosaic changes, changes located deep in intronic sequences and changes affecting promoter regions that would not have been identified using exon-only based analyses. Conclusions Targeted NGS of the TSC1 and TSC2 loci is a suitable method to increase the yield of mutations identified in the TSC patient population. Electronic supplementary material The online version of this article (doi:10.1186/s12881-015-0155-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mark Nellist
- Department of Clinical Genetics, Ee-2426, Erasmus Medical Center, Wytemaweg 80, 3015, Rotterdam, CN, The Netherlands.
| | - Rutger W W Brouwer
- Center for Biomics, Erasmus Medical Center, Wytemaweg 80, 3015, Rotterdam, CN, The Netherlands.
| | - Christel E M Kockx
- Center for Biomics, Erasmus Medical Center, Wytemaweg 80, 3015, Rotterdam, CN, The Netherlands.
| | - Monique van Veghel-Plandsoen
- Department of Clinical Genetics, Ee-2426, Erasmus Medical Center, Wytemaweg 80, 3015, Rotterdam, CN, The Netherlands.
| | - Caroline Withagen-Hermans
- Department of Clinical Genetics, Ee-2426, Erasmus Medical Center, Wytemaweg 80, 3015, Rotterdam, CN, The Netherlands.
| | - Lida Prins-Bakker
- Department of Clinical Genetics, Ee-2426, Erasmus Medical Center, Wytemaweg 80, 3015, Rotterdam, CN, The Netherlands.
| | - Marianne Hoogeveen-Westerveld
- Department of Clinical Genetics, Ee-2426, Erasmus Medical Center, Wytemaweg 80, 3015, Rotterdam, CN, The Netherlands.
| | - Alan Mrsic
- Department of Clinical Genetics, Ee-2426, Erasmus Medical Center, Wytemaweg 80, 3015, Rotterdam, CN, The Netherlands.
| | - Mike M P van den Berg
- Department of Clinical Genetics, Ee-2426, Erasmus Medical Center, Wytemaweg 80, 3015, Rotterdam, CN, The Netherlands. .,Department of Ophthalmology, Erasmus Medical Center, Wytemaweg 80, 3015, Rotterdam, CN, The Netherlands.
| | - Anna E Koopmans
- Department of Clinical Genetics, Ee-2426, Erasmus Medical Center, Wytemaweg 80, 3015, Rotterdam, CN, The Netherlands. .,Department of Ophthalmology, Erasmus Medical Center, Wytemaweg 80, 3015, Rotterdam, CN, The Netherlands.
| | - Marie-Claire de Wit
- Department of Neurology, Sophia Children's Hospital, Erasmus Medical Center, Wytemaweg 80, 3015, Rotterdam, CN, The Netherlands.
| | - Floor E Jansen
- Department of Pediatric Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3508, Utrecht, EA, The Netherlands.
| | - Anneke J A Maat-Kievit
- Department of Clinical Genetics, Ee-2426, Erasmus Medical Center, Wytemaweg 80, 3015, Rotterdam, CN, The Netherlands.
| | - Ans van den Ouweland
- Department of Clinical Genetics, Ee-2426, Erasmus Medical Center, Wytemaweg 80, 3015, Rotterdam, CN, The Netherlands.
| | - Dicky Halley
- Department of Clinical Genetics, Ee-2426, Erasmus Medical Center, Wytemaweg 80, 3015, Rotterdam, CN, The Netherlands.
| | - Annelies de Klein
- Department of Clinical Genetics, Ee-2426, Erasmus Medical Center, Wytemaweg 80, 3015, Rotterdam, CN, The Netherlands.
| | - Wilfred F J van IJcken
- Center for Biomics, Erasmus Medical Center, Wytemaweg 80, 3015, Rotterdam, CN, The Netherlands.
| |
Collapse
|
52
|
Jariwala N, Rajasekaran D, Srivastava J, Gredler R, Akiel MA, Robertson CL, Emdad L, Fisher PB, Sarkar D. Role of the staphylococcal nuclease and tudor domain containing 1 in oncogenesis (review). Int J Oncol 2014; 46:465-73. [PMID: 25405367 PMCID: PMC4277250 DOI: 10.3892/ijo.2014.2766] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 10/03/2014] [Indexed: 12/26/2022] Open
Abstract
The staphylococcal nuclease and tudor domain containing 1 (SND1) is a multifunctional protein overexpressed in breast, prostate, colorectal and hepatocellular carcinomas and malignant glioma. Molecular studies have revealed the multifaceted activities of SND1 involved in regulating gene expression at transcriptional as well as post-transcriptional levels. Early studies identified SND1 as a transcriptional co-activator. SND1 is also a component of RNA-induced silencing complex (RISC) thus mediating RNAi function, a regulator of mRNA splicing, editing and stability, and plays a role in maintenance of cell viability. Such diverse actions allow the SND1 to modulate a complex array of molecular networks, thereby promoting carcinogenesis. Here, we describe the crucial role of SND1 in cancer development and progression, and highlight SND1 as a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Nidhi Jariwala
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Devaraja Rajasekaran
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Jyoti Srivastava
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Rachel Gredler
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Maaged A Akiel
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Chadia L Robertson
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Luni Emdad
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Devanand Sarkar
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
53
|
Hu DG, Meech R, McKinnon RA, Mackenzie PI. Transcriptional regulation of human UDP-glucuronosyltransferase genes. Drug Metab Rev 2014; 46:421-58. [PMID: 25336387 DOI: 10.3109/03602532.2014.973037] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Glucuronidation is an important metabolic pathway for many small endogenous and exogenous lipophilic compounds, including bilirubin, steroid hormones, bile acids, carcinogens and therapeutic drugs. Glucuronidation is primarily catalyzed by the UDP-glucuronosyltransferase (UGT) 1A and two subfamilies, including nine functional UGT1A enzymes (1A1, 1A3-1A10) and 10 functional UGT2 enzymes (2A1, 2A2, 2A3, 2B4, 2B7, 2B10, 2B11, 2B15, 2B17 and 2B28). Most UGTs are expressed in the liver and this expression relates to the major role of hepatic glucuronidation in systemic clearance of toxic lipophilic compounds. Hepatic glucuronidation activity protects the body from chemical insults and governs the therapeutic efficacy of drugs that are inactivated by UGTs. UGT mRNAs have also been detected in over 20 extrahepatic tissues with a unique complement of UGT mRNAs seen in almost every tissue. This extrahepatic glucuronidation activity helps to maintain homeostasis and hence regulates biological activity of endogenous molecules that are primarily inactivated by UGTs. Deciphering the molecular mechanisms underlying tissue-specific UGT expression has been the subject of a large number of studies over the last two decades. These studies have shown that the constitutive and inducible expression of UGTs is primarily regulated by tissue-specific and ligand-activated transcription factors (TFs) via their binding to cis-regulatory elements (CREs) in UGT promoters and enhancers. This review first briefly summarizes published UGT gene transcriptional studies and the experimental models and tools utilized in these studies, and then describes in detail the TFs and their respective CREs that have been identified in the promoters and/or enhancers of individual UGT genes.
Collapse
Affiliation(s)
- Dong Gui Hu
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University School of Medicine, Flinders Medical Centre , Bedford Park, SA , Australia
| | | | | | | |
Collapse
|
54
|
Situational awareness: regulation of the myb transcription factor in differentiation, the cell cycle and oncogenesis. Cancers (Basel) 2014; 6:2049-71. [PMID: 25279451 PMCID: PMC4276956 DOI: 10.3390/cancers6042049] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Revised: 08/11/2014] [Accepted: 09/26/2014] [Indexed: 12/02/2022] Open
Abstract
This review summarizes the mechanisms that control the activity of the c-Myb transcription factor in normal cells and tumors, and discusses how c-Myb plays a role in the regulation of the cell cycle. Oncogenic versions of c-Myb contribute to the development of leukemias and solid tumors such as adenoid cystic carcinoma, breast cancer and colon cancer. The activity and specificity of the c-Myb protein seems to be controlled through changes in protein-protein interactions, so understanding how it is regulated could lead to the development of novel therapeutic strategies.
Collapse
|
55
|
Björner S, Fitzpatrick PA, Li Y, Allred C, Howell A, Ringberg A, Olsson H, Miller CJ, Axelson H, Landberg G. Epithelial and stromal microRNA signatures of columnar cell hyperplasia linking Let-7c to precancerous and cancerous breast cancer cell proliferation. PLoS One 2014; 9:e105099. [PMID: 25122196 PMCID: PMC4133372 DOI: 10.1371/journal.pone.0105099] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 07/21/2014] [Indexed: 01/22/2023] Open
Abstract
Columnar cell hyperplasia (CCH) is the earliest histologically identifiable breast lesion linked to cancer progression and is characterized by increased proliferation, decreased apoptosis and elevated oestrogen receptor α (ERα) expression. The mechanisms underlying the initiation of these lesions have not been clarified but might involve early and fundamental changes in cancer progression. MiRNAs are key regulators of several biological processes, acting by influencing the post-transcriptional regulation of numerous targets, thus making miRNAs potential candidates in cancer initiation. Here we have defined novel epithelial as well as stromal miRNA signatures from columnar cell hyperplasia lesions compared to normal terminal duct lobular units by using microdissection and miRNA microarrays. Let-7c were among the identified downregulated epithelial miRNAs and its functions were delineated in unique CCH derived cells and breast cancer cell line MCF-7 suggesting anti-proliferative traits potentially due to effects on Myb and ERα. MiR-132 was upregulated in the stroma surrounding CCH compared to stoma surrounding normal terminal duct lobular units (TDLUs), and overexpression of miR-132 in immortalized fibroblasts and in fibroblasts co-cultured with epithelial CCH cells caused substantial expression changes of genes involved in metabolism, DNA damage and cell motility. The miRNA signatures identified in CCH indicate early changes in the epithelial and stromal compartment of CCH and could represent early key alterations in breast cancer progression that potentially could be targeted in novel prevention or treatment schedules.
Collapse
Affiliation(s)
- Sofie Björner
- Center for Molecular Pathology, Skåne University Hospital, Department of Laboratory Medicine Malmö, Lund University, Malmö, Sweden
- Breakthrough Breast Cancer Research Unit, Institute of Cancer Sciences, University of Manchester, Manchester Academic Health Science Centre, Paterson Institute for Cancer Research, The Christie National Health Service Foundation Trust, Manchester, United Kingdom
- Sahlgrenska Cancer Center, Department of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Paul A. Fitzpatrick
- Sahlgrenska Cancer Center, Department of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Yaoyong Li
- Cancer Research UK Applied Computational Biology and Bioinformatics Group, Paterson Institute for Cancer Research, Manchester, United Kingdom
| | - Craig Allred
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Anthony Howell
- Breakthrough Breast Cancer Research Unit, Institute of Cancer Sciences, University of Manchester, Manchester Academic Health Science Centre, Paterson Institute for Cancer Research, The Christie National Health Service Foundation Trust, Manchester, United Kingdom
| | - Anita Ringberg
- Department of Plastic and Reconstructive Surgery, SUS Malmö, Institute of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Håkan Olsson
- Department of Oncology, Skåne University Hospital, Institute of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Crispin J. Miller
- Cancer Research UK Applied Computational Biology and Bioinformatics Group, Paterson Institute for Cancer Research, Manchester, United Kingdom
| | - Håkan Axelson
- Translational Cancer Research, Medicon Village, Department of Laboratory Medicine Malmö, Lund University, Lund, Sweden
| | - Göran Landberg
- Center for Molecular Pathology, Skåne University Hospital, Department of Laboratory Medicine Malmö, Lund University, Malmö, Sweden
- Breakthrough Breast Cancer Research Unit, Institute of Cancer Sciences, University of Manchester, Manchester Academic Health Science Centre, Paterson Institute for Cancer Research, The Christie National Health Service Foundation Trust, Manchester, United Kingdom
- Sahlgrenska Cancer Center, Department of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
56
|
Stenman G, Persson F, Andersson MK. Diagnostic and therapeutic implications of new molecular biomarkers in salivary gland cancers. Oral Oncol 2014; 50:683-90. [DOI: 10.1016/j.oraloncology.2014.04.008] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 04/22/2014] [Accepted: 04/26/2014] [Indexed: 12/19/2022]
|
57
|
Hann SR. MYC cofactors: molecular switches controlling diverse biological outcomes. Cold Spring Harb Perspect Med 2014; 4:a014399. [PMID: 24939054 DOI: 10.1101/cshperspect.a014399] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The transcription factor MYC has fundamental roles in proliferation, apoptosis, tumorigenesis, and stem cell pluripotency. Over the last 30 years extensive information has been gathered on the numerous cofactors that interact with MYC and the target genes that are regulated by MYC as a means of understanding the molecular mechanisms controlling its diverse roles. Despite significant advances and perhaps because the amount of information learned about MYC is overwhelming, there has been little consensus on the molecular functions of MYC that mediate its critical biological roles. In this perspective, the major MYC cofactors that regulate the various transcriptional activities of MYC, including canonical and noncanonical transactivation and transcriptional repression, will be reviewed and a model of how these transcriptional mechanisms control MYC-mediated proliferation, apoptosis, and tumorigenesis will be presented. The basis of the model is that a variety of cofactors form dynamic MYC transcriptional complexes that can switch the molecular and biological functions of MYC to yield a diverse range of outcomes in a cell-type- and context-dependent fashion.
Collapse
Affiliation(s)
- Stephen R Hann
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-2175
| |
Collapse
|
58
|
Estrogen induces Vav1 expression in human breast cancer cells. PLoS One 2014; 9:e99052. [PMID: 24905577 PMCID: PMC4048212 DOI: 10.1371/journal.pone.0099052] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2013] [Accepted: 05/09/2014] [Indexed: 12/21/2022] Open
Abstract
Vav1, a guanine nucleotide exchange factor (GEF) for Rho family GTPases, is a hematopoietic protein involved in a variety of cellular events. In recent years, aberrant expression of Vav1 has been reported in non-hematopoietic cancers including human breast cancer. It remains to be answered how Vav1 is expressed and what Vav1 does in its non-resident tissues. In this study, we aimed to explore the mechanism for Vav1 expression in breast cancer cells in correlation with estrogen-ER pathway. We not only verified the ectopic expression of Vav1 in human breast cancer cell lines, but also observed that Vav1 expression was induced by 17β-estradiol (E2), a typical estrogen receptor (ER) ligand, in ER-positive cell lines. On the other hand, Tamoxifen, a selective estrogen receptor modulator (SERM), and ICI 182,780, an ER antagonist, suppressed the expression of Vav1. The estrogen receptor modulating Vav1 expression was identified to be α form, not β. Furthermore, treatment of E2 increased the transcription of vav1 gene by enhancing the promoter activity, though there was no recognizable estrogen response element (ERE). Nevertheless, two regions at the vav1 gene promoter were defined to be responsible for E2-induced activation of vav1 promoter. Chromatin immunoprecipitation (ChIP) and co-immunoprecipitation (Co-IP) analyses suggested that ERα might access to the vav1 promoter via interacting with transcription factors, c-Myb and ELF-1. Consequently, the enhanced expression of Vav1 led to the elevation of Cyclin D1 and the progression of cell cycle. The present study implies that estrogen-ER modulates the transcription and expression of Vav1, which may contribute to the proliferation of cancerous cells.
Collapse
|
59
|
A supervised network analysis on gene expression profiles of breast tumors predicts a 41-gene prognostic signature of the transcription factor MYB across molecular subtypes. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2014; 2014:813067. [PMID: 24639887 PMCID: PMC3930188 DOI: 10.1155/2014/813067] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 10/07/2013] [Accepted: 10/20/2013] [Indexed: 02/05/2023]
Abstract
Background. MYB is predicted to be a favorable prognostic predictor in a breast cancer population. We proposed to find the inferred mechanism(s) relevant to the prognostic features of MYB via a supervised network analysis. Methods. Both coefficient of intrinsic dependence (CID) and Galton Pierson's correlation coefficient (GPCC) were combined and designated as CIDUGPCC. It is for the univariate network analysis. Multivariate CID is for the multivariate network analysis. Other analyses using bioinformatic tools and statistical methods are included. Results. ARNT2 is predicted to be the essential gene partner of MYB. We classified four prognostic relevant gene subpools in three breast cancer cohorts as feature types I–IV. Only the probes in feature type II are the potential prognostic feature of MYB. Moreover, we further validated 41 prognosis relevant probes to be the favorable prognostic signature. Surprisingly, two additional family members of MYB are elevated to promote poor prognosis when both levels of MYB and ARNT2 decline. Both MYBL1 and MYBL2 may partially decrease the tumor suppressive activities that are predicted to be up-regulated by MYB and ARNT2. Conclusions. The major prognostic feature of MYB is predicted to be determined by the MYB subnetwork (41 probes) that is relevant across subtypes.
Collapse
|
60
|
Kasper LH, Fukuyama T, Lerach S, Chang Y, Xu W, Wu S, Boyd KL, Brindle PK. Genetic interaction between mutations in c-Myb and the KIX domains of CBP and p300 affects multiple blood cell lineages and influences both gene activation and repression. PLoS One 2013; 8:e82684. [PMID: 24340053 PMCID: PMC3858336 DOI: 10.1371/journal.pone.0082684] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 10/28/2013] [Indexed: 02/06/2023] Open
Abstract
Adult blood cell production or definitive hematopoiesis requires the transcription factor c-Myb. The closely related KAT3 histone acetyltransferases CBP (CREBBP) and p300 (EP300) bind c-Myb through their KIX domains and mice homozygous for a p300 KIX domain mutation exhibit multiple blood defects. Perplexingly, mice homozygous for the same KIX domain mutation in CBP have normal blood. Here we test the hypothesis that the CBP KIX domain contributes subordinately to hematopoiesis via a genetic interaction with c-Myb. We assessed hematopoiesis in mice bearing compound mutations of c-Myb and/or the KIX domains of CBP and p300, and measured the effect of KIX domain mutations on c-Myb-dependent gene expression. We found that in the context of a p300 KIX mutation, the CBP KIX domain mutation affects platelets, B cells, T cells, and red cells. Gene interaction (epistasis) analysis provides mechanistic evidence that blood defects in KIX mutant mice are consistent with reduced c-Myb and KIX interaction. Lastly, we demonstrated that the CBP and p300 KIX domains contribute to both c-Myb-dependent gene activation and repression. Together these results suggest that the KIX domains of CBP, and especially p300, are principal mediators of c-Myb-dependent gene activation and repression that is required for definitive hematopoiesis.
Collapse
Affiliation(s)
- Lawryn H. Kasper
- Department of Biochemistry, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Tomofusa Fukuyama
- Department of Biochemistry, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Stephanie Lerach
- Department of Biochemistry, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Yunchao Chang
- Department of Biochemistry, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Wu Xu
- Department of Biochemistry, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Song Wu
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Kelli L. Boyd
- Veterinary Pathology Core, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Paul K. Brindle
- Department of Biochemistry, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| |
Collapse
|
61
|
Hugo HJ, Pereira L, Suryadinata R, Drabsch Y, Gonda TJ, Gunasinghe NPAD, Pinto C, Soo ETL, van Denderen BJW, Hill P, Ramsay RG, Sarcevic B, Newgreen DF, Thompson EW. Direct repression of MYB by ZEB1 suppresses proliferation and epithelial gene expression during epithelial-to-mesenchymal transition of breast cancer cells. Breast Cancer Res 2013; 15:R113. [PMID: 24283570 PMCID: PMC3979034 DOI: 10.1186/bcr3580] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 10/31/2013] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION Epithelial-to-mesenchymal transition (EMT) promotes cell migration and is important in metastasis. Cellular proliferation is often downregulated during EMT, and the reverse transition (MET) in metastases appears to be required for restoration of proliferation in secondary tumors. We studied the interplay between EMT and proliferation control by MYB in breast cancer cells. METHODS MYB, ZEB1, and CDH1 expression levels were manipulated by lentiviral small-hairpin RNA (shRNA)-mediated knockdown/overexpression, and verified with Western blotting, immunocytochemistry, and qRT-PCR. Proliferation was assessed with bromodeoxyuridine pulse labeling and flow cytometry, and sulforhodamine B assays. EMT was induced with epidermal growth factor for 9 days or by exposure to hypoxia (1% oxygen) for up to 5 days, and assessed with qRT-PCR, cell morphology, and colony morphology. Protein expression in human breast cancers was assessed with immunohistochemistry. ZEB1-MYB promoter binding and repression were determined with Chromatin Immunoprecipitation Assay and a luciferase reporter assay, respectively. Student paired t tests, Mann-Whitney, and repeated measures two-way ANOVA tests determined statistical significance (P < 0.05). RESULTS Parental PMC42-ET cells displayed higher expression of ZEB1 and lower expression of MYB than did the PMC42-LA epithelial variant. Knockdown of ZEB1 in PMC42-ET and MDA-MB-231 cells caused increased expression of MYB and a transition to a more epithelial phenotype, which in PMC42-ET cells was coupled with increased proliferation. Indeed, we observed an inverse relation between MYB and ZEB1 expression in two in vitro EMT cell models, in matched human breast tumors and lymph node metastases, and in human breast cancer cell lines. Knockdown of MYB in PMC42-LA cells (MYBsh-LA) led to morphologic changes and protein expression consistent with an EMT. ZEB1 expression was raised in MYBsh-LA cells and significantly repressed in MYB-overexpressing MDA-MB-231 cells, which also showed reduced random migration and a shift from mesenchymal to epithelial colony morphology in two dimensional monolayer cultures. Finally, we detected binding of ZEB1 to MYB promoter in PMC42-ET cells, and ZEB1 overexpression repressed MYB promoter activity. CONCLUSIONS This work identifies ZEB1 as a transcriptional repressor of MYB and suggests a reciprocal MYB-ZEB1 repressive relation, providing a mechanism through which proliferation and the epithelial phenotype may be coordinately modulated in breast cancer cells.
Collapse
|
62
|
Edavana VK, Penney RB, Yao-Borengasser A, Williams S, Rogers L, Dhakal IB, Kadlubar S. Fulvestrant up regulates UGT1A4 and MRPs through ERα and c-Myb pathways: a possible primary drug disposition mechanism. SPRINGERPLUS 2013; 2:620. [PMID: 24298433 PMCID: PMC3841332 DOI: 10.1186/2193-1801-2-620] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 10/30/2013] [Indexed: 11/17/2022]
Abstract
Fulvestrant (Faslodex™) is a pure antiestrogen that is effective in treating estrogen receptor-(ER) positive breast cancer tumors that are resistant to selective estrogen receptor modulators such as tamoxifen. Clinical trials investigating the utility of adding fulvestrant to other therapeutics have not been shown to affect cytochrome P450-mediated metabolism. Effects on phase II metabolism and drug resistance have not been explored. This study demonstrates that fulvestrant up regulates the expression of UDP glucuronosyltransferase 1A4 (UGT1A4) >2.5- and >3.5-fold in MCF7 and HepG2 cells, respectively. Up regulation occurred in a time- and concentration-dependent manner, and was inhibited by siRNA silencing of ERα. Fulvestrant also up regulates multidrug resistance-associated proteins (MRPs). There was an up regulation of MRP2 (1.5- and 3.5-fold), and MRP3 (5.5- and 4.5-fold) in MCF7 and HepG2 cell lines, respectively, and an up regulation of MRP1 (4-fold) in MCF7 cells. UGT1A4 mRNA up regulation was significantly correlated with UGT1A4 protein expression, anastrozole glucuronidation, ERα mRNA expression and MRP mRNA expression, but not with ERα protein expression. Genetic variants in the UGT1A4 promoter (-163A, -217G and -219T) reduced the basal activity of UGT1A4 by 40-60%. In silico analysis indicated that transcription factor c-Myb binding capacity may be affected by these variations. Luciferase activity assays demonstrate that silencing c-Myb abolished UGT1A4 up regulation by fulvestrant in promoters with the common genotype (-163G, -217 T and -219C) in MCF7 cells. These data indicate that fulvestrant can influence the disposition of other UGT1A4 substrates. These findings suggest a clinically significant role for UGT1A4 and MRPs in drug efficacy.
Collapse
Affiliation(s)
- Vineetha K Edavana
- Division of Medical Genetics, College of Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham, #580, Little Rock, AR 72205 USA
| | | | | | | | | | | | | |
Collapse
|
63
|
Proteome changes induced by c-myb silencing in human chronic myeloid leukemia cells suggest molecular mechanisms and putative biomarkers of hematopoietic malignancies. J Proteomics 2013; 96:200-22. [PMID: 24220303 DOI: 10.1016/j.jprot.2013.10.040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 10/18/2013] [Accepted: 10/30/2013] [Indexed: 11/23/2022]
Abstract
UNLABELLED To shed light on the molecular mechanisms associated with aberrant accumulation of c-Myb in chronic myeloid leukemia, comparative proteomic analysis was performed on c-myb RNAi-specifically silenced K562 cells, sampled on a time-course basis. 2D-DIGE technology highlighted 37 differentially-represented proteins that were further characterized by nLC-ESI-LIT-MS/MS and validated by western blotting and qRT-PCR analysis. Most of the deregulated proteins were related to protein folding, energy/primary metabolism, transcription/translation regulation and oxidative stress response. Protein network analysis suggested that glycolysis, gluconeogenesis and protein ubiquitination biosynthesis pathways were highly represented, confirming also the pivotal role of c-Myc. A specific reduced representation was observed for glyceraldehyde-3-phosphate-dehydrogenase and α-enolase, suggesting a possible role of c-Myb in the activation of aerobic glycolysis. A reduced amount was also observed for stress responsive heat shock 70kDa protein and 78kDa glucose-regulated protein, previously identified as direct targets of c-Myb. Among over-represented proteins, worth mentioning is the chromatin modifier chromobox protein homolog 3 that contributes to silencing of E2F- and Myc-responsive genes in quiescent G0 cells. Data here presented, while providing novel insights onto the molecular mechanisms underlying c-Myb activity, indicate potential protein biomarkers for monitoring the progression of chronic myeloid leukemia. BIOLOGICAL SIGNIFICANCE Myeloid leukemia is a malignant disease of the hematopoietic system in which cells of myeloid lineages accumulate to an undifferentiated state. In particular, it was shown that an aberrant accumulation of the c-Myb transcriptional factor is associated with the suppression of normal differentiation processes promoting the development of the hematopoietic malignancies. Many efforts have been recently made to identify novel genes directly targeted by c-Myb at a transcriptome level. In this work, we originally describe a differential proteomic approach to facilitate the comprehension of the regulation of the protein networks exerted by c-Myb. Our study reveals a complex network of proteins regulated by c-Myb. The functional heterogeneity of these proteins emphasizes the pleiotropic role of c-Myb as a regulator of genes that are crucial for energy production and stress response in leukemia. In fact, variations in glyceraldehyde-3-phosphate-dehydrogenase and α-enolase suggest a possible role of c-Myb in the activation of aerobic glycolysis. Moreover, significant differences were found for heat shock 70kDa protein and 78kDa glucose-regulated protein known as direct c-Myb targets. This work highlights potential protein biomarkers to look into disease progression and to develop translational medicine approaches in myeloid leukemia.
Collapse
|
64
|
Identification and characterization of a nanog homolog in Japanese flounder (Paralichthys olivaceus). Gene 2013; 531:411-21. [PMID: 24013085 DOI: 10.1016/j.gene.2013.08.030] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 07/28/2013] [Accepted: 08/10/2013] [Indexed: 02/06/2023]
Abstract
The homeodomain-containing transcription factor nanog plays a key role in maintaining the pluripotency and self-renewal of embryonic stem cells in mammals. Stem cells offered as a significant and effective tool for generation of transgenic animals and preservation of genetic resources. The molecular genetic organization and expression of nanog gene in marine fish have not been reported yet. In this study, we isolated and characterized the flounder nanog gene as a first step towards understanding the mechanism of the plurpotency of fish stem cells and develop a potential molecular marker to identify the stem cells in vivo and in vitro. Phylogenetic, gene structure and chromosome synteny analysis provided the evidence that Po-nanog is homologous to the mammalian nanog gene. Protein sequence comparison showed that flounder Nanog shared low similarity with other vertebrate orthologs except for a conserved homeodomain. Quantitative RT-PCR analysis showed that flounder nanog was maternally expressed, and the transcripts were present from the one-cell stage to the neurula stage with the peaking at blastula stage. Whole mount in situ hybridization analyses demonstrated that the transcripts were present in all blastomeres of the early embryo. Tissue distribution analysis indicated that nanog was detectable only in gonads. Further, the expression was significantly high in ovary than in testis. In situ hybridization revealed that the transcripts were located in the cytoplasm of the oogonia and oocytes in ovary, only in the spermatogonia but no spermatocytes or spermatids in testis. The promoter region was also analyzed to have several basal core promoter elements and transcription factor binding sites. All these results suggest that Po-Nanog may have a conservative function between teleosts and mammals.
Collapse
|
65
|
Bobowski M, Vincent A, Steenackers A, Colomb F, Van Seuningen I, Julien S, Delannoy P. Estradiol represses the G(D3) synthase gene ST8SIA1 expression in human breast cancer cells by preventing NFκB binding to ST8SIA1 promoter. PLoS One 2013; 8:e62559. [PMID: 23626833 PMCID: PMC3633854 DOI: 10.1371/journal.pone.0062559] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 03/23/2013] [Indexed: 12/13/2022] Open
Abstract
Recent data have underlined a possible role of G(D3) synthase (GD3S) and complex gangliosides in Estrogen Receptor (ER) negative breast cancer progression. Here, we describe the main transcript of the GD3S coding gene ST8SIA1 expressed in breast tumors. We characterized the corresponding core promoter in Hs578T breast cancer cells and showed that estradiol decreases ST8SIA1 mRNA expression in ER-positive MCF-7 cells and ERα-transfected ER-negative Hs578T cells. The activity of the core promoter sequence of ST8SIA1 is also repressed by estradiol. The core promoter of ST8SIA1 contains two putative Estrogen Response Elements (ERE) that were not found to be involved in the promoter activity pathway. However, NFκB was shown to be involved in ST8SIA1 transcriptional activation and we demonstrated that estradiol prevents NFκB to bind to ST8SIA1 core promoter in ERα expressing breast cancer cells by inhibiting p65 and p50 nucleus localization. The activation of NFκB pathway in ER-negative tumors, due to the absence of estradiol signaling, might explain the overexpression of G(D3) synthase in this tumor subtype.
Collapse
Affiliation(s)
- Marie Bobowski
- University Lille Nord de France, Lille, France
- Université des Sciences et Technologies de Lille (USTL), Unité de Glycobiologie Structurale et Fonctionnelle (UGSF), Villeneuve d’Ascq, France
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 8576, Villeneuve d’Ascq, France
| | - Audrey Vincent
- University Lille Nord de France, Lille, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), Jean Pierre Aubert Research Center, Lille, France
- Centre Hospitalier Régional et Universitaire de Lille, Lille, France
| | - Agata Steenackers
- University Lille Nord de France, Lille, France
- Université des Sciences et Technologies de Lille (USTL), Unité de Glycobiologie Structurale et Fonctionnelle (UGSF), Villeneuve d’Ascq, France
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 8576, Villeneuve d’Ascq, France
| | - Florent Colomb
- University Lille Nord de France, Lille, France
- Université des Sciences et Technologies de Lille (USTL), Unité de Glycobiologie Structurale et Fonctionnelle (UGSF), Villeneuve d’Ascq, France
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 8576, Villeneuve d’Ascq, France
| | - Isabelle Van Seuningen
- University Lille Nord de France, Lille, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), Jean Pierre Aubert Research Center, Lille, France
- Centre Hospitalier Régional et Universitaire de Lille, Lille, France
| | - Sylvain Julien
- University Lille Nord de France, Lille, France
- Université des Sciences et Technologies de Lille (USTL), Unité de Glycobiologie Structurale et Fonctionnelle (UGSF), Villeneuve d’Ascq, France
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 8576, Villeneuve d’Ascq, France
| | - Philippe Delannoy
- University Lille Nord de France, Lille, France
- Université des Sciences et Technologies de Lille (USTL), Unité de Glycobiologie Structurale et Fonctionnelle (UGSF), Villeneuve d’Ascq, France
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 8576, Villeneuve d’Ascq, France
- * E-mail:
| |
Collapse
|
66
|
Armengol S, Arretxe E, Rodríguez L, Ochoa B, Chico Y, Martínez MJ. NF-κB, Sp1 and NF-Y as transcriptional regulators of human SND1 gene. Biochimie 2012; 95:735-42. [PMID: 23160072 DOI: 10.1016/j.biochi.2012.10.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 10/30/2012] [Indexed: 11/19/2022]
Abstract
Staphylococcal nuclease domain-containing protein 1 (SND1), also called Tudor-SN, is required for many biological events ranging from gene expression to cell growth regulation. Promoter regulation of SND1 gene and its molecular mechanism have remained elusive to date. In this work, we have identified SND1 as a new target gene for NF-κB, Sp1 and NF-Y transcription factors. We isolated and characterized a 3808 bp sequence corresponding to the human SND1 gene promoter (GenBank ID: EF690304). It lacks the typical TATA-box element and contains a CpG island with several Sp1 binding sites at the 3' end, and a highly conserved 300 bp segment with two inverted CCAAT boxes that bind NF-Y, in addition to NF-κB sites and other cis-regulatory elements. Electrophoretic mobility shift assays and chromatin immunoprecipitation experiments confirmed the ability of SND1 promoter to bind NF-κB, Sp1 and NF-Y in vitro and in vivo. Deletion analysis of the 5'-flanking region by luciferase reporter assays, showed the minimum promoter activity 112 base-pair upstream from the transcription start site, and an enhancer region between -112 and -274 bp responsible for the maximal transcriptional activity of the promoter. Site-directed mutagenesis of the CCAAT and GC boxes and the NF-κB elements within the proximal region substantially reduced SND1 promoter activity. Proinflammatory cytokine TNF-α caused an increase of SND1 promoter activity that is mediated, at least in part, via NF-κB as mutation in the NF-κB sites impaired the promoter stimulation. We provide for the first time the characterization of the human SND1 promoter activity and establish a transcriptional network associated to the key transcription factors NF-κB, Sp1 and NF-Y that operates in the control of the SND1 gene expression.
Collapse
Affiliation(s)
- Sandra Armengol
- Department of Physiology, Faculty of Medicine and Dentistry, University of the Basque Country UPV/EHU, Barrio Sarriena s/n, 48940 Leioa, Vizcaya, Spain
| | | | | | | | | | | |
Collapse
|
67
|
Lorenzo PI, Brendeford EM, Gilfillan S, Gavrilov AA, Leedsak M, Razin SV, Eskeland R, Sæther T, Gabrielsen OS. Identification of c-Myb Target Genes in K562 Cells Reveals a Role for c-Myb as a Master Regulator. Genes Cancer 2012; 2:805-17. [PMID: 22393465 DOI: 10.1177/1947601911428224] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Accepted: 10/08/2011] [Indexed: 11/17/2022] Open
Abstract
The c-Myb transcription factor is an important regulator of hematopoietic cell development. c-Myb is expressed in immature hematopoietic cells and plays a direct role in lineage fate selection, cell cycle progression, and differentiation of myeloid as well as B- and T-lymphoid progenitor cells. As a DNA-binding transcription factor, c-Myb regulates specific gene programs through activation of target genes. Still, our understanding of these programs is incomplete. Here, we report a set of novel c-Myb target genes, identified using a combined approach: specific c-Myb knockdown by 2 different siRNAs and subsequent global expression profiling, combined with the confirmation of direct binding of c-Myb to the target promoters by ChIP assays. The combination of these 2 approaches, as well as additional validation such as cloning and testing the promoters in reporter assays, confirmed that MYADM, LMO2, GATA2, STAT5A, and IKZF1 are target genes of c-Myb. Additional studies, using chromosome conformation capture, demonstrated that c-Myb target genes may directly interact with each other, indicating that these genes may be coordinately regulated. Of the 5 novel target genes identified, 3 are transcription factors, and one is a transcriptional co-regulator, supporting a role of c-Myb as a master regulator controlling the expression of other transcriptional regulators in the hematopoietic system.
Collapse
|
68
|
Siu A, Virtanen C, Jongstra J. PIM kinase isoform specific regulation of MIG6 expression and EGFR signaling in prostate cancer cells. Oncotarget 2012; 2:1134-44. [PMID: 22193779 PMCID: PMC3282072 DOI: 10.18632/oncotarget.386] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The PIM family of oncogenic serine/threonine kinases regulates tumour cell proliferation. To identify proliferative signaling pathways that are regulated by PIM kinases we analyzed gene expression differences in DU-145 and PC3 prostate cancer derived cells induced by treatment with the recently developed highly selective PIM kinase inhibitor M-110. This identified 97 genes the expression of which is affected by M-110 in both cell lines. We then focused on the M-110 induced up regulation of the MIG6 gene that encodes a negative regulator of EGFR signaling. Here we show that M-110 and the structurally unrelated PIM kinase inhibitor SGI-1776 up regulate MIG6 in DU-145 and PC3 cells. Knockdown of PIM-1 but not of PIM-2 or PIM-3 also up regulates MIG6 expression, which identifies MIG6 as a PIM-1 regulated gene. In agreement with the role of MIG6 protein as a negative regulator of EGFR signaling we found that M-110 treatment inhibits EGF induced EGFR activation and the activation of the downstream ERK MAPkinase pathway. The biological significance of these findings are demonstrated by the fact that co-treatment of DU-145 or PC3 cells with the EGFR tyrosine kinase inhibitor Gefitinib and M-110 or SGI-1776 has synergistic inhibitory effects on cell proliferation. These experiments define a novel biological function of PIM-1 as a co-regulator of EGFR signaling and suggest that PIM inhibitors may be used in combination therapies to increase the efficacy of EGFR tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Allan Siu
- Department of Immunology, University of Toronto, Toronto, Canada
| | | | | |
Collapse
|
69
|
Knopfová L, Beneš P, Pekarčíková L, Hermanová M, Masařík M, Pernicová Z, Souček K, Smarda J. c-Myb regulates matrix metalloproteinases 1/9, and cathepsin D: implications for matrix-dependent breast cancer cell invasion and metastasis. Mol Cancer 2012; 11:15. [PMID: 22439866 PMCID: PMC3325857 DOI: 10.1186/1476-4598-11-15] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Accepted: 03/23/2012] [Indexed: 12/11/2022] Open
Abstract
Background The c-Myb transcription factor is essential for the maintenance of stem-progenitor cells in bone marrow, colon epithelia, and neurogenic niches. c-Myb malfunction contributes to several types of malignancies including breast cancer. However, the function of c-Myb in the metastatic spread of breast tumors remains unexplored. In this study, we report a novel role of c-Myb in the control of specific proteases that regulate the matrix-dependent invasion of breast cancer cells. Results Ectopically expressed c-Myb enhanced migration and ability of human MDA-MB-231 and mouse 4T1 mammary cancer cells to invade Matrigel but not the collagen I matrix in vitro. c-Myb strongly increased the expression/activity of cathepsin D and matrix metalloproteinase (MMP) 9 and significantly downregulated MMP1. The gene coding for cathepsin D was suggested as the c-Myb-responsive gene and downstream effector of the migration-promoting function of c-Myb. Finally, we demonstrated that c-Myb delayed the growth of mammary tumors in BALB/c mice and affected the metastatic potential of breast cancer cells in an organ-specific manner. Conclusions This study identified c-Myb as a matrix-dependent regulator of invasive behavior of breast cancer cells.
Collapse
Affiliation(s)
- Lucia Knopfová
- Department of Experimental Biology, Faculty of Science, Masaryk University, and International Clinical Research Center, CBCE, St. Anne's University Hospital, Brno, Czech Republic
| | | | | | | | | | | | | | | |
Collapse
|
70
|
Regulation of the human catalytic subunit of telomerase (hTERT). Gene 2012; 498:135-46. [PMID: 22381618 DOI: 10.1016/j.gene.2012.01.095] [Citation(s) in RCA: 205] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Revised: 01/29/2012] [Accepted: 01/30/2012] [Indexed: 12/12/2022]
Abstract
Over the past decade, there has been much interest in the regulation of telomerase, the enzyme responsible for maintaining the integrity of chromosomal ends, and its crucial role in cellular immortalization, tumorigenesis, and the progression of cancer. Telomerase activity is characterized by the expression of the telomerase reverse transcriptase (TERT) gene, suggesting that TERT serves as the major limiting agent for telomerase activity. Recent discoveries have led to characterization of various interactants that aid in the regulation of human TERT (hTERT), including numerous transcription factors; further supporting the pivotal role that transcription plays in both the expression and repression of telomerase. Several studies have suggested that epigenetic modulation of the hTERT core promoter region may provide an additional level of regulation. Although these studies have provided essential information on the regulation of hTERT, there has been ambiguity of the role of methylation within the core promoter region and the subsequent binding of various activating and repressive agents. As a result, we found it necessary to consolidate and summarize these recent developments and elucidate these discrepancies. In this review, we focus on the co-regulation of hTERT via transcriptional regulation, the presence or absence of various activators and repressors, as well as the epigenetic pathways of DNA methylation and histone modifications.
Collapse
|
71
|
Miao RY, Drabsch Y, Cross RS, Cheasley D, Carpinteri S, Pereira L, Malaterre J, Gonda TJ, Anderson RL, Ramsay RG. MYB is essential for mammary tumorigenesis. Cancer Res 2011; 71:7029-37. [PMID: 21948968 DOI: 10.1158/0008-5472.can-11-1015] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
MYB oncogene upregulation is associated with estrogen receptor (ER)-positive breast cancer, but disease requirements for MYB function in vivo have not been explored. In this study, we provide evidence of a critical requirement for MYB functions in models of human and murine breast cancer. In human breast cancer, we found that MYB expression was critical for tumor cell growth both in vitro and in vivo in xenograft settings. In transgenic knockout mice, tissue-specific deletion of the murine MYB gene caused a transient defect in mammary gland development that was reflected in delayed ductal branching and defective apical bud formation. In mouse mammary tumor virus (MMTV)-NEU mice where tumors are initiated by activation of HER2, MYB deletion was sufficient to abolish tumor formation. In the more aggressive MMTV-PyMT model system, MYB deletion delayed tumorigenesis significantly. Together, the findings in these transgenic knockout models implied that MYB was critical during an early window in mammary development when it was essential for tumor initiation, even though MYB loss did not exert a lasting impact upon normal mammary function. Two important MYB-target genes that promote cell survival, BCL2 and GRP78/BIP, were each elevated compared with nontransformed mammary epithelial cells, thereby promoting survival as confirmed in colony formation assays in vitro. Taken together, our findings establish a role for MYB at the hub of ER- and HER2-dependent pathways in mammary carcinogenesis.
Collapse
Affiliation(s)
- Rebecca Yu Miao
- Peter MacCallum Cancer Centre and Department of Pathology, University of Melbourne, Melbourne, Victoria, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|