1
|
Shen X, Zhou C, Feng H, Li J, Xia T, Cheng X, Zhao R, Zou D. ETV1 Positively Correlated With Immune Infiltration and Poor Clinical Prognosis in Colorectal Cancer. Front Immunol 2022; 13:939806. [PMID: 35860243 PMCID: PMC9291282 DOI: 10.3389/fimmu.2022.939806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/03/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectiveNumerous studies recently suggested that the immune microenvironment could influence the development of colorectal cancer (CRC). These findings implied that the infiltration of immune cells could be a promising prognostic biomarker for CRC.MethodsFurthermore, the Oncomine database and R2 platform analysis were applied in our research to validate CRC clinical prognosis via expression levels of polyoma enhancer activator 3 (PEA3) members. We explored the correlation of ETV1, ETV4, and ETV5 with tumor-infiltrating immune cells (TIICs) in CRC tumor microenvironments via the Tumor Immune Estimation Resource (TIMER) and Gene Expression Profiling Interactive Analysis (GEPIA). Immunohistochemistry (IHC) was used to validate our CRC clinical data.ResultsOur findings indicated that the upregulation of PEA3 members including ETV1 and ETV5 was positively associated with poor prognosis in CRC patients. Meanwhile, ETV1 and ETV5 may play significant roles in the development progress of CRC. Furthermore, ETV1 tends to be associated with immune infiltration of CRC, especially with cancer-associated fibroblasts and M2 macrophages.ConclusionThese findings revealed that ETV1 and ETV5 played significant roles in the development of CRC. Moreover, ETV1 was significantly associated with the infiltration of cancer-associated fibroblasts and M2 macrophages in CRC. Targeting ETV1 can be a potential auspicious approach for CRC treatment.
Collapse
Affiliation(s)
- Xiaonan Shen
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chunhua Zhou
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haoran Feng
- Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jialu Li
- State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Tianxue Xia
- Department of Gastroenterology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xi Cheng
- Department of General Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Xi Cheng, ; Ren Zhao, ; Duowu Zou,
| | - Ren Zhao
- Department of General Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Xi Cheng, ; Ren Zhao, ; Duowu Zou,
| | - Duowu Zou
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Xi Cheng, ; Ren Zhao, ; Duowu Zou,
| |
Collapse
|
2
|
Cavazzola LR, Carvalhal GF, Deves C, Renck D, Almeida R, Santos DIS. Relative mRNA expression of prostate-derived E-twenty-six factor and E-twenty-six variant 4 transcription factors, and of uridine phosphorylase-1 and thymidine phosphorylase enzymes, in benign and malignant prostatic tissue. Oncol Lett 2015; 9:2886-2894. [PMID: 26137165 DOI: 10.3892/ol.2015.3093] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 03/10/2015] [Indexed: 12/25/2022] Open
Abstract
Prostate cancer is the most frequent urological tumor, and the second most common cancer diagnosed in men. Incidence and mortality are variable and appear to depend on behavioral factors and genetic predisposition. The prostate-derived E-twenty-six factor (PDEF) and E-twenty-six variant 4 (ETV4) transcription factors, and the thymidine phosphorylase (TP) and uridine phosphorylase-1 (UP-1) enzymes, are reported to be components of the pathways leading to tumorigenesis and/or metastasis in a number of tumors. The present study aimed to analyze the mRNA expression levels of these proteins in prostatic cancerous and benign tissue, and their association with clinical and pathological variables. Using quantitative reverse transcription polymerase chain reaction, the mRNA expression levels of PDEF, ETV4, TP and UP-1 were studied in 52 tissue samples (31 of benign prostatic hyperplasia and 21 of prostate adenocarcinomas) obtained from patients treated by transurethral resection of the prostate or by radical prostatectomy. Relative expression was assessed using the ∆-CT method. Data was analyzed using Spearman's tests for correlation. P<0.05 was considered to indicate a statistically significant difference. The results revealed that PDEF, ETV4, UP-1 and TP were expressed in 85.7, 90.5, 95.2 and 100% of the prostate cancer samples, and in 90.3, 96.8, 90.3 and 96.8% of the benign samples, respectively. PDEF and ETV4 exhibited a significantly higher relative expression level in the tumor samples compared with their benign counterparts. The relative expression of TP and UP-1 did not differ significantly between benign and cancerous prostate tissues. The relative expression of TP was moderately and significantly correlated with the expression of ETV4 in the benign tissues. The relative expression of UP-1 was significantly lower in T3 compared with T1 and T2 cancers. These findings indicate that PDEF, ETV4, TP and UP-1 are typically expressed in benign and malignant prostatic tissues. Further studies are necessary to define the role of these proteins as therapeutic targets in prostate cancer.
Collapse
Affiliation(s)
- Luciane Rostirola Cavazzola
- Center for Research on Molecular and Functional Biology, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90619-900, Brazil
| | - Gustavo Franco Carvalhal
- Department of Urology, School of Medicine, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90619-900, Brazil
| | - Candida Deves
- Center for Research on Molecular and Functional Biology, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90619-900, Brazil
| | - Daiana Renck
- Center for Research on Molecular and Functional Biology, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90619-900, Brazil
| | - Ricardo Almeida
- Department of Urology, School of Medicine, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90619-900, Brazil
| | - DIóGENES Santiago Santos
- Center for Research on Molecular and Functional Biology, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90619-900, Brazil
| |
Collapse
|
3
|
Yuan ZY, Dai T, Wang SS, Peng RJ, Li XH, Qin T, Song LB, Wang X. Overexpression of ETV4 protein in triple-negative breast cancer is associated with a higher risk of distant metastasis. Onco Targets Ther 2014; 7:1733-42. [PMID: 25328406 PMCID: PMC4196788 DOI: 10.2147/ott.s66692] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Patients with triple-negative breast cancer (TNBC) present a higher probability of distant metastasis and lack of effective targeted therapy. ETS translocation variant 4 (ETV4) is an ETS (E-26) transcription factor and has been associated with tumor metastasis. However, the clinical and functional significance of ETV4 in TNBC still remains unclear. METHODS A human tumor metastasis polymerase chain reaction array was used to profile differential expression of tumor metastasis-related genes in TNBC tissue. Real-time reverse transcription and Western blot analyses were performed to verify ETV4 expression in TNBC cells and tissue. Immunohistochemistry was used to detect expression of ETV4 protein in 135 TNBC tissue samples for association between ETV4 protein expression and clinical outcomes. RESULTS A total total of eight upregulated (CCL7, KISS1, MET, MMP7, NR4A3, ETV4, TIMP3, and TSHR) and three downregulated (ITGA7, SSTR, and MMP2) genes were identified between TNBC tissue and the luminal subtype of breast cancer tissue. ETV4 messenger ribonucleic acid was more than five-fold upregulated in TNBC tissue compared with the control tissue. ETV4 overexpression was found in 57.0% of 135 TNBC cases. Overexpression of ETV4 protein was associated with an advanced stage and a higher proportion of positive lymph node and lymphovascular invasion. Patients with an ETV4-overexpressed tumor had a significantly higher risk of developing distant metastasis (P<0.0001) and shorter overall survival and disease-free survival. Overexpression of ETV4 protein was an independent predictor of short disease-free survival of TNBC patients (P=0.021). CONCLUSION Overexpression of ETV4 protein increases risk of developing distant metastasis and results in a poor prognosis for TNBC patients. Thus, ETV4 might be a novel target for developing an alternative therapeutic strategy for prevention of TNBC distant metastasis.
Collapse
Affiliation(s)
- Zhong-Yu Yuan
- State Key Laboratory of Oncology in South China, Guangzhou, People's Republic of China ; Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China ; Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Ting Dai
- State Key Laboratory of Oncology in South China, Guangzhou, People's Republic of China ; Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Shu-Sen Wang
- State Key Laboratory of Oncology in South China, Guangzhou, People's Republic of China ; Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China ; Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Rou-Jun Peng
- State Key Laboratory of Oncology in South China, Guangzhou, People's Republic of China ; Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China ; Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Xing-Hua Li
- State Key Laboratory of Oncology in South China, Guangzhou, People's Republic of China ; Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Tao Qin
- State Key Laboratory of Oncology in South China, Guangzhou, People's Republic of China ; Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China ; Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Li-Bing Song
- State Key Laboratory of Oncology in South China, Guangzhou, People's Republic of China ; Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Xi Wang
- State Key Laboratory of Oncology in South China, Guangzhou, People's Republic of China ; Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China ; Department of Breast Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| |
Collapse
|
4
|
Kar A, Gutierrez-Hartmann A. Molecular mechanisms of ETS transcription factor-mediated tumorigenesis. Crit Rev Biochem Mol Biol 2013; 48:522-43. [PMID: 24066765 DOI: 10.3109/10409238.2013.838202] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The E26 transformation-specific (ETS) family of transcription factors is critical for development, differentiation, proliferation and also has a role in apoptosis and tissue remodeling. Changes in expression of ETS proteins therefore have a significant impact on normal physiology of the cell. Transcriptional consequences of ETS protein deregulation by overexpression, gene fusion, and modulation by RAS/MAPK signaling are linked to alterations in normal cell functions, and lead to unlimited increased proliferation, sustained angiogenesis, invasion and metastasis. Existing data show that ETS proteins control pathways in epithelial cells as well as stromal compartments, and the crosstalk between the two is essential for normal development and cancer. In this review, we have focused on ETS factors with a known contribution in cancer development. Instead of focusing on a prototype, we address cancer associated ETS proteins and have highlighted the diverse mechanisms by which they affect carcinogenesis. Finally, we discuss strategies for ETS factor targeting as a potential means for cancer therapeutics.
Collapse
|
5
|
Speiser JJ, Erşahin C, Osipo C. The functional role of Notch signaling in triple-negative breast cancer. VITAMINS AND HORMONES 2013; 93:277-306. [PMID: 23810012 DOI: 10.1016/b978-0-12-416673-8.00013-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The term "triple-negative breast cancer" (TNBC) is a heterogeneous subtype of breast cancer. Unfortunately, due to the lack of expression of hormone receptors and human epidermal growth factor receptor-2, therefore the lack of US Food and Drug Administration-approved targeted therapies, TNBC has the worst prognosis of all subtypes of breast cancer. Notch signaling has emerged as a pro-oncogene in several human malignancies and has particularly been associated with the triple-negative subtype of breast cancer. This chapter explores the role of Notch signaling in triple negative and other subtypes of breast cancer, the relationship of Notch with other breast cancer biomarkers, prognostic indicators associated with Notch, and potential therapeutic strategies targeting Notch inhibition. Hopefully, better understanding of this signaling pathway in the future will lead to optimal molecular therapeutic treatments for TNBC patients, improving their quality of life and outcome.
Collapse
Affiliation(s)
- Jodi J Speiser
- Department of Pathology, Loyola University Chicago Division of Health Sciences, Maywood, Illinois, USA
| | | | | |
Collapse
|
6
|
Oh S, Shin S, Janknecht R. ETV1, 4 and 5: an oncogenic subfamily of ETS transcription factors. Biochim Biophys Acta Rev Cancer 2012; 1826:1-12. [PMID: 22425584 DOI: 10.1016/j.bbcan.2012.02.002] [Citation(s) in RCA: 163] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Revised: 02/26/2012] [Accepted: 02/27/2012] [Indexed: 12/30/2022]
Abstract
The homologous ETV1, ETV4 and ETV5 proteins form the PEA3 subfamily of ETS transcription factors. In Ewing tumors, chromosomal translocations affecting ETV1 or ETV4 are an underlying cause of carcinogenesis. Likewise, chromosomal rearrangements of the ETV1, ETV4 or ETV5 gene occur in prostate tumors and are thought to be one of the major driving forces in the genesis of prostate cancer. In addition, these three ETS proteins are implicated in melanomas, breast and other types of cancer. Complex posttranslational modifications govern the activity of PEA3 factors, which can promote cell proliferation, motility and invasion. Here, we review evidence for a role of ETV1, 4 and 5 as oncoproteins and describe modes of their action. Modulation of their activation or interaction with cofactors as well as inhibiting crucial target gene products may ultimately be exploited to treat various cancers that are dependent on the PEA3 group of ETS transcription factors.
Collapse
Affiliation(s)
- Sangphil Oh
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | | | | |
Collapse
|
7
|
NOTCH-1 and NOTCH-4 are novel gene targets of PEA3 in breast cancer: novel therapeutic implications. Breast Cancer Res 2011; 13:R63. [PMID: 21679465 PMCID: PMC3218952 DOI: 10.1186/bcr2900] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2011] [Revised: 05/03/2011] [Accepted: 06/14/2011] [Indexed: 02/06/2023] Open
Abstract
Introduction Women with triple-negative breast cancer have the worst prognosis, frequently present with metastatic tumors and have few targeted therapy options. Notch-1 and Notch-4 are potent breast oncogenes that are overexpressed in triple-negative and other subtypes of breast cancer. PEA3, an ETS transcription factor, is also overexpressed in triple-negative and other breast cancer subtypes. We investigated whether PEA3 could be the critical transcriptional activator of Notch receptors in MDA-MB-231 and other breast cancer cells. Methods Real-time PCR and Western blot analysis were performed to detect Notch-1, Notch-2, Notch-3 and Notch-4 receptor expression in breast cancer cells when PEA3 was knocked down by siRNA. Chromatin immunoprecipitation was performed to identify promoter regions for Notch genes that recruited PEA3. TAM-67 and c-Jun siRNA were used to identify that c-Jun was necessary for PEA3 enrichment on the Notch-4 promoter. A Notch-4 luciferase reporter was used to confirm that endogenous PEA3 or AP-1 activated the Notch-4 promoter region. Cell cycle analysis, trypan blue exclusion, annexin V flow cytometry, colony formation assay and an in vivo xenograft study were performed to determine the biological significance of targeting PEA3 via siRNA, Notch signaling via a γ-secretase inhibitor, or both. Results Herein we provide new evidence for transcriptional regulation of Notch by PEA3 in breast cancer. PEA3 activates Notch-1 transcription in MCF-7, MDA-MB-231 and SKBr3 breast cancer cells. PEA3 activates Notch-4 transcription in MDA-MB-231 cells where PEA3 levels are endogenously high. In SKBr3 and BT474 breast cancer cells where PEA3 levels are low, overexpression of PEA3 increases Notch-4 transcripts. Chromatin immunoprecipitation confirmed the enrichment of PEA3 on Notch-1 and Notch-4 promoters in MDA-MB-231 cells. PEA3 recruitment to Notch-1 was AP-1-independent, whereas PEA3 recruitment to Notch-4 was c-JUN-dependent. Importantly, the combined inhibition of Notch signaling via a γ-secretase inhibitor (MRK-003 GSI) and knockdown of PEA3 arrested growth in the G1 phase, decreased both anchorage-dependent and anchorage-independent growth and significantly increased apoptotic cells in vitro. Moreover, either PEA3 knockdown or MRK-003 GSI treatment significantly reduced tumor growth of MDA-MB-231 xenografts in vivo. Conclusions Taken together, the results from this study demonstrate for the first time that Notch-1 and Notch-4 are novel transcriptional targets of PEA3 in breast cancer cells. Targeting of PEA3 and/or Notch pathways might provide a new therapeutic strategy for triple-negative and possibly other breast cancer subtypes.
Collapse
|
8
|
Tai W, Mahato R, Cheng K. The role of HER2 in cancer therapy and targeted drug delivery. J Control Release 2010; 146:264-75. [PMID: 20385184 DOI: 10.1016/j.jconrel.2010.04.009] [Citation(s) in RCA: 403] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2010] [Accepted: 04/05/2010] [Indexed: 12/16/2022]
Abstract
HER2 is highly expressed in a significant proportion of breast cancer, ovarian cancer, and gastric cancer. Since the discovery of its role in tumorigenesis, HER2 has received great attention in cancer research during the past two decades. Successful development of the humanized monoclonal anti-HER2 antibody (Trastuzumab) for the treatment of breast cancer further spurred scientists to develop various HER2 specific antibodies, dimerization inhibitors and kinase inhibitors for cancer therapy. On the other hand, the high expression of HER2 and the accessibility of its extracellular domain make HER2 an ideal target for the targeted delivery of anti-tumor drugs as well as imaging agents. Although there is no natural ligand for HER2, artificial ligands targeting HER2 have been developed and applied in various targeted drug delivery systems. The emphasis of this review is to elucidate the roles of HER2 in cancer therapy and targeted drug delivery. The structure and signal pathway of HER2 will be briefly described. The role of HER2 in tumorigenesis and its relationship with other tumor markers will be discussed. For the HER2 targeted cancer therapy, numerous strategies including the blockage of receptor dimerization, inhibition of the tyrosine kinase activity, and interruption of the downstream signal pathway will be summarized. For the targeted drug delivery to HER2 positive tumor cells, various targeting ligands and their delivery systems will be described in details.
Collapse
Affiliation(s)
- Wanyi Tai
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | | | | |
Collapse
|
9
|
Baker R, Kent CV, Silbermann RA, Hassell JA, Young LJT, Howe LR. Pea3 transcription factors and wnt1-induced mouse mammary neoplasia. PLoS One 2010; 5:e8854. [PMID: 20107508 PMCID: PMC2809747 DOI: 10.1371/journal.pone.0008854] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2009] [Accepted: 01/05/2010] [Indexed: 12/21/2022] Open
Abstract
The role of the PEA3 subfamily of Ets transcription factors in breast neoplasia is controversial. Although overexpression of PEA3 (E1AF/ETV4), and of the related factors ERM (ETV5) and ER81 (ETV1), have been observed in human and mouse breast tumors, PEA3 factors have also been ascribed a tumor suppressor function. Here, we utilized the MMTV/Wnt1 mouse strain to further interrogate the role of PEA3 transcription factors in mammary tumorigenesis based on our previous observation that Pea3 is highly expressed in MMTV/Wnt1 mammary tumors. Pea3 expression in mouse mammary tissues was visualized using a Pea3NLSlacZ reporter strain. In normal mammary glands, Pea3 expression is predominantly confined to myoepithelial cells. Wnt1 transgene expression induced marked amplification of this cell compartment in nontumorous mammary glands, accompanied by an apparent increase in Pea3 expression. The pattern of Pea3 expression in MMTV/Wnt1 mammary glands recapitulated the cellular profile of activated β-catenin/TCF signaling, which was visualized using both β-catenin immunohistochemistry and the β-catenin/TCF-responsive reporter Axin2NLSlacZ. To test the requirement for PEA3 factors in Wnt1-induced tumorigenesis, we employed a mammary-targeted dominant negative PEA3 transgene, ΔNPEA3En. Expression of ΔNPEA3En delayed early-onset tumor formation in MMTV/Wnt1 virgin females (P = 0.03), suggesting a requirement for PEA3 factor function for Wnt1-driven tumor formation. Consistent with this observation, expression of the ΔNPEA3En transgene was profoundly reduced in mammary tumors compared to nontumorous mammary glands from bigenic MMTV/Wnt1, MMTV/ΔNPEA3En mice (P = 0.01). Our data provide the first description of Wnt1-mediated expansion of the Pea3-expressing myoepithelial compartment in nontumorous mammary glands. Consistent with this observation, mammary myoepithelium was selectively responsive to Wnt1. Together these data suggest the MMTV/Wnt1 strain as a potential model of basal breast cancer. Furthermore, this study provides evidence for a protumorigenic role of PEA3 factors in breast neoplasia, and supports targeting the PEA3 transcription factor family in breast cancer.
Collapse
Affiliation(s)
- Rebecca Baker
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, New York, United States of America
- Strang Cancer Research Laboratory, Rockefeller University, New York, New York, United States of America
| | - Claire V. Kent
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, New York, United States of America
| | - Rachel A. Silbermann
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, New York, United States of America
- Strang Cancer Research Laboratory, Rockefeller University, New York, New York, United States of America
| | - John A. Hassell
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Lawrence J. T. Young
- Center for Comparative Medicine, University of California Davis, Davis, California, United States of America
| | - Louise R. Howe
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, New York, United States of America
- Strang Cancer Research Laboratory, Rockefeller University, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
10
|
Abstract
Profiling of differentially expressed proteins is perhaps the most important and useful approach in developing tools for risk assessment in a population, diagnostic screening, and therapeutics. Proteomic markers have potential for identifying individuals at high risk of developing cancer; however, these markers have not been extensively used in cancer epidemiologic studies. Several markers have to be clinically validated. In this chapter, methods used in proteomic analysis of clinical samples, challenges in the proteomics and cancer epidemiology, and their potential solutions are discussed.
Collapse
|
11
|
Myers E, Hill ADK, Kelly G, McDermott EW, O'Higgins NJ, Young LS. A positive role for PEA3 in HER2-mediated breast tumour progression. Br J Cancer 2006; 95:1404-9. [PMID: 17060941 PMCID: PMC2360603 DOI: 10.1038/sj.bjc.6603427] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Overexpression of HER2 is associated with an adverse prognosis in breast cancer. Despite this, the mechanism of its transcriptional regulation remains poorly understood. PEA3, a MAP kinase (MAPK)-activated member of the Ets transcription factor family has been implicated in the transcriptional regulation of HER2. The direction of its modulation remains controversial. We assessed relative levels of PEA3 expression and DNA binding in primary breast cultures derived from patient tumours (n=18) in the presence of an activated MAPK pathway using Western blotting and shift analysis. Expression of PEA3 in breast tumours from patients of known HER2 status (n=107) was examined by immunohistochemistry. In primary breast cancer cell cultures, growth factors induced interaction between PEA3 and its DNA response element. Upregulation of PEA3 expression in the presence of growth factors associated with HER2 positivity and axillary lymph node metastasis (P=0.034 and 0.049, respectively). PEA3 expression in breast cancer tissue associated with reduced disease-free survival (P<0.001), Grade III tumours (P<0.0001) and axillary lymph node metastasis (P=0.026). Co-expression of PEA3 and HER2 significantly associated with rate of recurrence compared to patients who expressed HER2 alone (P=0.0039). These data support a positive role for PEA3 in HER2-mediated oncogenesis in breast cancer.
Collapse
MESH Headings
- Axilla/pathology
- Biomarkers, Tumor/metabolism
- Blotting, Western
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Disease Progression
- Electrophoretic Mobility Shift Assay
- Epidermal Growth Factor/pharmacology
- Female
- Fibroblast Growth Factor 2/pharmacology
- Fluorescent Antibody Technique
- Gene Expression Regulation, Neoplastic
- Humans
- Immunoenzyme Techniques
- Lymph Nodes/pathology
- Lymphatic Metastasis/pathology
- Middle Aged
- Neoplasm Recurrence, Local/metabolism
- Neoplasm Recurrence, Local/pathology
- Promoter Regions, Genetic
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/metabolism
- Survival Rate
- Transcription Factors/metabolism
- Transcription, Genetic
- Tumor Cells, Cultured/drug effects
- raf Kinases/metabolism
Collapse
Affiliation(s)
- E Myers
- School of Medicine and Medical Science, Saint Vincent's University Hospital, Dublin, Ireland
- Conway Institute, University College Dublin, Dublin, Ireland
| | - A D K Hill
- School of Medicine and Medical Science, Saint Vincent's University Hospital, Dublin, Ireland
- Conway Institute, University College Dublin, Dublin, Ireland
| | - G Kelly
- School of Mathematical Sciences, Dublin, Ireland
| | - E W McDermott
- School of Medicine and Medical Science, Saint Vincent's University Hospital, Dublin, Ireland
| | - N J O'Higgins
- School of Medicine and Medical Science, Saint Vincent's University Hospital, Dublin, Ireland
| | - L S Young
- School of Medicine and Medical Science, Saint Vincent's University Hospital, Dublin, Ireland
- Conway Institute, University College Dublin, Dublin, Ireland
- Surgical Research Group, Royal College of Surgeons in Ireland, York House, York Street, Dublin 2, Ireland. E-mail:
| |
Collapse
|