1
|
Bidany-Mizrahi T, Shweiki A, Maroun K, Abu-Tair L, Mali B, Aqeilan RI. Unveiling the relationship between WWOX and BRCA1 in mammary tumorigenicity and in DNA repair pathway selection. Cell Death Discov 2024; 10:145. [PMID: 38499540 PMCID: PMC10948869 DOI: 10.1038/s41420-024-01878-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 02/07/2024] [Accepted: 02/20/2024] [Indexed: 03/20/2024] Open
Abstract
Breast cancer is the leading cause of cancer-related deaths in women worldwide, with the basal-like or triple-negative breast cancer (TNBC) subtype being particularly aggressive and challenging to treat. Understanding the molecular mechanisms driving the development and progression of TNBC is essential. We previously showed that WW domain-containing oxidoreductase (WWOX) is commonly inactivated in TNBC and is implicated in the DNA damage response (DDR) through ATM and ATR activation. In this study, we investigated the interplay between WWOX and BRCA1, both frequently inactivated in TNBC, on mammary tumor development and on DNA double-strand break (DSB) repair choice. We generated and characterized a transgenic mouse model (K14-Cre;Brca1fl/fl;Wwoxfl/fl) and observed that mice lacking both WWOX and BRCA1 developed basal-like mammary tumors and exhibited a decrease in 53BP1 foci and an increase in RAD51 foci, suggesting impaired DSB repair. We examined human TNBC cell lines harboring wild-type and mutant BRCA1 and found that WWOX expression promoted NHEJ repair in cells with wild-type BRCA1. Our findings suggest that WWOX and BRCA1 play an important role in DSB repair pathway choice in mammary epithelial cells, underscoring their functional interaction and significance in breast carcinogenesis.
Collapse
Affiliation(s)
- Tirza Bidany-Mizrahi
- The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Aya Shweiki
- The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Kian Maroun
- The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Lina Abu-Tair
- The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Bella Mali
- Department of Pathology, Hadassah University Hospital, Jerusalem, Israel
| | - Rami I Aqeilan
- The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
- Cyprus Cancer Research Institute (CCRI), Nicosia, Cyprus.
| |
Collapse
|
2
|
Pospiech K, Orzechowska M, Nowakowska M, Anusewicz D, Płuciennik E, Kośla K, Bednarek AK. TGFα-EGFR pathway in breast carcinogenesis, association with WWOX expression and estrogen activation. J Appl Genet 2022; 63:339-359. [PMID: 35290621 PMCID: PMC8979909 DOI: 10.1007/s13353-022-00690-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 02/22/2022] [Accepted: 03/02/2022] [Indexed: 11/29/2022]
Abstract
WWOX is a tumor-suppressive steroid dehydrogenase, which relationship with hormone receptors was shown both in animal models and breast cancer patients. Herein, through nAnT-iCAGE high-throughput gene expression profiling, we studied the interplay of estrogen receptors and the WWOX in breast cancer cell lines (MCF7, T47D, MDA-MB-231, BT20) under estrogen stimulation and either introduction of the WWOX gene by retroviral transfection (MDA-MB-231, T47D) or silenced with shRNA (MCF7, BT20). Additionally, we evaluated the consequent biological characteristics by proliferation, apoptosis, invasion, and adhesion assays. TGFα-EGFR signaling was found to be significantly affected in all examined breast cancer cell lines in response to estrogen and strongly associated with the level of WWOX expression, especially in ER-positive MCF7 cells. Under the influence of 17β-estradiol presence, biological characteristics of the cell lines were also delineated. The study revealed modulation of adhesion, invasion, and apoptosis. The obtained results point at a complex role of the WWOX gene in the carcinogenesis of the breast tissue, which seems to be closely related to the presence of estrogen α and/or β receptors.
Collapse
Affiliation(s)
- Karolina Pospiech
- Department of Molecular Carcinogenesis, Medical University of Lodz, Lodz, Poland
| | | | - Magdalena Nowakowska
- Department of Molecular Carcinogenesis, Medical University of Lodz, Lodz, Poland
| | - Dorota Anusewicz
- Department of Molecular Carcinogenesis, Medical University of Lodz, Lodz, Poland
| | - Elżbieta Płuciennik
- Department of Molecular Carcinogenesis, Medical University of Lodz, Lodz, Poland
| | - Katarzyna Kośla
- Department of Molecular Carcinogenesis, Medical University of Lodz, Lodz, Poland
| | - Andrzej K Bednarek
- Department of Molecular Carcinogenesis, Medical University of Lodz, Lodz, Poland.
| |
Collapse
|
3
|
Kałuzińska Ż, Kołat D, Kośla K, Orzechowska M, Bednarek AK, Płuciennik E. In vitro and in silico assessment of the effect of WWOX expression on invasiveness pathways associated with AP-2 transcription factors in bladder cancer. BMC Urol 2021; 21:36. [PMID: 33691672 PMCID: PMC7944886 DOI: 10.1186/s12894-021-00806-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 03/02/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND WW Domain Containing Oxidoreductase (WWOX) belongs to the unusual tumor suppressors, whose molecular function is not fully understood in bladder cancer, especially regarding interaction with Activator Protein 2 (AP-2) α/γ transcription factors. Thus, using lentiviral systems we created an in vitro model overexpressing or downregulating WWOX in CAL-29 cell line to assess invasiveness pathways. Surprisingly, while WWOX overexpression was accompanied with increased expression of both AP-2 factors, its downregulation only affected AP-2α level but not AP-2γ which remained high. METHODS Using cellular models and unpaired t-test or Wilcoxon test, we investigated significant changes in biological processes: clonogenicity, extracellular matrix adhesion, metalloproteinases activity, 3D culture growth, proliferation, mitochondrial redox potential and invasiveness. Relative gene expression acquired through Real-Time qPCR has been analyzed by Welch's t-test. Additionally, using oncoprint analysis we distinguished groups for bioinformatics analyzes in order to perform a follow-up of in vitro experiments. RESULTS Downregulation of WWOX in bladder cancer cell line intensified ability of single cell to grow into colony, mitochondrial redox potential and proliferation rate. Moreover, these cells shown elevated pro-MMP-2/9 activity but reduced adhesion to collagen I or laminin I, as well as distinct 3D culture growth. Through global in silico profiling we determined that WWOX alters disease-free survival of bladder cancer patients and modulates vital processes through AP-2 downstream effectors. CONCLUSIONS Our research indicates that WWOX possesses tumor suppressor properties in bladder cancer but consecutive examination is required to entirely understand the contribution of AP-2γ or AP-2α.
Collapse
Affiliation(s)
- Żaneta Kałuzińska
- Department of Molecular Carcinogenesis, Medical University of Lodz, 90-752, Lodz, Poland.
| | - Damian Kołat
- Department of Molecular Carcinogenesis, Medical University of Lodz, 90-752, Lodz, Poland
| | - Katarzyna Kośla
- Department of Molecular Carcinogenesis, Medical University of Lodz, 90-752, Lodz, Poland
| | - Magdalena Orzechowska
- Department of Molecular Carcinogenesis, Medical University of Lodz, 90-752, Lodz, Poland
| | - Andrzej K Bednarek
- Department of Molecular Carcinogenesis, Medical University of Lodz, 90-752, Lodz, Poland
| | - Elżbieta Płuciennik
- Department of Molecular Carcinogenesis, Medical University of Lodz, 90-752, Lodz, Poland
| |
Collapse
|
4
|
Abdeen SK, Aqeilan RI. Decoding the link between WWOX and p53 in aggressive breast cancer. Cell Cycle 2019; 18:1177-1186. [PMID: 31075076 PMCID: PMC6592247 DOI: 10.1080/15384101.2019.1616998] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 04/14/2019] [Accepted: 04/18/2019] [Indexed: 10/26/2022] Open
Abstract
Basal-like breast cancer (BLBC) and triple-negative breast cancer (TNBC) are aggressive forms of human breast cancer with poor prognosis and limited treatment response. Molecular understanding of BLBC and TNBC biology is instrumental to improve detection and management of these deadly diseases. Tumor suppressors WW domain-containing oxidoreductase (WWOX) and TP53 are altered in BLBC and in TNBC. Nevertheless, the functional interplay between WWOX and p53 is poorly understood. In a recent study by Abdeen and colleagues, it has been demonstrated that WWOX loss drives BLBC formation via deregulating p53 functions. In this review, we highlight important signaling pathways regulated by WWOX and p53 that are related to estrogen receptor signaling, epithelial-to-mesenchymal transition, and genomic instability and how they impact BLBC and TNBC development.
Collapse
Affiliation(s)
- Suhaib K. Abdeen
- Lautenberg Center for Immunology and Cancer Research, IMRIC, Hebrew University-Hadassah Medical School, IMRIC, Jerusalem, Israel
| | - Rami I. Aqeilan
- Lautenberg Center for Immunology and Cancer Research, IMRIC, Hebrew University-Hadassah Medical School, IMRIC, Jerusalem, Israel
- Department of Cancer Biology and Genetics, The Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
5
|
Khawaled S, Suh SS, Abdeen SK, Monin J, Distefano R, Nigita G, Croce CM, Aqeilan RI. WWOX Inhibits Metastasis of Triple-Negative Breast Cancer Cells via Modulation of miRNAs. Cancer Res 2019; 79:1784-1798. [PMID: 30622118 DOI: 10.1158/0008-5472.can-18-0614] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 04/30/2018] [Accepted: 01/03/2019] [Indexed: 11/16/2022]
Abstract
Triple-negative breast cancer (TNBC) is a heterogeneous, highly aggressive, and difficult to treat tumor type. The tumor suppressor WWOX spans FRA16D, a common fragile site that is commonly altered in breast cancer. Despite recent progress, the role of WWOX in TNBC metastasis is unknown. Here we report that WWOX inactivation correlates with advanced stages of TNBC and that its levels are frequently altered in TNBC cells. Ectopic restoration of WWOX in WWOX-negative TNBC cells inhibited metastasis while its depletion in WWOX-positive TNBC cells promoted metastasis. WWOX was a negative regulator of c-MYC, which regulated miR-146a expression and consequently fibronectin levels, contributing to an epithelial status of the cell. Treatment of TNBC cells with anti-miR-146a rescued the WWOX antimetastatic phenotype. Moreover, overexpression of MYC in WWOX-expressing TNBC cells overrode WWOX effects on miR-146a and fibronectin levels. Altogether, our data uncover an essential role for WWOX in antagonizing TNBC progression and highlight its potential use as a biomarker for metastasis. SIGNIFICANCE: These findings highlight the mechanism by which the tumor suppressor WWOX regulates metastasis of triple-negative breast cancer.See related commentary by Sharma, p. 1746.
Collapse
Affiliation(s)
- Saleh Khawaled
- Lautenberg Center for Immunology and Cancer Research, Hebrew University-Hadassah Medical School, IMRIC, Jerusalem, Israel
| | - Sung Suk Suh
- Department of Bioscience, Mokpo National University, Muan, Republic of Korea
| | - Suhaib K Abdeen
- Lautenberg Center for Immunology and Cancer Research, Hebrew University-Hadassah Medical School, IMRIC, Jerusalem, Israel
| | - Jonathan Monin
- Lautenberg Center for Immunology and Cancer Research, Hebrew University-Hadassah Medical School, IMRIC, Jerusalem, Israel
| | - Rosario Distefano
- Department of Cancer Biology and Genetics, Wexner Medical Center, The Ohio State University, Columbus, Ohio
| | - Giovanni Nigita
- Department of Cancer Biology and Genetics, Wexner Medical Center, The Ohio State University, Columbus, Ohio
| | - Carlo M Croce
- Department of Cancer Biology and Genetics, Wexner Medical Center, The Ohio State University, Columbus, Ohio
| | - Rami I Aqeilan
- Lautenberg Center for Immunology and Cancer Research, Hebrew University-Hadassah Medical School, IMRIC, Jerusalem, Israel. .,Department of Cancer Biology and Genetics, Wexner Medical Center, The Ohio State University, Columbus, Ohio
| |
Collapse
|
6
|
Jamous A, Salah Z. WW-Domain Containing Protein Roles in Breast Tumorigenesis. Front Oncol 2018; 8:580. [PMID: 30619734 PMCID: PMC6300493 DOI: 10.3389/fonc.2018.00580] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 11/19/2018] [Indexed: 12/13/2022] Open
Abstract
Protein-protein interactions are key factors in executing protein function. These interactions are mediated through different protein domains or modules. An important domain found in many different types of proteins is WW domain. WW domain-containing proteins were shown to be involved in many human diseases including cancer. Some of these proteins function as either tumor suppressor genes or oncogenes, while others show dual identity. Some of these proteins act on their own and alter the function(s) of specific or multiple proteins implicated in cancer, others interact with their partners to compose WW domain modular pathway. In this review, we discuss the role of WW domain-containing proteins in breast tumorigenesis. We give examples of specific WW domain containing proteins that play roles in breast tumorigenesis and explain the mechanisms through which these proteins lead to breast cancer initiation and progression. We discuss also the possibility of using these proteins as biomarkers or therapeutic targets.
Collapse
Affiliation(s)
- Abrar Jamous
- Al Quds-Bard College for Arts and Sciences, Al Quds University, Abu Dis, Palestine
| | - Zaidoun Salah
- Al Quds-Bard College for Arts and Sciences, Al Quds University, Abu Dis, Palestine
| |
Collapse
|
7
|
Chang R, Song L, Xu Y, Wu Y, Dai C, Wang X, Sun X, Hou Y, Li W, Zhan X, Zhan L. Loss of Wwox drives metastasis in triple-negative breast cancer by JAK2/STAT3 axis. Nat Commun 2018; 9:3486. [PMID: 30154439 PMCID: PMC6113304 DOI: 10.1038/s41467-018-05852-8] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 07/11/2018] [Indexed: 12/19/2022] Open
Abstract
Loss of WW domain-containing oxidoreductase (Wwox) expression has been observed in breast cancer (BC). However, its regulatory effects are largely unknown, especially in triple-negative breast cancer (TNBC). Herein, gene expression profiling revealed that JAK/STAT3 pathway was one of the most differentially modulated pathways in basal-like BC cells. The lower expression of Wwox was significantly correlated with high activation of STAT3 in basal-like cells and TNBC tissues. Overexpression of Wwox markedly inhibited proliferation and metastasis of BC cells by suppressing STAT3 activation, which is to interact with JAK2 to inhibit JAK2 and STAT3 phosphorylation. Furthermore, Wwox limited STAT3 binding to the interleukin-6 promoter, repressing expression of the IL-6 cytokine. Altogether, our data established that Wwox suppresses BC cell metastasis and proliferation by JAK2/STAT3 pathway. Targeting of Wwox with STAT3 could offer a promising therapeutic strategy for TNBC. In breast cancer, the loss of expression of WW domain-containing oxireductase (Wwox) has been observed. Here, the authors illustrate that in triple negative breast cancer models Wwox suppresses metastasis and proliferation via the JAK2/STAT3 pathway.
Collapse
Affiliation(s)
- Renxu Chang
- Key Laboratory of Nutrition, Metabolism, and Food Safety, Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of the Chinese Academy of Sciences, Shanghai, 200031, China
| | - Lele Song
- Key Laboratory of Nutrition, Metabolism, and Food Safety, Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of the Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yi Xu
- Key Laboratory of Nutrition, Metabolism, and Food Safety, Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of the Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yanjun Wu
- Key Laboratory of Nutrition, Metabolism, and Food Safety, Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of the Chinese Academy of Sciences, Shanghai, 200031, China
| | - Cheng Dai
- Key Laboratory of Nutrition, Metabolism, and Food Safety, Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of the Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xinyu Wang
- Key Laboratory of Nutrition, Metabolism, and Food Safety, Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of the Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xia Sun
- Key Laboratory of Nutrition, Metabolism, and Food Safety, Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of the Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wei Li
- Department of General Surgery, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Zhejiang, 310020, China
| | - Xianbao Zhan
- Department of Oncology, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Lixing Zhan
- Key Laboratory of Nutrition, Metabolism, and Food Safety, Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of the Chinese Academy of Sciences, Shanghai, 200031, China. .,Department of Cellular and Genetic Medicine, Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
8
|
Pospiech K, Płuciennik E, Bednarek AK. WWOX Tumor Suppressor Gene in Breast Cancer, a Historical Perspective and Future Directions. Front Oncol 2018; 8:345. [PMID: 30211123 PMCID: PMC6121138 DOI: 10.3389/fonc.2018.00345] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 08/06/2018] [Indexed: 11/18/2022] Open
Abstract
The WWOX tumor suppressor gene is located at 16q23. 1–23.2, which covers the region of FRA16D—a common fragile sites. Deletions within the WWOX coding sequence are observed in up to 80% of breast cancer cases, which makes it one of the most common genetic alterations in this tumor type. The WWOX gene is known to play a role in breast cancer: increased expression of WWOX inhibits cell proliferation in suspension, reduces tumor growth rates in xenographic transplants, but also enhances cell migration through the basal membrane and contributes to morphological changes in 3D matrix-based cell cultures. The WWOX protein may act in several ways, as it has three functional domains—two WW domains, responsible for protein-protein interactions and an SDR domain (short dehydrogenase/reductase domain) which catalyzes conversions of low molecular weight ligands, most likely steroids. In epithelial cells, WWOX modulates gene transcription through interaction with p73, AP-2γ, and ERBB4 proteins. In steroid hormone-regulated tissues like mammary gland epithelium, the WWOX SDR domain acts as a steroid dehydrogenase. The relationship between WWOX and hormone receptors was shown in an animal model, where WWOX(C3H)+/–mice exhibited loss of both ER and PR receptors. Moreover, in breast cancer specimens, a positive correlation was observed between WWOX expression and ER status. On the other hand, decreased WWOX expression was associated with worse prognosis, namely higher relapse and mortality rates in BC patients. Recently, it was shown that genomic instability might be driven by the loss of WWOX expression. It was reported that WWOX plays role in DNA damage response (DDR) and DNA repair by regulating ATM activation through physical interaction. A genome caretaker function has also been proposed for WWOX, as it was found that WWOX sufficiency decreases homology directed repair (HDR) and supports non-homologous end-joining (NHEJ) repair as the dominant DSB repair pathway by Brca1-Wwox interaction. In breast cancer cells, WWOX was also found to modulate the expression of glycolysis pathway genes, through hypoxia-inducible transcription factor 1α (HIF1α) regulation. The paper presents the current state of knowledge regarding the WWOX tumor suppressor gene in breast cancer, as well as future research perspectives.
Collapse
Affiliation(s)
- Karolina Pospiech
- Department of Molecular Carcinogenesis, Medical University of Lodz, Lodz, Poland
| | - Elzbieta Płuciennik
- Department of Molecular Carcinogenesis, Medical University of Lodz, Lodz, Poland
| | - Andrzej K Bednarek
- Department of Molecular Carcinogenesis, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
9
|
Somatic loss of WWOX is associated with TP53 perturbation in basal-like breast cancer. Cell Death Dis 2018; 9:832. [PMID: 30082886 PMCID: PMC6079009 DOI: 10.1038/s41419-018-0896-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 07/11/2018] [Accepted: 07/12/2018] [Indexed: 12/16/2022]
Abstract
Inactivation of WW domain-containing oxidoreductase (WWOX), the gene product of the common fragile site FRA16D, is a common event in breast cancer and is associated with worse prognosis of triple-negative breast cancer (TNBC) and basal-like breast cancer (BLBC). Despite recent progress, the role of WWOX in driving breast carcinogenesis remains unknown. Here we report that ablation of Wwox in mammary tumor-susceptible mice results in increased tumorigenesis, and that the resultant tumors resemble human BLBC. Interestingly, copy number loss of Trp53 and downregulation of its transcript levels were observed in the Wwox knockout tumors. Moreover, tumors isolated from Wwox and Trp53 mutant mice were indistinguishable histologically and transcriptionally. Finally, we find that deletion of TP53 and WWOX co-occurred and is associated with poor survival of breast cancer patients. Altogether, our data uncover an essential role for WWOX as a bona fide breast cancer tumor suppressor through the maintenance of p53 stability.
Collapse
|
10
|
Choi HJ, Park JH, Park JH, Lee KB, Oh SM. Pc2-mediated SUMOylation of WWOX is essential for its suppression of DU145 prostate tumorigenesis. FEBS Lett 2015; 589:3977-88. [PMID: 26592150 DOI: 10.1016/j.febslet.2015.11.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 11/11/2015] [Accepted: 11/18/2015] [Indexed: 11/19/2022]
Abstract
Tumor suppressor WW domain-containing oxidoreductase (WWOX) is depleted in various cancer types. Here we report that WWOX is modified by small ubiquitin-like modifier (SUMO) proteins and represses DU145 prostate cancer tumorigenesis in a SUMOylation-dependent manner. Ectopic WWOX was shown to associate with SUMO2/3 or E2 Ubc9. Furthermore, we revealed that WWOX SUMOylation was promoted by E3 ligase polycomb2 (Pc2), and that WWOX associated with Pc2. Meanwhile, anisomycin-induced activator protein-1 (AP-1) activity was markedly diminished by co-expression of SUMO and WWOX. Also, WWOX wild type (WT), but not WWOX SUMO mutant (K176A) markedly reduced both DU145 prostate cancer cell proliferation and xenograft tumorigenesis. Collectively, our findings demonstrate that SUMO modification of WWOX is essential for its suppressive activity for DU145 prostate cancer tumorigenesis.
Collapse
Affiliation(s)
- Hye-Jin Choi
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 302-832, Republic of Korea
| | - Jung-Hwan Park
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 302-832, Republic of Korea
| | - Jong-Hwan Park
- Laboratory Animal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju 500-757, Republic of Korea
| | - Kyung Bok Lee
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 302-832, Republic of Korea
| | - Sang-Muk Oh
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 302-832, Republic of Korea.
| |
Collapse
|
11
|
Zhang M, Zhang X, Zhao S, Wang Y, Di W, Zhao G, Yang M, Zhang Q. Prognostic value of survivin and EGFR protein expression in triple-negative breast cancer (TNBC) patients. Target Oncol 2015; 9:349-57. [PMID: 24233638 DOI: 10.1007/s11523-013-0300-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Accepted: 10/30/2013] [Indexed: 12/12/2022]
Abstract
Triple-negative breast cancer (TNBC) is a particular type of breast cancer which is characterized by its biological aggressiveness, worse prognosis, and lack of prognostic markers or therapeutic targets in contrast with hormonal receptor-positive and human epidermal growth factor receptor 2-positive (HER2+) breast cancers. We aimed to evaluate survivin and epidermal growth factor receptor (EGFR) expression and their prognostic value and determine their relationships with the clinicopathological parameters of TNBC. A total of 136 patients who had undergone a resection of primary TNBC were enrolled at the Third Affiliated Hospital of Harbin Medical University from March 2003 to September 2005. Expression of ER, PR, HER2, EGFR, and survivin was assessed by immunohistochemistry. The association of TNBC and other clinicopathological variables and the prognostic value of survivin and EGFR expression were evaluated. Survivin was expressed in 62 (45.6 %) cases and EGFR was expressed in 82 (60.3 %) cases. Survivin expression was associated with menopausal status (P = 0.011), tumor size (P = 0.037), and lymph node status (P = 0.001). EGFR expression was associated with menopausal status (P = 0.029), lymph node status (P = 0.004), P53 expression (P = 0.001), Ki-67 expression (P = 0.028), and lymphatic vascular invasion (P = 0.037). A multivariate analysis demonstrated that tumor size (hazard ratio (HR) 1.587, 95 % confidence interval (CI) 1.081–2.330, P = 0.018 for disease-free survival (DFS); HR 1.606, 95%CI 1.096–2.354, P = 0.015 for overall survival (OS)), lymph node status (HR 2.873, 95%CI 1.544–5.344, P = 0.001 for DFS; HR 2.915, 95%CI 1.553–5.471, P = 0.001 for OS), tumor grade (HR 1.914, 95%CI 1.218–3.007, P = 0.005 for DFS; HR 1.983, 95%CI 1.228–3.203, P = 0.005 for OS), EGFR (HR 3.008, 95%CI 1.331–6.792, P = 0.008 for DFS; HR 3.151, 95%CI 1.374–7.226, P = 0.007 for OS), and survivin (HR 1.573, 95%CI 1.087–2.277, P = 0.016 for DFS; HR 1.607, 95%CI 1.088–2.374, P = 0.017 for OS) were of prognostic significance for disease-free and overall survival. We draw a conclusion from the present study that survivin and EGFR expression are useful prognostic markers of TNBC and might be useful for molecular targeting therapy of TNBC treatment.
Collapse
|
12
|
Płuciennik E, Nowakowska M, Pospiech K, Stępień A, Wołkowicz M, Gałdyszyńska M, Popęda M, Wójcik-Krowiranda K, Bieńkiewicz A, Bednarek AK. The role of WWOX tumor suppressor gene in the regulation of EMT process via regulation of CDH1-ZEB1-VIM expression in endometrial cancer. Int J Oncol 2015; 46:2639-48. [PMID: 25892250 DOI: 10.3892/ijo.2015.2964] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 03/20/2015] [Indexed: 11/06/2022] Open
Abstract
This study defines the role of WWOX in the regulation of epithelial to mesenchymal transition. A group of 164 endometrial adenocarcinoma patients was studied as well as an ECC1 well-differentiated steroid-responsive endometrial cell line, which was transducted with WWOX cDNA by a retroviral system. The relationship between WWOX gene and EMT marker (CDH1, VIM, ZEB1, SNAI1) expression on mRNA (RT-qPCR) and protein levels (western blotting) was evaluated. The EMT processes were also analysed in vitro by adhesion of cells to extracellular matrix proteins, migration through a basement membrane, anchorage-independent growth and MMP activity assay. DNA microarrays (HumanOneArray™) were used to determine WWOX-dependent pathways in an ECC1 cell line. A positive correlation was observed between WWOX and ZEB1, and a negative correlation between CDH1 and VIM. WWOX expression was found to inversely correlate with the risk of recurrence of tumors in patients. However, in the WWOX-expressing ECC1 cell line, WWOX expression was found to be inversely related with VIM and positively with CDH1. The ECC1/WWOX cell line variant demonstrated increased migratory capacity, with increased expression of metalloproteinases MMP2/MMP9. However, these cells were not able to form colonies in suspension and revealed decreased adhesion to fibronectin and fibrinogen. Microarray analysis demonstrated that WWOX has an impact on the variety of cellular pathways including the cadherin and integrin signalling pathways. Our results suggest that the WWOX gene plays a role in the regulation of EMT processes in endometrial cancer by controlling the expression of proteins associated with cell motility, thus influencing tissue remodeling, with the suppression of mesenchymal markers.
Collapse
Affiliation(s)
- Elżbieta Płuciennik
- Department of Molecular Carcinogenesis, Medical University of Lodz, PL 90-752 Lodz, Poland
| | - Magdalena Nowakowska
- Department of Molecular Carcinogenesis, Medical University of Lodz, PL 90-752 Lodz, Poland
| | - Karolina Pospiech
- Department of Molecular Carcinogenesis, Medical University of Lodz, PL 90-752 Lodz, Poland
| | - Anna Stępień
- Laboratory of Clinical and Transplant Immunology and Genetics, Copernicus Memorial Hospital in Lodz, PL 90-752 Lodz, Poland
| | - Mateusz Wołkowicz
- Bio-Tech Consulting Sp. z o.o, Faculty of Biomedical Science and Postgraduate Education, Medical University of Lodz, PL 90-752 Lodz, Poland
| | - Małgorzata Gałdyszyńska
- Faculty of Biomedical Sciences and Postgraduate Education, Medical University of Lodz, PL 90-752 Lodz, Poland
| | - Marta Popęda
- Faculty of Biomedical Sciences and Postgraduate Education, Medical University of Lodz, PL 90-752 Lodz, Poland
| | | | - Andrzej Bieńkiewicz
- Clinical Division of Gynecological Oncology, Medical University of Lodz, PL 90-752 Lodz, Poland
| | - Andrzej K Bednarek
- Department of Molecular Carcinogenesis, Medical University of Lodz, PL 90-752 Lodz, Poland
| |
Collapse
|
13
|
Abstract
WWOX, the WW domain-containing oxidoreductase gene at chromosome region 16q23.3-q24.1, spanning chromosomal fragile site FRA16D, encodes the 46 kDa Wwox protein, a tumor suppressor that is lost or reduced in expression in a wide variety of cancers, including breast, prostate, ovarian, and lung. The function of Wwox as a tumor suppressor implies that it serves a function in the prevention of carcinogenesis. Indeed, in vitro studies show that Wwox protein interacts with many binding partners to regulate cellular apoptosis, proliferation, and/or maturation. It has been reported that newborn Wwox knockout mice exhibit nascent osteosarcomas while Wwox(+/-) mice exhibit increased incidence of spontaneous and induced tumors. Furthermore, absence or reduction of Wwox expression in mouse xenograft models results in increased tumorigenesis, which can be rescued by Wwox re-expression, though there is not universal agreement among investigators regarding the role of Wwox loss in these experimental models. Despite this proposed tumor suppressor function, the overlap of the human WWOX locus with FRA16D sensitizes the gene to protein-inactivating deletions caused by replication stress. The high frequency of deletions within the WWOX locus in cancers of various types, without the hallmark protein inactivation-associated mutations of "classical" tumor suppressors, has led to the proposal that WWOX deletions in cancers are passenger events that occur in early cancer progenitor cells due to fragility of the genetic locus, rather than driver events which provide the cancer cell a selective advantage. Recently, a proposed epigenetic cause of chromosomal fragility has suggested a novel mechanism for early fragile site instability and has implications regarding the involvement of tumor suppressor genes at chromosomal fragile sites in cancer. In this review, we provide an overview of the evidence for WWOX as a tumor suppressor gene and put this into the context of fragility associated with the FRA16D locus.
Collapse
Affiliation(s)
- Morgan S Schrock
- Biomedical Sciences Graduate Program, Ohio State University Wexner Medical Center, Columbus, Ohio 43210, USA Department of Molecular Virology, Immunology and Medical Genetics, Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, Ohio 43210, USA
| | - Kay Huebner
- Department of Molecular Virology, Immunology and Medical Genetics, Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, Ohio 43210, USA
| |
Collapse
|
14
|
Li J, Liu J, Ren Y, Liu P. Roles of the WWOX in pathogenesis and endocrine therapy of breast cancer. Exp Biol Med (Maywood) 2015; 240:324-8. [PMID: 25476151 PMCID: PMC4935229 DOI: 10.1177/1535370214561587] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Breast cancer is one of the most common malignancies, often with complicated etiology and poor clinical outcome. In recent years, a critical role has emerged for the WW domain-containing oxidoreductase (WWOX) in breast cancer. WWOX is a tumor suppressor; it is deleted or attenuated in 29-63.2% of breast cancer tissues and is associated with a poor prognosis of breast cancer patients. WWOX heterozygous knockout mice show a higher incidence of mammary tumors and impaired branching morphogenesis. At the molecular level, WWOX interacts with AP2γ, ErbB4, SMAD3, and WBP2 suppressing their transcription activities in breast cancer cell lines. This review provides comprehensive insights into the current knowledge of WWOX activities in the pathogenesis and endocrine therapy of breast cancer.
Collapse
Affiliation(s)
- Juan Li
- Center for Translational Medicine, The First Affiliated Hospital, Xian Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, PR China
| | - Jie Liu
- Center for Translational Medicine, The First Affiliated Hospital, Xian Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, PR China
| | - Yu Ren
- Department of Surgical Oncology, The First Affiliated Hospital, Xian Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, PR China
| | - Peijun Liu
- Center for Translational Medicine, The First Affiliated Hospital, Xian Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, PR China
| |
Collapse
|
15
|
Huang C, Tian Y, Peng R, Zhang C, Wang D, Han S, Jiao C, Wang X, Zhang H, Wang Y, Li X. Association of downregulation of WWOX with poor prognosis in patients with intrahepatic cholangiocarcinoma after curative resection. J Gastroenterol Hepatol 2015; 30:421-433. [PMID: 25168293 DOI: 10.1111/jgh.12722] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/03/2014] [Indexed: 12/30/2022]
Abstract
BACKGROUND AND AIM Downregulation of the WW domain containing oxidoreductase (WWOX) has been reported to be involved in tumorigenesis in several neoplasms. This study sought to investigate the expression and role of WWOX in intrahepatic cholangiocarcinoma (ICC). METHODS WWOX expression was measured by quantitative real-time polymerase chain reaction (PCR), immunoblot, immunofluorescence, and immunohistochemistry. The prognostic significance was assessed by Kaplan-Meier and Cox regression analyses. The role of WWOX in proliferation, anchorage-independent growth, gene expression regulation, and tumorigenesis was assessed by WWOX re-expression using lentivirus. Methylation-specific PCR was performed to evaluate the methylation status of the WWOX gene regulatory region. A DNA methyltransferase inhibitor, 5-aza-2'-deoxycytidine (AZA), was used to activate the endogenous WWOX gene in ICC cells both in vitro and in vivo. RESULTS The expression of WWOX in ICC tissues was much lower than that in nontumorous samples and showed reverse correlation with proliferative status. Restoration of WWOX expression resulted in suppression of the growth of WWOX-deficient ICC cells through activation of the intrinsic apoptotic signaling pathway, but did not affect growth of WWOX-sufficient human intrahepatic biliary epithelial derived non-cancer cells. Multivariate analyses revealed that downregulation of WWOX was an unfavorable predictor for overall survival and cumulative recurrence rates. The WWOX gene regulatory region was frequently methylated in ICC tissues and cell lines, and intratumoral WWOX restoration, through AZA injection, suppressed tumor growth in nude mice. CONCLUSION Downregulation of WWOX may occur as a result of hypermethylation and implies a poor prognosis in ICC; WWOX re-expression may be a potential molecular therapeutic target for ICC.
Collapse
Affiliation(s)
- Changjun Huang
- Department of General Surgery, Luohe Central Hospital Affiliated to Luohe Medical College, Luohe, China; Liver Transplantation Center, First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Living Donor Liver Transplantation, Ministry of Public Health, Nanjing, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Lo JY, Chou YT, Lai FJ, Hsu LJ. Regulation of cell signaling and apoptosis by tumor suppressor WWOX. Exp Biol Med (Maywood) 2015; 240:383-91. [PMID: 25595191 DOI: 10.1177/1535370214566747] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Human fragile WWOX gene encodes a tumor suppressor WW domain-containing oxidoreductase (named WWOX, FOR, or WOX1). Functional suppression of WWOX prevents apoptotic cell death induced by a variety of stress stimuli, such as tumor necrosis factor, UV radiation, and chemotherapeutic drug treatment. Loss of WWOX gene expression due to gene deletions, loss of heterozygosity, chromosomal translocations, or epigenetic silencing is frequently observed in human malignant cancer cells. Acquisition of chemoresistance in squamous cell carcinoma, osteosarcoma, and breast cancer cells is associated with WWOX deficiency. WWOX protein physically interacts with many signaling molecules and exerts its regulatory effects on gene transcription and protein stability and subcellular localization to control cell survival, proliferation, differentiation, autophagy, and metabolism. In this review, we provide an overview of the recent advances in understanding the molecular mechanisms by which WWOX regulates cellular functions and stress responses. A potential scenario is that activation of WWOX by anticancer drugs is needed to overcome chemoresistance and trigger cancer cell death, suggesting that WWOX can be regarded as a prognostic marker and a candidate molecule for targeted cancer therapies.
Collapse
Affiliation(s)
- Jui-Yen Lo
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, Tainan 70101, Taiwan
| | - Ying-Tsen Chou
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, Tainan 70101, Taiwan
| | - Feng-Jie Lai
- Department of Dermatology, Chimei Medical Center, Tainan 71004, Taiwan
| | - Li-Jin Hsu
- Department of Medical Laboratory Science and Biotechnology Center of Infectious Disease and Signaling Research and Research Center for Medical Laboratory Biotechnology, National Cheng Kung University Medical College, Tainan 70101, Taiwan
| |
Collapse
|
17
|
Ge F, Chen W, Yang R, Zhou Z, Chang N, Chen C, Zou T, Liu R, Tan J, Ren G. WWOX suppresses KLF5 expression and breast cancer cell growth. Chin J Cancer Res 2014; 26:511-6. [PMID: 25400415 DOI: 10.3978/j.issn.1000-9604.2014.09.03] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 08/05/2014] [Indexed: 12/31/2022] Open
Abstract
The WW domain-containing oxidoreductase (WWOX) is a tumor suppressor in a variety of cancers, including breast cancer. Reduced WWOX expression is associated with the basal-like subtype and a relatively poor disease-free survival rate among breast cancer patients. Though several WWOX partners have been identified, the functional mechanisms of WWOX's role in cancers have not been fully addressed to date. In the current study, we found WWOX suppresses expression of KLF5-an oncogenic transcription factor-at protein level, and suppresses cancer cell proliferation in both bladder and breast cancer cell lines. Furthermore, we demonstrated that WWOX physically interacts with KLF5 via the former's WW domains and the latter's PY motifs. Interestingly, we found the expression of WWOX negatively correlates with KLF5 expression in a panel of breast cancer cell lines. Taken together, we conjecture that WWOX may suppress cancer cell proliferation partially by reducing the expression of KLF5.
Collapse
Affiliation(s)
- Fei Ge
- 1 Department of Endocrine Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China ; 2 Department of Breast Surgery, 3 Second Department of Internal Medicine of Oncology, The 3rd Affiliated Hospital of Kunming Medical University, Kunming 650118, China ; 4 Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China ; 5 Institute of Molecular Medicine, National Cheng Kung University Medical College, Tainan, Taiwan, China
| | - Wenlin Chen
- 1 Department of Endocrine Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China ; 2 Department of Breast Surgery, 3 Second Department of Internal Medicine of Oncology, The 3rd Affiliated Hospital of Kunming Medical University, Kunming 650118, China ; 4 Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China ; 5 Institute of Molecular Medicine, National Cheng Kung University Medical College, Tainan, Taiwan, China
| | - Runxiang Yang
- 1 Department of Endocrine Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China ; 2 Department of Breast Surgery, 3 Second Department of Internal Medicine of Oncology, The 3rd Affiliated Hospital of Kunming Medical University, Kunming 650118, China ; 4 Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China ; 5 Institute of Molecular Medicine, National Cheng Kung University Medical College, Tainan, Taiwan, China
| | - Zhongmei Zhou
- 1 Department of Endocrine Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China ; 2 Department of Breast Surgery, 3 Second Department of Internal Medicine of Oncology, The 3rd Affiliated Hospital of Kunming Medical University, Kunming 650118, China ; 4 Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China ; 5 Institute of Molecular Medicine, National Cheng Kung University Medical College, Tainan, Taiwan, China
| | - Nanshan Chang
- 1 Department of Endocrine Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China ; 2 Department of Breast Surgery, 3 Second Department of Internal Medicine of Oncology, The 3rd Affiliated Hospital of Kunming Medical University, Kunming 650118, China ; 4 Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China ; 5 Institute of Molecular Medicine, National Cheng Kung University Medical College, Tainan, Taiwan, China
| | - Ceshi Chen
- 1 Department of Endocrine Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China ; 2 Department of Breast Surgery, 3 Second Department of Internal Medicine of Oncology, The 3rd Affiliated Hospital of Kunming Medical University, Kunming 650118, China ; 4 Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China ; 5 Institute of Molecular Medicine, National Cheng Kung University Medical College, Tainan, Taiwan, China
| | - Tianning Zou
- 1 Department of Endocrine Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China ; 2 Department of Breast Surgery, 3 Second Department of Internal Medicine of Oncology, The 3rd Affiliated Hospital of Kunming Medical University, Kunming 650118, China ; 4 Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China ; 5 Institute of Molecular Medicine, National Cheng Kung University Medical College, Tainan, Taiwan, China
| | - Rong Liu
- 1 Department of Endocrine Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China ; 2 Department of Breast Surgery, 3 Second Department of Internal Medicine of Oncology, The 3rd Affiliated Hospital of Kunming Medical University, Kunming 650118, China ; 4 Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China ; 5 Institute of Molecular Medicine, National Cheng Kung University Medical College, Tainan, Taiwan, China
| | - Jing Tan
- 1 Department of Endocrine Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China ; 2 Department of Breast Surgery, 3 Second Department of Internal Medicine of Oncology, The 3rd Affiliated Hospital of Kunming Medical University, Kunming 650118, China ; 4 Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China ; 5 Institute of Molecular Medicine, National Cheng Kung University Medical College, Tainan, Taiwan, China
| | - Guosheng Ren
- 1 Department of Endocrine Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China ; 2 Department of Breast Surgery, 3 Second Department of Internal Medicine of Oncology, The 3rd Affiliated Hospital of Kunming Medical University, Kunming 650118, China ; 4 Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China ; 5 Institute of Molecular Medicine, National Cheng Kung University Medical College, Tainan, Taiwan, China
| |
Collapse
|
18
|
Gasparini P, Fassan M, Cascione L, Guler G, Balci S, Irkkan C, Paisie C, Lovat F, Morrison C, Zhang J, Scarpa A, Croce CM, Shapiro CL, Huebner K. Androgen receptor status is a prognostic marker in non-basal triple negative breast cancers and determines novel therapeutic options. PLoS One 2014; 9:e88525. [PMID: 24505496 PMCID: PMC3914993 DOI: 10.1371/journal.pone.0088525] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 01/07/2014] [Indexed: 12/20/2022] Open
Abstract
Triple negative breast cancers are a heterogeneous group of tumors characterized by poor patient survival and lack of targeted therapeutics. Androgen receptor has been associated with triple negative breast cancer pathogenesis, but its role in the different subtypes has not been clearly defined. We examined androgen receptor protein expression by immunohistochemical analysis in 678 breast cancers, including 396 triple negative cancers. Fifty matched lymph node metastases were also examined. Association of expression status with clinical (race, survival) and pathological (basal, non-basal subtype, stage, grade) features was also evaluated. In 160 triple negative breast cancers, mRNA microarray expression profiling was performed, and differences according to androgen receptor status were analyzed. In triple negative cancers the percentage of androgen receptor positive cases was lower (24.8% vs 81.6% of non-triple negative cases), especially in African American women (16.7% vs 25.5% of cancers of white women). No significant difference in androgen receptor expression was observed in primary tumors vs matched metastatic lesions. Positive androgen receptor immunoreactivity was inversely correlated with tumor grade (p<0.01) and associated with better overall patient survival (p = 0.032) in the non-basal triple negative cancer group. In the microarray study, expression of three genes (HER4, TNFSF10, CDK6) showed significant deregulation in association with androgen receptor status; eg CDK6, a novel therapeutic target in triple negative cancers, showed significantly higher expression level in androgen receptor negative cases (p<0.01). These findings confirm the prognostic impact of androgen receptor expression in non-basal triple negative breast cancers, and suggest targeting of new androgen receptor-related molecular pathways in patients with these cancers.
Collapse
Affiliation(s)
- Pierluigi Gasparini
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Wexner Medical Center and Comprehensive Cancer Center, Columbus, Ohio, United States of America
| | - Matteo Fassan
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Wexner Medical Center and Comprehensive Cancer Center, Columbus, Ohio, United States of America
- ARC-NET Research Centre, University and Hospital Trust of Verona, Verona Italy
| | - Luciano Cascione
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Wexner Medical Center and Comprehensive Cancer Center, Columbus, Ohio, United States of America
| | - Gulnur Guler
- Department of Pathology, Hacettepe University, Ankara Turkey
| | - Serdar Balci
- Department of Pathology, Hacettepe University, Ankara Turkey
| | - Cigdem Irkkan
- Department of Pathology, Hacettepe University, Ankara Turkey
| | - Carolyn Paisie
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Wexner Medical Center and Comprehensive Cancer Center, Columbus, Ohio, United States of America
| | - Francesca Lovat
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Wexner Medical Center and Comprehensive Cancer Center, Columbus, Ohio, United States of America
| | - Carl Morrison
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, New York, United States of America
| | - Jianying Zhang
- Bioinformatics Shared Resource, Comprehensive Cancer Center, Ohio State University, Columbus, Ohio, United States of America
| | - Aldo Scarpa
- ARC-NET Research Centre, University and Hospital Trust of Verona, Verona Italy
| | - Carlo M. Croce
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Wexner Medical Center and Comprehensive Cancer Center, Columbus, Ohio, United States of America
| | - Charles L. Shapiro
- Division of Medical Oncology and the Breast Program, James Cancer Hospital and Ohio State University Comprehensive Cancer Center, Columbus, Ohio, United States of America
| | - Kay Huebner
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Wexner Medical Center and Comprehensive Cancer Center, Columbus, Ohio, United States of America
| |
Collapse
|
19
|
Ferguson BW, Gao X, Zelazowski MJ, Lee J, Jeter CR, Abba MC, Aldaz CM. The cancer gene WWOX behaves as an inhibitor of SMAD3 transcriptional activity via direct binding. BMC Cancer 2013; 13:593. [PMID: 24330518 PMCID: PMC3871008 DOI: 10.1186/1471-2407-13-593] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 12/06/2013] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND The WW domain containing protein WWOX has been postulated to behave as a tumor suppressor in breast and other cancers. Expression of this protein is lost in over 70% of ER negative tumors. This prompted us to investigate the phenotypic and gene expression effects of loss of WWOX expression in breast cells. METHODS Gene expression microarrays and standard in vitro assays were performed on stably silenced WWOX (shRNA) normal breast cells. Bioinformatic analyses were used to identify gene networks and transcriptional regulators affected by WWOX silencing. Co-immunoprecipitations and GST-pulldowns were used to demonstrate a direct interaction between WWOX and SMAD3. Reporter assays, ChIP, confocal microscopy and in silico analyses were employed to determine the effect of WWOX silencing on TGFβ-signaling. RESULTS WWOX silencing affected cell proliferation, motility, attachment and deregulated expression of genes involved in cell cycle, motility and DNA damage. Interestingly, we detected an enrichment of targets activated by the SMAD3 transcription factor, including significant upregulation of ANGPTL4, FST, PTHLH and SERPINE1 transcripts. Importantly, we demonstrate that the WWOX protein physically interacts with SMAD3 via WW domain 1. Furthermore, WWOX expression dramatically decreases SMAD3 occupancy at the ANGPTL4 and SERPINE1 promoters and significantly quenches activation of a TGFβ responsive reporter. Additionally, WWOX expression leads to redistribution of SMAD3 from the nuclear to the cytoplasmic compartment. Since the TGFβ target ANGPTL4 plays a key role in lung metastasis development, we performed a meta-analysis of ANGPTL4 expression relative to WWOX in microarray datasets from breast carcinomas. We observed a significant inverse correlation between WWOX and ANGPTL4. Furthermore, the WWOX(lo)/ANGPTL4(hi) cluster of breast tumors is enriched in triple-negative and basal-like sub-types. Tumors with this gene expression signature could represent candidates for anti-TGFβ targeted therapies. CONCLUSIONS We show for the first time that WWOX modulates SMAD3 signaling in breast cells via direct WW-domain mediated binding and potential cytoplasmic sequestration of SMAD3 protein. Since loss of WWOX expression increases with breast cancer progression and it behaves as an inhibitor of SMAD3 transcriptional activity these observations may help explain, at least in part, the paradoxical pro-tumorigenic effects of TGFβ signaling in advanced breast cancer.
Collapse
Affiliation(s)
- Brent W Ferguson
- Department of Molecular Carcinogenesis, Science Park, The University of Texas M.D. Anderson Cancer Center, Smithville, TX 78957, USA
| | - Xinsheng Gao
- Department of Molecular Carcinogenesis, Science Park, The University of Texas M.D. Anderson Cancer Center, Smithville, TX 78957, USA
| | - Maciej J Zelazowski
- Department of Molecular Carcinogenesis, Science Park, The University of Texas M.D. Anderson Cancer Center, Smithville, TX 78957, USA
| | - Jaeho Lee
- Department of Molecular Carcinogenesis, Science Park, The University of Texas M.D. Anderson Cancer Center, Smithville, TX 78957, USA
| | - Collene R Jeter
- Department of Molecular Carcinogenesis, Science Park, The University of Texas M.D. Anderson Cancer Center, Smithville, TX 78957, USA
| | - Martin C Abba
- CINIBA, Facultad de Medicina, Universidad Nacional de La Plata, La Plata, Argentina
| | - C Marcelo Aldaz
- Department of Molecular Carcinogenesis, Science Park, The University of Texas M.D. Anderson Cancer Center, Smithville, TX 78957, USA
| |
Collapse
|
20
|
Göthlin Eremo A, Wegman P, Stål O, Nordenskjöld B, Fornander T, Wingren S. Wwox expression may predict benefit from adjuvant tamoxifen in randomized breast cancer patients. Oncol Rep 2013; 29:1467-74. [PMID: 23381945 DOI: 10.3892/or.2013.2261] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 01/04/2013] [Indexed: 11/06/2022] Open
Abstract
Reduced or absent Wwox expression has recently been associated with tamoxifen resistance in breast cancer and has also been proposed as a candidate predictive marker for treatment. We aimed to investigate the correlation of Wwox expression with the outcome of tamoxifen treatment by examining tissues from 912 randomized breast cancer patients. Paraffin-embedded tissues from patient tumors were arranged on tissue microarray, and Wwox protein was stained using immunohistochemistry. After microscopic examination, the results were analyzed with Cox regression, Kaplan-Meier survival curves and the log-rank test. In the group of cases having a tumor absent for Wwox expression, there was no difference in recurrence-free survival between treated and untreated patients (P=0.81). For treated cases with a tumor expressing moderate or strong Wwox protein, recurrence-free survival was improved (P=0.001 and P=0.003, respectively). The test for interaction between Wwox and treatment response demonstrated a decreased risk of recurrence for treated patients with a moderate or strong Wwox expression (HR=0.31, 95% CI 0.10-0.98 and HR=0.28, 95% CI 0.08-0.97, respectively). Our results indicate that patients with high expression of Wwox may gain more benefit from treatment with tamoxifen.
Collapse
Affiliation(s)
- Anna Göthlin Eremo
- School of Health and Medical Sciences, Örebro University, SE-70182 Örebro, Sweden.
| | | | | | | | | | | |
Collapse
|
21
|
Płuciennik E, Nowakowska M, Wujcicka WI, Sitkiewicz A, Kazanowska B, Zielińska E, Bednarek AK. Genetic alterations of WWOX in Wilms' tumor are involved in its carcinogenesis. Oncol Rep 2012; 28:1417-22. [PMID: 22842668 DOI: 10.3892/or.2012.1940] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Accepted: 05/18/2012] [Indexed: 11/06/2022] Open
Abstract
Loss of heterozygosity (LOH) in 16q appears in ~20-30% cases of Wilms' tumor. Within this region, known as common fragile site FRA16D, the WWOX tumor suppressor gene is located. Abnormalities of WWOX gene expression levels were observed in many tumor types and were associated with worse prognosis. The purpose of this study was to investigate the role of the WWOX tumor suppressor gene in Wilms' tumor samples. We evaluated the correlation between expression of WWOX and genes involved in proliferation (Ki67), apoptosis (BCL2, BAX), signal transduction (ERBB4, ERBB2, EGFR), cell cycle (CCNE1, CCND1), cell adhesion (CDH1) and transcription (TP73) using real-time RT-PCR in 23 tumor samples. We also analyzed the potential causes of WWOX gene expression reduction i.e., promoter methylation status (MethylScreen method) and loss of heterozygosity (LOH) status. We revealed a positive correlation between WWOX expression and BCL2, BCL2/BAX ratio, EGFR, ERBB4 isoform JM-a, TP73 and negative correlation with both cyclins. Loss of heterozygosity of the WWOX gene was observed only at intron 8, however, it had no influence on the reduction of its expression levels. Contrary to LOH, methylation of the region covering the 3' end of the promoter and part of exon 1 was associated with statistically significant reduction of WWOX gene expression levels. In the present study we reveal that in Wilms' tumors the WWOX expression levels are positively associated with the process of apoptosis, signal transduction through the ErbB4 pathway and EGFR and negatively with the regulation of the cell cycle (by cyclin E1 and D1). Moreover, our analysis indicates that in this type of tumor the expression of the WWOX gene can be regulated by an epigenetic mechanism--its promoter methylation.
Collapse
Affiliation(s)
- Elżbieta Płuciennik
- Department of Molecular Cancerogenesis, Medical University of Lodz, 90-752 Lodz, Poland.
| | | | | | | | | | | | | |
Collapse
|
22
|
Aberrant expression of WWOX protein in epithelial ovarian cancer: a clinicopathologic and immunohistochemical study. Int J Gynecol Pathol 2012; 31:125-32. [PMID: 22317867 DOI: 10.1097/pgp.0b013e3182297fd2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Epithelial ovarian cancer is the most frequent cause of death from gynecologic cancer. The WW domain-containing oxidoreductase (WWOX) gene is located at 16q23.3-24.1, a region that spans the second most common human fragile site, FRA16D. Abnormalities affecting WWOX at the genomic and/or expression level(s) have been reported in numerous neoplasias and cancer-derived cell lines. The goal of the study was to evaluate WWOX protein expression in epithelial ovarian carcinoma tissues to determine whether they correlated with clincopathologic parameters. We performed WWOX expression analyses by means of immunohistochemistry on 112 epithelial ovarian carcinoma tissues, and ovarian carcinoma-derived SKOV3, 3AO cells. The basic significant level was fixed at P<0.05. Loss of WWOX expression was observed in 32 (28.6%) of 112 ovarian carcinoma samples and was positively correlated with negative estrogen receptor (ER) (P<0.001) and negative progesterone receptor (PR) (P=0.001). A statistically significant correlation was observed between the lack of WWOX expression and the advanced International Federation of Gynecology and Obstetrics (FIGO) stages (P=0.02). Furthermore, negative WWOX staining was significantly correlated with lymph node metastasis (P=0.013), whereas no significant differences were found between WWOX and HER-2/neu staining (P=0.79). WWOX protein expression was moderately detectable in SKOV3 cells but not in 3AO cells. Our results indicate that loss of WWOX expression in epithelial ovarian carcinomas correlates with negative ER, negative PR, advanced FIGO stages, and lymph node metastases.
Collapse
|
23
|
Tian Y, Zhang J, Yan S, Qiu L, Li Z. FATS expression is associated with cisplatin sensitivity in non small cell lung cancer. Lung Cancer 2012; 76:416-22. [DOI: 10.1016/j.lungcan.2011.11.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Revised: 09/26/2011] [Accepted: 11/07/2011] [Indexed: 01/21/2023]
|
24
|
Ferguson BW, Gao X, Kil H, Lee J, Benavides F, Abba MC, Aldaz CM. Conditional Wwox deletion in mouse mammary gland by means of two Cre recombinase approaches. PLoS One 2012; 7:e36618. [PMID: 22574198 PMCID: PMC3344920 DOI: 10.1371/journal.pone.0036618] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Accepted: 04/03/2012] [Indexed: 11/23/2022] Open
Abstract
Loss of WWOX expression has been reported in many different cancers including breast cancer. Elucidating the function of this gene in adult tissues has not been possible with full Wwox knockout models. Here we characterize the first conditional models of Wwox ablation in mouse mammary epithelium utilizing two transgenic lines expressing Cre recombinase, keratin 5-Cre (BK5-Cre) and MMTV-Cre. In the BK5-Cre model we observed very efficient Wwox ablation in KO mammary glands. However, BK5-Cre Wwox KO animals die prematurely for unknown reasons. In the MMTV-Cre model we observed significant ablation of Wwox in mammary epithelium with no effect on survival. In both of these models we found that Wwox deletion resulted in impaired mammary branching morphogenesis. We demonstrate that loss of Wwox is not carcinogenic in our KO models. Furthermore, no evidence of increase proliferation or development of premalignant lesions was observed. In none of the models did loss of a single Wwox allele (i.e. haploinsufficiency) have any observable phenotypic effect in mammary gland. To better understand the function of Wwox in the mammary gland, transcriptome profiling was performed. We observed that Wwox ablation results in the deregulation of genes involved in various cellular processes. We found that expression of the non-canonical Wnt ligand, Wnt5a, was significantly upregulated in Wwox KO mammary epithelium. Interestingly, we also determined that components of the Jak/Stat3 signaling pathway were upregulated in KO mice and this correlated with a very robust increase in phospho-Stat3 signaling, which warrants further testing. Even though the loss of Wwox expression in breast and other cancers is very well documented, our findings suggest that Wwox does not act as a classical tumor suppressor as previously thought.
Collapse
Affiliation(s)
- Brent W. Ferguson
- Department of Molecular Carcinogenesis, The University of Texas M.D. Anderson Cancer Center, Smithville, Texas, United States of America
| | - Xinsheng Gao
- Department of Molecular Carcinogenesis, The University of Texas M.D. Anderson Cancer Center, Smithville, Texas, United States of America
| | - Hyunsuk Kil
- Department of Molecular Carcinogenesis, The University of Texas M.D. Anderson Cancer Center, Smithville, Texas, United States of America
| | - Jaeho Lee
- Department of Molecular Carcinogenesis, The University of Texas M.D. Anderson Cancer Center, Smithville, Texas, United States of America
| | - Fernando Benavides
- Department of Molecular Carcinogenesis, The University of Texas M.D. Anderson Cancer Center, Smithville, Texas, United States of America
| | - Martin C. Abba
- CINIBA, Facultad de Medicina, Universidad Nacional de La Plata, La Plata, Argentina
| | - C. Marcelo Aldaz
- Department of Molecular Carcinogenesis, The University of Texas M.D. Anderson Cancer Center, Smithville, Texas, United States of America
- * E-mail:
| |
Collapse
|
25
|
Scott DJ, Devonshire AS, Adeleye YA, Schutte ME, Rodrigues MR, Wilkes TM, Sacco MG, Gribaldo L, Fabbri M, Coecke S, Whelan M, Skinner N, Bennett A, White A, Foy CA. Inter- and intra-laboratory study to determine the reproducibility of toxicogenomics datasets. Toxicology 2011; 290:50-8. [PMID: 21871943 DOI: 10.1016/j.tox.2011.08.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2011] [Revised: 08/09/2011] [Accepted: 08/12/2011] [Indexed: 10/17/2022]
Abstract
The application of toxicogenomics as a predictive tool for chemical risk assessment has been under evaluation by the toxicology community for more than a decade. However, it predominately remains a tool for investigative research rather than for regulatory risk assessment. In this study, we assessed whether the current generation of microarray technology in combination with an in vitro experimental design was capable of generating robust, reproducible data of sufficient quality to show promise as a tool for regulatory risk assessment. To this end, we designed a prospective collaborative study to determine the level of inter- and intra-laboratory reproducibility between three independent laboratories. All test centres (TCs) adopted the same protocols for all aspects of the toxicogenomic experiment including cell culture, chemical exposure, RNA extraction, microarray data generation and analysis. As a case study, the genotoxic carcinogen benzo[a]pyrene (B[a]P) and the human hepatoma cell line HepG2 were used to generate three comparable toxicogenomic data sets. High levels of technical reproducibility were demonstrated using a widely employed gene expression microarray platform. While differences at the global transcriptome level were observed between the TCs, a common subset of B[a]P responsive genes (n=400 gene probes) was identified at all TCs which included many genes previously reported in the literature as B[a]P responsive. These data show promise that the current generation of microarray technology, in combination with a standard in vitro experimental design, can produce robust data that can be generated reproducibly in independent laboratories. Future work will need to determine whether such reproducible in vitro model(s) can be predictive for a range of toxic chemicals with different mechanisms of action and thus be considered as part of future testing regimes for regulatory risk assessment.
Collapse
Affiliation(s)
- D J Scott
- LGC, Queens Rd, Teddington, TW11 0LY, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Shibata H, Miuma S, Saldivar JC, Huebner K. Response of subtype-specific human breast cancer-derived cells to poly(ADP-ribose) polymerase and checkpoint kinase 1 inhibition. Cancer Sci 2011; 102:1882-8. [PMID: 21707865 DOI: 10.1111/j.1349-7006.2011.02016.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
When DNA damage is detected, checkpoint signal networks are activated to stop the cell cycle, and DNA repair processes begin. Inhibitory compounds targeting components of DNA damage response pathways have been identified and are being used in clinical trials, in combination with chemotherapeutic agents, to enhance cancer therapy. Inhibitors of checkpoint kinases, Chk1 and Chk2, have been shown to sensitize tumor cells to DNA damaging agents, and treatment of BRCA1/2-deficient tumor cells, as well as triple negative breast cancers, with poly(ADP-ribose) polymerase (PARP) inhibitors has shown promise. But systematic studies to determine which tumor subtypes are likely to respond to these specific inhibitors have not been reported. The current study was designed to test sensitivity of specific breast cancer subtype-derived cells to two classes of these new inhibitory drugs, PARP and Chk1 inhibitors. Luminal, HER2 overexpressing, and triple negative breast cancer-derived cells were tested for sensitivity to killing by PARP inhibitors, ABT-888 and BSI-201, and Chk1 inhibitor, PF-00477736, alone or in combination with gemcitabine or carboplatin. Each of the triple negative breast cancer cell lines showed strong sensitivity to the Chk1 inhibitor, but only the BRCA1-deficient breast cancer cell lines showed sensitivity to the PARP inhibitors, suggesting that in vitro testing of cancer cell lines of specific subtypes, with panels of the different PARP and Chk1 inhibitors, will contribute to stratification of patients for clinical trials using these classes of inhibitors.
Collapse
Affiliation(s)
- Hidetaka Shibata
- Molecular Virology, Immunology and Medical Genetics, and Comprehensive Cancer Center, The Ohio State University Medical Center, Columbus, Ohio, USA
| | | | | | | |
Collapse
|
27
|
Abstract
Breast cancer is the leading cause of cancer-related death in women worldwide. Expression of the WWOX tumor suppressor is absent or reduced in a large proportion of breast tumors suggesting that loss of WWOX may contribute to breast tumorigenesis. Wwox-deficient mice die by 3-4 weeks of age precluding adult tumor analysis. To evaluate the effect of WWOX-altered expression on mammary tumor formation, the Wwox-heterozygous allele was back crossed onto the C3H mammary tumor-susceptible genetic background (Wwox(C3H)+/-) and incidence of mammary tumor formation was evaluated. Although 50% of the female Wwox(C3H)+/- mice developed mammary carcinomas, only 7% of Wwox(C3H)+/+ mice did. Intriguingly, mammary tumors in Wwox(C3H)+/- mice frequently lost WWOX protein expression suggesting a genetic predisposition toward mammary tumorigenesis. Immunohistochemical staining of hormone receptors revealed loss of estrogen receptor-α (ER) and progesterone receptor in the majority of these tumors. In vitro, depletion of WWOX in MCF7 ER-positive cells led to reduced ER expression and reduced sensitivity to tamoxifen and estrogen treatment and was associated with enhanced survival and anchorage-independent growth. Finally, cDNA array analyses of murine normal mammary epithelial cells and mammary tumors identified 163 significantly downreguated and 129 upregulated genes in the tumors. The majority of differentially expressed genes were part of pathways involved in cellular movement, cell-to-cell signaling and interaction, cellular development, cellular growth and proliferation and cell death. These changes in gene expression of mouse mammary tumors in Wwox(C3H)+/- mice resemble, at least in part, human breast cancer development. Our findings demonstrate the critical role that the WWOX tumor suppressor gene has in preventing tumorigenesis in breast cancer.
Collapse
|
28
|
Żelazowski MJ, Płuciennik E, Pasz-Walczak G, Potemski P, Kordek R, Bednarek AK. WWOX expression in colorectal cancer--a real-time quantitative RT-PCR study. Tumour Biol 2011; 32:551-60. [PMID: 21347750 PMCID: PMC3093543 DOI: 10.1007/s13277-010-0150-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Accepted: 12/14/2010] [Indexed: 01/16/2023] Open
Abstract
The WWOX gene is a tumour suppressor gene affected in various types of malignancies. Numerous studies showed either loss or reduction of the WWOX expression in variety of tumours, including breast, ovary, liver, stomach and pancreas. Recent study demonstrated that breast cancer patients exhibiting higher WWOX expression showed significantly longer disease-free survival in contrast to the group with lower relative WWOX level. This work was undertaken to show whether similar phenomena take place in colon tumours and cell lines. To assess the correlation of WWOX gene expression with prognosis and cancer recurrence in 99 colorectal cancer patients, we performed qRT-PCR analysis. We also performed analysis of WWOX promoter methylation status using MethylScreen method and analysis of loss of heterozygosity (LOH) status at two WWOX-related loci, previously shown to be frequently deleted in various types of tumours. A significantly better disease-free survival was observed among patients with tumours exhibiting high level of WWOX (hazard ratio = 0.39; p = 0.0452; Mantel-Cox log-rank test), but in multivariate analysis it was not an independent prognostic factor. We also found that although in colorectal cancer WWOX expression varies among patients and correlates with DFS, the exact mode of decrease in this type of tumour was not found. We failed to find the evidence of LOH in WWOX region, or hypermethylation in promoter regions of this gene. Although we provide the evidence for tumour-suppressive role of WWOX gene expression in colon, we were unable to identify the molecular mechanism responsible for this.
Collapse
Affiliation(s)
- Maciej Jakub Żelazowski
- Department of Molecular Carcinogenesis, Medical University of Łódź, Zeligowskiego Str 7/9, 90-752 Łódź, Poland.
| | | | | | | | | | | |
Collapse
|
29
|
Woodfield GW, Chen Y, Bair TB, Domann FE, Weigel RJ. Identification of primary gene targets of TFAP2C in hormone responsive breast carcinoma cells. Genes Chromosomes Cancer 2010; 49:948-62. [PMID: 20629094 DOI: 10.1002/gcc.20807] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The TFAP2C transcription factor is involved in mammary development, differentiation, and oncogenesis. Previous studies established a role for TFAP2C in the regulation of ESR1 (ERalpha) and ERBB2 (Her2) in breast carcinomas. However, the role of TFAP2C in different breast cancer phenotypes has not been examined in detail. To develop a more complete characterization of TFAP2C target genes, ChIP-seq with anti-TFAP2C antibody and expression arrays with TFAP2C knock down were analyzed in MCF-7 breast carcinoma cells. Genomic sequences common to the ChIP-seq data set defined the consensus sequence for TFAP2C chromatin binding as the nine base sequence SCCTSRGGS (S = G/C, r = A/G), which closely matches the previously defined optimal in vitro binding site. Comparing expression arrays before and after knock down of TFAP2C with ChIP-seq data demonstrated a conservative estimate that 8% of genes altered by TFAP2C expression are primary target genes and includes genes that are both induced and repressed by TFAP2C. A set of 447 primary target genes of TFAP2C was identified, which included ESR1 (ERalpha), FREM2, RET, FOXA1, WWOX, GREB1, MYC, and members of the retinoic acid response pathway. The identification of ESR1, WWOX, GREB1, and FOXA1 as primary targets confirmed the role of TFAP2C in hormone response. TFAP2C plays a critical role in gene regulation in hormone responsive breast cancer and its target genes are different than for the Her2 breast cancer phenotype.
Collapse
Affiliation(s)
- George W Woodfield
- Department of Surgery, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | | | | | | | | |
Collapse
|
30
|
Guler G, Himmetoglu C, Jimenez RE, Geyer SM, Wang WP, Costinean S, Pilarski RT, Morrison C, Suren D, Liu J, Chen J, Kamal J, Shapiro CL, Huebner K. Aberrant expression of DNA damage response proteins is associated with breast cancer subtype and clinical features. Breast Cancer Res Treat 2010; 129:421-32. [PMID: 21069451 DOI: 10.1007/s10549-010-1248-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Accepted: 10/26/2010] [Indexed: 12/18/2022]
Abstract
Landmark studies of the status of DNA damage checkpoints and associated repair functions in preneoplastic and neoplastic cells has focused attention on importance of these pathways in cancer development, and inhibitors of repair pathways are in clinical trials for treatment of triple negative breast cancer. Cancer heterogeneity suggests that specific cancer subtypes will have distinct mechanisms of DNA damage survival, dependent on biological context. In this study, status of DNA damage response (DDR)-associated proteins was examined in breast cancer subtypes in association with clinical features; 479 breast cancers were examined for expression of DDR proteins γH2AX, BRCA1, pChk2, and p53, DNA damage-sensitive tumor suppressors Fhit and Wwox, and Wwox-interacting proteins Ap2α, Ap2γ, ErbB4, and correlations among proteins, tumor subtypes, and clinical features were assessed. In a multivariable model, triple negative cancers showed significantly reduced Fhit and Wwox, increased p53 and Ap2γ protein expression, and were significantly more likely than other subtype tumors to exhibit aberrant expression of two or more DDR-associated proteins. Disease-free survival was associated with subtype, Fhit and membrane ErbB4 expression level and aberrant expression of multiple DDR-associated proteins. These results suggest that definition of specific DNA repair and checkpoint defects in subgroups of triple negative cancer might identify new treatment targets. Expression of Wwox and its interactor, ErbB4, was highly significantly reduced in metastatic tissues vs. matched primary tissues, suggesting that Wwox signal pathway loss contributes to lymph node metastasis, perhaps by allowing survival of tumor cells that have detached from basement membranes, as proposed for the role of Wwox in ovarian cancer spread.
Collapse
Affiliation(s)
- Gulnur Guler
- Department of Pathology, Hacettepe University, Ankara, Turkey
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Salah Z, Aqeilan R, Huebner K. WWOX gene and gene product: tumor suppression through specific protein interactions. Future Oncol 2010; 6:249-59. [PMID: 20146584 DOI: 10.2217/fon.09.152] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The WWOX gene, an archetypal fragile gene, encompasses a chromosomal fragile site at 16q23.2, and encodes the approximately 46-kDa Wwox protein, with WW domains that interact with a growing list of interesting proteins. If the function of a protein is defined by the company it keeps, then Wwox is involved in numerous important signal pathways for bone and germ-cell development, cellular and animal growth and death, transcriptional control and suppression of cancer development. Because alterations to genes at fragile sites are exquisitely sensitive to replication stress-induced DNA damage, there has been an ongoing scientific discussion questioning whether such gene expression alterations provide a selective advantage for clonal expansion of neoplastic cells, and a parallel discussion on why important genes would be present at sites that are susceptible to inactivation. We offer some answers through a description of known WWOX functions.
Collapse
Affiliation(s)
- Zaidoun Salah
- The Lautenberg Center for Immunology, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Pharmacy Building, Jerusalem 91120, Israel.
| | | | | |
Collapse
|
32
|
Perez EA, Moreno-Aspitia A, Aubrey Thompson E, Andorfer CA. Adjuvant therapy of triple negative breast cancer. Breast Cancer Res Treat 2010; 120:285-91. [PMID: 20094772 PMCID: PMC3918886 DOI: 10.1007/s10549-010-0736-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2009] [Accepted: 01/06/2010] [Indexed: 12/28/2022]
Abstract
Patients with the triple negative subtype of breast cancer have an overall poor outcome, with earlier relapses, distinct patterns of metastases, and lack of specific targets for treatment selection. Classification of these tumors has begun to be modified by inclusion of immunohistochemistry for various markers, and gene profiling. Further characterization of this subtype of breast cancer may aid in the identification of new targeted therapies. Anthracyclines and taxanes remain the standard of care in the adjuvant setting. However, novel anti-angiogenesis, anti-tubulin, and DNA repair agents are already under evaluation in (neo) adjuvant trials. Molecular characterization is being included in trials to identify optimal adjuvant strategies. The aim of this manuscript is to review data concerning the molecular characterization of triple negative breast cancers as well as the clinical outcomes of treating patients with existing adjuvant treatments, and to highlight newer adjuvant research strategies in development.
Collapse
Affiliation(s)
- Edith A Perez
- Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA.
| | | | | | | |
Collapse
|
33
|
Immunohistochemical expression of epidermal growth factor receptor in breast cancer. Breast Cancer 2010; 18:37-41. [DOI: 10.1007/s12282-010-0200-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2009] [Accepted: 01/27/2010] [Indexed: 10/19/2022]
|
34
|
Hassan MI, Naiyer A, Ahmad F. Fragile histidine triad protein: structure, function, and its association with tumorogenesis. J Cancer Res Clin Oncol 2010; 136:333-50. [PMID: 20033706 DOI: 10.1007/s00432-009-0751-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2009] [Accepted: 12/09/2009] [Indexed: 01/31/2023]
Abstract
BACKGROUND The human fragile histidine triad (FHIT) gene is a putative tumor suppressor gene, which is located at chromosome region 3p14.2. It was suggested that the loss of heterozygosity (LOH), homozygous deletions, and abnormal expression of the FHIT gene were involved in several types of human malignancies. MATERIALS AND METHODS To determine the role of FHIT in various cancers, we have performed structural and functional analysis of FHIT in detail. RESULTS AND DISCUSSION The protein FHIT catalyzes the Mg(2+) dependent hydrolysis of P1-5 cent-O-adenosine-P3-5 cent-O-adenosine triphosphate, Ap3A, to AMP, and ADP. The reaction is thought to follow a two-step mechanism. Histidine triad proteins, named for a motif related to the sequence H-cent-H-cent-H-cent-cent- (cent, a hydrophobic amino acid), belong to superfamily of nucleotide hydrolases and transferases. This enzyme acts on the R-phosphate of ribonucleotides, and contain a approximately 30-kDa domain that is typically a homodimer of approximately 15 kDa polypeptides with catalytic site. CONCLUSION Here we have gathered information is known about biological activities of FHIT, the structural and biochemical bases for their functions. Our approach may provide a comparative framework for further investigation of FHIT.
Collapse
Affiliation(s)
- Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | | | | |
Collapse
|
35
|
Study of FHIT and WWOX expression in mucoepidermoid carcinoma and adenoid cystic carcinoma of salivary gland. Oral Oncol 2010; 46:195-9. [DOI: 10.1016/j.oraloncology.2009.12.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2009] [Revised: 12/07/2009] [Accepted: 12/08/2009] [Indexed: 11/21/2022]
|
36
|
Okumura H, Ishii H, Pichiorri F, Croce CM, Mori M, Huebner K. Fragile gene product, Fhit, in oxidative and replicative stress responses. Cancer Sci 2009; 100:1145-50. [PMID: 19486340 PMCID: PMC11159339 DOI: 10.1111/j.1349-7006.2009.01168.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2009] [Revised: 03/12/2009] [Accepted: 03/15/2009] [Indexed: 10/21/2022] Open
Abstract
Though the fragile histidine triad gene product, Fhit, was discovered and characterized as a tumor suppressor 13 years ago, its sequence, structure, and cellular location did not provide clues to aid discovery of its mechanisms of suppression. Recently, using chemical cross-linkers and immunoprecipitation, a Fhit protein complex was identified that includes Hsp60 and Hsp10 which may mediate Fhit stability and mitochondrial localization, where Fhit binds and stabilizes ferredoxin reductase (Fdxr); when Fdxr is overexpressed, it can lead to production of reactive oxygen species (ROS) that induce apoptosis. Cancer cells expressing endogenous or exogenous Fhit, when exposed to H(2)O(2), an oxidative stress, produce higher levels of apoptosis-inducing ROS than matched, Fhit-negative cells; the Fhit-negative cancer cells survive, carrying DNA damage. In addition to this mitochondrial function, Fhit-overexpression in cancer cells exposed to replicative stress-inducing agents leads to enhanced caspase 3 activation and apoptosis, due to defective Chk1 activation. Thus, damage to the fragile FHIT locus leads to reduced expression of Fhit protein, and makes a two-pronged contribution to development of preneoplastic clonal expansion: (1) absence or reduction of Fhit leads to reduced expression of Fdxr and reduced ROS-induced apoptosis; (2) cells that escape ROS- or replicative stress-induced apoptosis can carry misrepaired DNA damage. The aberrant DNA damage response checkpoint in Fhit-deficient preneoplasias and cancers may make these lesions targets for inhibitors of proteins such as Parp1 and Chk1 with important roles in checkpoint responses, as observed for BRCA1-deficient cancer cells that also exhibit DNA damage repair deficiencies.
Collapse
Affiliation(s)
- Hiroshi Okumura
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | | | | | | | | | | |
Collapse
|