1
|
Nierengarten MB. First person profile: Ruth M. O'Regan, MD: Dr O'Regan promotes work-life balance, collaboration, equal access to education, and care for all. Cancer 2025; 131:e35809. [PMID: 40233160 DOI: 10.1002/cncr.35809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
This news section offers Cancer readers timely information on events, public policy analysis, topical issues, and personalities. This issue includes a profile of Dr Ruth M. O’Regan, who promotes work‐life balance, collaboration, equal access to education, and care for all. Also, adding two cycles of the immunotherapy blinatumomab to chemotherapy for children with newly diagnosed standard‐risk B‐cell ALL improved outcomes significantly, and high response rates were seen with novel CAR T‐cell therapy for adults with advanced B‐cell ALL.
Collapse
|
2
|
Mustafa M, Abbas K, Alam M, Ahmad W, Moinuddin, Usmani N, Siddiqui SA, Habib S. Molecular pathways and therapeutic targets linked to triple-negative breast cancer (TNBC). Mol Cell Biochem 2024; 479:895-913. [PMID: 37247161 DOI: 10.1007/s11010-023-04772-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 05/18/2023] [Indexed: 05/30/2023]
Abstract
Cancer is a group of diseases characterized by uncontrolled cellular growth, abnormal morphology, and altered proliferation. Cancerous cells lose their ability to act as anchors, allowing them to spread throughout the body and infiltrate nearby cells, tissues, and organs. If these cells are not identified and treated promptly, they will likely spread. Around 70% of female breast cancers are caused by a mutation in the BRCA gene, specifically BRCA1. The absence of progesterone, oestrogen and HER2 receptors (human epidermal growth factor) distinguishes the TNBC subtype of breast cancer. There were approximately 6,85,000 deaths worldwide and 2.3 million new breast cancer cases in women in 2020. Breast cancer is the most common cancer globally, affecting 7.8 million people at the end of 2020. Compared to other cancer types, breast cancer causes more women to lose disability-adjusted life years (DALYs). Worldwide, women can develop breast cancer at any age after puberty, but rates increase with age. The maintenance of mammary stem cell stemness is disrupted in TNBC, governed by signalling cascades controlling healthy mammary gland growth and development. Interpreting these essential cascades may facilitate an in-depth understanding of TNBC cancer and the search for an appropriate therapeutic target. Its treatment remains challenging because it lacks specific receptors, which renders hormone therapy and medications ineffective. In addition to radiotherapy, numerous recognized chemotherapeutic medicines are available as inhibitors of signalling pathways, while others are currently undergoing clinical trials. This article summarizes the vital druggable targets, therapeutic approaches, and strategies associated with TNBC.
Collapse
Affiliation(s)
- Mohd Mustafa
- Department of Biochemistry, J.N. Medical College, Aligarh Muslim University, Aligarh, 202002, India
| | - Kashif Abbas
- Department of Zoology, Aligarh Muslim University, Aligarh, India
| | - Mudassir Alam
- Department of Zoology, Aligarh Muslim University, Aligarh, India
| | - Waleem Ahmad
- Department of Medicine, J.N. Medical College, Aligarh Muslim University, Aligarh, India
| | - Moinuddin
- Department of Biochemistry, J.N. Medical College, Aligarh Muslim University, Aligarh, 202002, India
| | - Nazura Usmani
- Department of Zoology, Aligarh Muslim University, Aligarh, India
| | - Shahid Ali Siddiqui
- Department of Radiotherapy, J.N. Medical College, Aligarh Muslim University, Aligarh, India
| | - Safia Habib
- Department of Biochemistry, J.N. Medical College, Aligarh Muslim University, Aligarh, 202002, India.
| |
Collapse
|
3
|
Merckens A, Sieler M, Keil S, Dittmar T. Altered Phenotypes of Breast Epithelial × Breast Cancer Hybrids after ZEB1 Knock-Out. Int J Mol Sci 2023; 24:17310. [PMID: 38139138 PMCID: PMC10744253 DOI: 10.3390/ijms242417310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
ZEB1 plays a pivotal role in epithelial-to-mesenchymal transition (EMT), (cancer) cell stemness and cancer therapy resistance. The M13HS tumor hybrids, which were derived from spontaneous fusion events between the M13SV1-EGFP-Neo breast epithelial cells and HS578T-Hyg breast cancer cells, express ZEB1 and exhibit prospective cancer stem cell properties. To explore a possible correlation between the ZEB1 and stemness/ EMT-related properties in M13HS tumor hybrids, ZEB1 was knocked-out by CRISPR/Cas9. Colony formation, mammosphere formation, cell migration, invasion assays, flow cytometry and Western blot analyses were performed for the characterization of ZEB1 knock-out cells. The ZEB1 knock-out in M13HS tumor cells was not correlated with the down-regulation of the EMT-related markers N-CADHERIN (CDH2) and VIMENTIN and up-regulation of miR-200c-3p. Nonetheless, both the colony formation and mammosphere formation capacities of the M13HS ZEB1 knock-out cells were markedly reduced. Interestingly, the M13HS-2 ZEB1-KO cells harbored a markedly higher fraction of ALDH1-positive cells. The Transwell/ Boyden chamber migration assay data indicated a reduced migratory activity of the M13HS ZEB1-knock-out tumor hybrids, whereas in scratch/ wound-healing assays only the M13SH-8 ZEB1-knock-out cells possessed a reduced locomotory activity. Similarly, only the M13HS-8 ZEB1-knock-out tumor hybrids showed a reduced invasion capacity. Although the ZEB1 knock-out resulted in only moderate phenotypic changes, our data support the role of ZEB1 in EMT and stemness.
Collapse
Affiliation(s)
| | | | | | - Thomas Dittmar
- Institute of Immunology, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, Stockumer Str. 10, 58448 Witten, Germany; (A.M.); (M.S.); (S.K.)
| |
Collapse
|
4
|
Rajarajan S, Snijesh VP, Anupama CE, Nair MG, Mavatkar AD, Naidu CM, Patil S, Nimbalkar VP, Alexander A, Pillai M, Jolly MK, Sabarinathan R, Ramesh RS, Bs S, Prabhu JS. An androgen receptor regulated gene score is associated with epithelial to mesenchymal transition features in triple negative breast cancers. Transl Oncol 2023; 37:101761. [PMID: 37603927 PMCID: PMC10465938 DOI: 10.1016/j.tranon.2023.101761] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 07/13/2023] [Accepted: 08/07/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUND Androgen receptor (AR) is considered a marker of better prognosis in hormone receptor positive breast cancers (BC), however, its role in triple negative breast cancer (TNBC) is controversial. This may be attributed to intrinsic molecular differences or scoring methods for AR positivity. We derived AR regulated gene score and examined its utility in BC subtypes. METHODS AR regulated genes were derived by applying a bioinformatic pipeline on publicly available microarray data sets of AR+ BC cell lines and gene score was calculated as average expression of six AR regulated genes. Tumors were divided into AR high and low based on gene score and associations with clinical parameters, circulating androgens, survival and epithelial to mesenchymal transition (EMT) markers were examined, further evaluated in invitro models and public datasets. RESULTS 53% (133/249) tumors were classified as AR gene score high and were associated with significantly better clinical parameters, disease-free survival (86.13 vs 72.69 months, log rank p = 0.032) when compared to AR low tumors. 36% of TNBC (N = 66) were AR gene score high with higher expression of EMT markers (p = 0.024) and had high intratumoral levels of 5α-reductase, enzyme involved in intracrine androgen metabolism. In MDA-MB-453 treated with dihydrotestosterone, SLUG expression increased, E-cadherin decreased with increase in migration and these changes were reversed with bicalutamide. Similar results were obtained in public datasets. CONCLUSION Deciphering the role of AR in BC is difficult based on AR protein levels alone. Our results support the context dependent function of AR in driving better prognosis in ER positive tumors and EMT features in TNBC tumors.
Collapse
Affiliation(s)
- Savitha Rajarajan
- Division of Molecular Medicine, St. John's Research Institute, St. John's Medical College, Bengaluru, India; Centre for Doctoral Studies, Manipal Academy of Higher Education (MAHE), Manipal, India
| | - V P Snijesh
- Division of Molecular Medicine, St. John's Research Institute, St. John's Medical College, Bengaluru, India; Centre for Doctoral Studies, Manipal Academy of Higher Education (MAHE), Manipal, India
| | - C E Anupama
- Division of Molecular Medicine, St. John's Research Institute, St. John's Medical College, Bengaluru, India
| | - Madhumathy G Nair
- Division of Molecular Medicine, St. John's Research Institute, St. John's Medical College, Bengaluru, India
| | - Apoorva D Mavatkar
- Division of Molecular Medicine, St. John's Research Institute, St. John's Medical College, Bengaluru, India
| | - Chandrakala M Naidu
- Division of Molecular Medicine, St. John's Research Institute, St. John's Medical College, Bengaluru, India
| | - Sharada Patil
- Division of Molecular Medicine, St. John's Research Institute, St. John's Medical College, Bengaluru, India
| | - Vidya P Nimbalkar
- Division of Molecular Medicine, St. John's Research Institute, St. John's Medical College, Bengaluru, India
| | - Annie Alexander
- Division of Molecular Medicine, St. John's Research Institute, St. John's Medical College, Bengaluru, India
| | - Maalavika Pillai
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru, India
| | - Mohit Kumar Jolly
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru, India
| | | | - Rakesh S Ramesh
- Department of Surgical Oncology, St. John's Medical College, Bengaluru, India
| | - Srinath Bs
- Department of Surgery, Sri Shankara Cancer Hospital and Research Centre, Bangalore, India
| | - Jyothi S Prabhu
- Division of Molecular Medicine, St. John's Research Institute, St. John's Medical College, Bengaluru, India.
| |
Collapse
|
5
|
Crespo B, Caceres S, Silvan G, Illera MJ, Illera JC. The inhibition of steroid hormones determines the fate of IPC-366 tumor cells, highlighting the crucial role of androgen production in tumor processes. Res Vet Sci 2023; 161:1-14. [PMID: 37290206 DOI: 10.1016/j.rvsc.2023.05.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 05/01/2023] [Accepted: 05/26/2023] [Indexed: 06/10/2023]
Abstract
Inflammatory mammary cancer (IMC) is a disease that affects female dogs. It is characterized by poor treatment options and no efficient targets. However, anti-androgenic and anti-estrogenic therapies could be effective because IMC has a great endocrine influence, affecting tumor progression. IPC-366 is a triple negative IMC cell line that has been postulated as a useful model to study this disease. Therefore, the aim of this study was to inhibit steroid hormones production at different points of the steroid pathway in order to determine its effect in cell viability and migration in vitro and tumor growth in vivo. For this purpose, Dutasteride (anti-5αReductase), Anastrozole (anti-aromatase) and ASP9521 (anti-17βHSD) and their combinations have been used. Results revealed that this cell line is positive to estrogen receptor β (ERβ) and androgen receptor (AR) and endocrine therapies reduce cell viability. Our results enforced the hypothesis that estrogens promote cell viability and migration in vitro due to the function of E1SO4 as an estrogen reservoir for E2 production that promotes the IMC cells proliferation. Also, an increase in androgen secretion was associated with a reduction in cell viability. Finally, in vivo assays showed large tumor reduction. Hormone assays determined that high estrogen levels and the reduction of androgen levels promote tumor growth in Balb/SCID IMC mice. In conclusion, estrogen levels reduction may be associated with a good prognosis. Also, activation of AR by increasing androgen production could result in effective therapy for IMC because their anti-proliferative effect.
Collapse
Affiliation(s)
- Belen Crespo
- Department Animal Physiology, Veterinary Medicine School, Complutense University of Madrid (UCM), 28040 Madrid, Spain.
| | - Sara Caceres
- Department Animal Physiology, Veterinary Medicine School, Complutense University of Madrid (UCM), 28040 Madrid, Spain.
| | - Gema Silvan
- Department Animal Physiology, Veterinary Medicine School, Complutense University of Madrid (UCM), 28040 Madrid, Spain.
| | - Maria Jose Illera
- Department Animal Physiology, Veterinary Medicine School, Complutense University of Madrid (UCM), 28040 Madrid, Spain.
| | - J C Illera
- Department Animal Physiology, Veterinary Medicine School, Complutense University of Madrid (UCM), 28040 Madrid, Spain.
| |
Collapse
|
6
|
Kang Y. Landscape of NcRNAs involved in drug resistance of breast cancer. Clin Transl Oncol 2023; 25:1869-1892. [PMID: 37067729 PMCID: PMC10250522 DOI: 10.1007/s12094-023-03189-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 07/02/2022] [Indexed: 04/18/2023]
Abstract
Breast cancer (BC) leads to the most amounts of deaths among women. Chemo-, endocrine-, and targeted therapies are the mainstay drug treatments for BC in the clinic. However, drug resistance is a major obstacle for BC patients, and it leads to poor prognosis. Accumulating evidences suggested that noncoding RNAs (ncRNAs) are intricately linked to a wide range of pathological processes, including drug resistance. Till date, the correlation between drug resistance and ncRNAs is not completely understood in BC. Herein, we comprehensively summarized a dysregulated ncRNAs landscape that promotes or inhibits drug resistance in chemo-, endocrine-, and targeted BC therapies. Our review will pave way for the effective management of drug resistance by targeting oncogenic ncRNAs, which, in turn will promote drug sensitivity of BC in the future.
Collapse
Affiliation(s)
- Yujuan Kang
- Department of Breast Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China.
| |
Collapse
|
7
|
Grypari IM, Pappa I, Papastergiou T, Zolota V, Bravou V, Melachrinou M, Megalooikonomou V, Tzelepi V. Elucidating the role of PRMTs in prostate cancer using open access databases and a patient cohort dataset. Histol Histopathol 2023; 38:287-302. [PMID: 36082942 DOI: 10.14670/hh-18-513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
Abstract
Protein arginine methylation is an understudied epigenetic mechanism catalyzed by enzymes known as Protein Methyltransferases of Arginine (PRMTs), while the opposite reaction is performed by Jumonji domain- containing protein 6 (JMJD6). There is increasing evidence that PRMTs are deregulated in prostate cancer (PCa). In this study, the expression of two PRMT members, PRMT2 and PRMT7 as well as JMJD6, a demethylase, was analyzed in PCa. Initially, we retrieved data from The Cancer Genome Atlas (TCGA) project and the Gene Expression Omnibus (GEO) database to explore the differential expression of various PRMT family members in patients with PCa and then applied immunohistochemistry in a patient cohort across the spectrum of PCa, including non-neoplastic prostate tissue and lymph node metastatic foci. The results from the TCGA analysis revealed that PRMT7, PRMT6 and PRMT3 expression increased while PRMT2, PRMT9 and JMJD6 levels decreased in the tumor compared to non-neoplastic prostate. Results from the GEO datasets were similar, albeit not identical with the TCGA results, with PRMT7 and PRMT3 being upregulated and PRMT2 and JMJD6 being downregulated in the tumor compared to non-neoplastic tissue in some of them. In addition, PRMT7 levels decreased with stage and grade progression in the TCGA analysis. In the patient cohort, both PRMTs and JMJD6 were overexpressed in PCa compared to non-neoplastic tissue, and nuclear PRMT2 and JMJD6 were upregulated in lymph node metastasis, too. PRMT7 and JMJD6 expression were upregulated with the progression of stage and JMJD6 was also increased with the elevation of grade. After androgen ablation therapy, nuclear expression of PRMT7 and JMJD6 were elevated compared to untreated tumors. PRMT2, PRMT7 and JMD6 were also correlated with markers of EMT and cell cycle regulators. Finally, our findings indicate that PRMTs and JMJD6 are involved in prostate cancer progression and revealed a potential interplay of PRMTs with EMT mediators, underscoring the need for therapeutic targeting of arginine methylation in prostate cancer.
Collapse
Affiliation(s)
- Ioanna Maria Grypari
- Department of Pathology, School of Medicine, University of Patras, Patras, Greece
| | - Ioanna Pappa
- Multidimensional Data Analysis and Knowledge Management Laboratory, Computer Engineering and Informatics Department, School of Engineering, University of Patras, Patras, Greece
| | - Thomas Papastergiou
- Multidimensional Data Analysis and Knowledge Management Laboratory, Computer Engineering and Informatics Department, School of Engineering, University of Patras, Patras, Greece
| | - Vasiliki Zolota
- Department of Pathology, School of Medicine, University of Patras, Patras, Greece
| | - Vasiliki Bravou
- Department of Anatomy, Histology and Embryology, School of Medicine, University of Patras, Patras, Greece
| | - Maria Melachrinou
- Department of Pathology, School of Medicine, University of Patras, Patras, Greece
| | - Vasileios Megalooikonomou
- Multidimensional Data Analysis and Knowledge Management Laboratory, Computer Engineering and Informatics Department, School of Engineering, University of Patras, Patras, Greece
| | - Vasiliki Tzelepi
- Department of Pathology, School of Medicine, University of Patras, Patras, Greece.
| |
Collapse
|
8
|
VARISLI LOKMAN, TOLAN VEYSEL, CEN JIYANH, VLAHOPOULOS SPIROS, CEN OSMAN. Dissecting the effects of androgen deprivation therapy on cadherin switching in advanced prostate cancer: A molecular perspective. Oncol Res 2023; 30:137-155. [PMID: 37305018 PMCID: PMC10208071 DOI: 10.32604/or.2022.026074] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 12/23/2022] [Indexed: 01/15/2023] Open
Abstract
Prostate cancer is one of the most often diagnosed malignancies in males and its prevalence is rising in both developed and developing countries. Androgen deprivation therapy has been used as a standard treatment approach for advanced prostate cancer for more than 80 years. The primary aim of androgen deprivation therapy is to decrease circulatory androgen and block androgen signaling. Although a partly remediation is accomplished at the beginning of treatment, some cell populations become refractory to androgen deprivation therapy and continue to metastasize. Recent evidences suggest that androgen deprivation therapy may cause cadherin switching, from E-cadherin to N-cadherin, which is the hallmark of epithelial-mesenchymal transition. Diverse direct and indirect mechanisms are involved in this switching and consequently, the cadherin pool changes from E-cadherin to N-cadherin in the epithelial cells. Since E-cadherin represses invasive and migrative behaviors of the tumor cells, the loss of E-cadherin disrupts epithelial tissue structure leading to the release of tumor cells into surrounding tissues and circulation. In this study, we review the androgen deprivation therapy-dependent cadherin switching in advanced prostate cancer with emphasis on its molecular basis especially the transcriptional factors regulated through TFG-β pathway.
Collapse
Affiliation(s)
- LOKMAN VARISLI
- Department of Molecular Biology and Genetics, Science Faculty, Dicle University, Diyarbakir, 21280, Turkey
- Cancer Research Center, Dicle University, Diyarbakir, 21280, Turkey
| | - VEYSEL TOLAN
- Department of Molecular Biology and Genetics, Science Faculty, Dicle University, Diyarbakir, 21280, Turkey
| | - JIYAN H. CEN
- Department of Chemical and Biomolecular Engineering, University of Illinois, Urbana, IL, 61801, USA
| | - SPIROS VLAHOPOULOS
- First Department of Pediatrics, National and Kapodistrian University of Athens, Athens, 11527, Greece
| | - OSMAN CEN
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Department of Natural Sciences and Engineering, John Wood College, Quincy, IL, 62305, USA
| |
Collapse
|
9
|
Stella S, Martorana F, Massimino M, Vitale SR, Manzella L, Vigneri P. Potential Therapeutic Targets for Luminal Androgen Receptor Breast Cancer: What We Know so Far. Onco Targets Ther 2023; 16:235-247. [PMID: 37056632 PMCID: PMC10089148 DOI: 10.2147/ott.s379867] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 03/24/2023] [Indexed: 04/15/2023] Open
Abstract
Luminal Androgen Receptor Breast Cancers (LAR BCs) are characterized by a triple negative phenotype and by the expression of Androgen Receptor (AR), coupled with luminal-like genomic features. This unique BC subtype, accounting for about 10% of all triple negative BC, has raised considerable interest given its ill-defined clinical behavior and the chance to exploit AR as a therapeutic target. The complexity of AR activity in BC cells, as revealed by decades of mechanistic studies, holds promise to offer additional therapeutic options beyond mere AR inhibition. Indeed, preclinical and translational evidence showed that several pathways and mediators, including PI3K/mToR, HER2, BRCA1, cell cycle and immune modulation, can be tackled in LAR BCs. Moving from bench to bedside, several clinical trials tested anti-androgen therapies in LAR BCs, but their results are inconsistent and often disappointing. More recently, studies exploring combinations of anti-androgen agents with other targeted therapies have been designed and are currently ongoing. While the results from these trials are awaited, a concerted effort will be needed to find the biological vulnerabilities of LAR BCs which may disclose new and effective therapeutic targets, eventually improving patients' outcomes.
Collapse
Affiliation(s)
- Stefania Stella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico “G. Rodolico - San Marco”, Catania, Italy
- Correspondence: Stefania Stella, University of Catania, Department of Clinical and Experimental Medicine, Center of Experimental Oncology and Hematology, A.O.U. Policlinico “G. Rodolico - San Marco”, Via S. Sofia, 78, Edificio 8D/2, Catania, Italy, Tel +39 95 378 1946, Email ;
| | - Federica Martorana
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico “G. Rodolico - San Marco”, Catania, Italy
| | - Michele Massimino
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico “G. Rodolico - San Marco”, Catania, Italy
| | - Silvia Rita Vitale
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico “G. Rodolico - San Marco”, Catania, Italy
| | - Livia Manzella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico “G. Rodolico - San Marco”, Catania, Italy
| | - Paolo Vigneri
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico “G. Rodolico - San Marco”, Catania, Italy
| |
Collapse
|
10
|
Tahtamouni L, Alzghoul A, Alderfer S, Sun J, Ahram M, Prasad A, Bamburg J. The role of activated androgen receptor in cofilin phospho-regulation depends on the molecular subtype of TNBC cell line and actin assembly dynamics. PLoS One 2022; 17:e0279746. [PMID: 36584207 PMCID: PMC9803305 DOI: 10.1371/journal.pone.0279746] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 12/13/2022] [Indexed: 12/31/2022] Open
Abstract
Triple negative breast cancer (TNBC) is highly metastatic and of poor prognosis. Metastasis involves coordinated actin filament dynamics mediated by cofilin and associated proteins. Activated androgen receptor (AR) is believed to contribute to TNBC tumorigenesis. Our current work studied roles of activated AR and cofilin phospho-regulation during migration of three AR+ TNBC cell lines to determine if altered cofilin regulation can explain their migratory differences. Untreated or AR agonist-treated BT549, MDA-MB-453, and SUM159PT cells were compared to cells silenced for cofilin (KD) or AR expression/function (bicalutamide). Cofilin-1 was found to be the only ADF/cofilin isoform expressed in each TNBC line. Despite a significant increase in cofilin kinase caused by androgens, the ratio of cofilin:p-cofilin (1:1) did not change in SUM159PT cells. BT549 and MDA-MB-453 cells contain high p-cofilin levels which underwent androgen-induced dephosphorylation through increased cofilin phosphatase expression, but surprisingly maintain a leading-edge with high p-cofilin/total cofilin not found in SUM159PT cells. Androgens enhanced cell polarization in all lines, stimulated wound healing and transwell migration rates and increased N/E-cadherin mRNA ratios while reducing cell adhesion in BT549 and MDA-MB-453 cells. Cofilin KD negated androgen effects in MDA-MB-453 except for cell adhesion, while in BT549 cells it abrogated androgen-reduced cell adhesion. In SUM159PT cells, cofilin KD with and without androgens had similar effects in almost all processes studied. AR dependency of the processes were confirmed. In conclusion, cofilin regulation downstream of active AR is dependent on which actin-mediated process is being examined in addition to being cell line-specific. Although MDA-MB-453 cells demonstrated some control of cofilin through an AR-dependent mechanism, other AR-dependent pathways need to be further studied. Non-cofilin-dependent mechanisms that modulate migration of SUM159PT cells need to be investigated. Categorizing TNBC behavior as AR responsive and/or cofilin dependent can inform on decisions for therapeutic treatment.
Collapse
Affiliation(s)
- Lubna Tahtamouni
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
- Department of Biochemistry and Molecular Biology, College of Natural Sciences, Colorado State University, Fort Collins, CO, United States of America
- * E-mail: ,
| | - Ahmad Alzghoul
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Sydney Alderfer
- Department of Chemical and Biological Engineering, School of Biomedical Engineering, Colorado State University, Fort Collins, CO, United States of America
| | - Jiangyu Sun
- Department of Biochemistry and Molecular Biology, College of Natural Sciences, Colorado State University, Fort Collins, CO, United States of America
| | - Mamoun Ahram
- Department of Physiology and Biochemistry, School of Medicine, The University of Jordan, Amman, Jordan
| | - Ashok Prasad
- Department of Chemical and Biological Engineering, School of Biomedical Engineering, Colorado State University, Fort Collins, CO, United States of America
| | - James Bamburg
- Department of Biochemistry and Molecular Biology, College of Natural Sciences, Colorado State University, Fort Collins, CO, United States of America
| |
Collapse
|
11
|
Cingir Koker S, Yalcin B, Dogan Turacli I. Metformin resistant MDA-MB-468 cells exhibit EMT-like phenotype and increased migration capacity. Mol Biol Rep 2022; 49:5973-5984. [PMID: 35355210 DOI: 10.1007/s11033-022-07381-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 03/16/2022] [Indexed: 10/18/2022]
Abstract
PURPOSE Metformin is one of the most prescribed drugs for the treatment of type II diabetes. Its anti-proliferative effect is also taken advantage for the treatment of cancer. Despite many of the studies mentioning the positive effects of metformin in inhibiting the proliferation of cancer cells, there are also studies which questions this idea as well. METHODS In this study, we investigated the most widely studied breast cancer cell lines, ER (+) MCF7 cells, TNBC MDA-MB-231 and MDA-MB-468 cells in terms of metastatic behavior under long-term metformin treatment. MCF7, MDA-MB-231 and MDA-MB-468 cells were gained resistant to metformin starting from 0.2 to 3.2 mM. RESULTS Compared to MCF7 and MDA-MB-231 cell lines, we only observed dramatic changes in MDA-MB-468 cells whose morphology has been changed towards mesenchymal like phenotype. Moreover, migration capacity of these cells was also significantly increased which were validated at both mRNA and protein levels as well as wound healing assay. In addition to EMT like phenotype and increasing migration capacity of metformin resistant MDA-MB-468 cells, they exhibited less sensitivity to PI3K inhibitor. CONCLUSIONS All together, our data pointed out that, metformin's effects should be questioned depending on the subtype of the breast cancer that's to be treated.
Collapse
Affiliation(s)
- Sahika Cingir Koker
- Department of Medical Biology, Ufuk University, Mevlana Bulvarı 86-88, 06520, Balgat, Ankara, Turkey.
- ONTAL, Ufuk University, Mevlana Bulvarı 86-88, 06520, Balgat, Ankara, Turkey.
| | - Banu Yalcin
- ONTAL, Ufuk University, Mevlana Bulvarı 86-88, 06520, Balgat, Ankara, Turkey
| | - Irem Dogan Turacli
- Department of Medical Biology, Ufuk University, Mevlana Bulvarı 86-88, 06520, Balgat, Ankara, Turkey
- ONTAL, Ufuk University, Mevlana Bulvarı 86-88, 06520, Balgat, Ankara, Turkey
| |
Collapse
|
12
|
Characteristics of the TDRD1 gene promoter in chickens. Mol Genet Genomics 2022; 297:903-910. [PMID: 35347417 DOI: 10.1007/s00438-022-01886-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 03/14/2022] [Indexed: 10/18/2022]
Abstract
Tudor domain containing 1 (TDRD1) is a member of the TDRD family and plays an important role in embryogenesis and gametogenesis. A detailed study of the characteristics of chicken TDRD1 can lay a foundation for the study of chicken spermatogonia stem cell formation and spermatogenesis. We cloned 2117 bp upstream fragment of TDRD1 promoter and constructed a series of recombinant vectors with different length deletions. The dual-luciferase experiments reveal that the upstream region of - 161 to 0 bp was its core transcription promoter region. Bioinformatics analysis predicted the possible binding of Transcription Factor 7 Like 2 (TCF7L2) and Zinc Finger E-Box-Binding Homeobox 1(ZEB1) transcription factors in the core region. The transcriptional activity of TDRD1 was significantly decreased after mutation of TCF7L2-binding site, while that of TDRD1 was significantly increased after mutation of ZEB1-binding site. Further, ChIP experiments verified that TCF7L2 enriched in the TDRD1 core transcriptional initiation region, suggesting that TCF7L2 and ZEB1 play an important role in the regulation of TDRD1. In summary, the region from - 161 to 0 bp is the core promoter region of TDRD1; ZEB1 and TCF7L2 bind to the TDRD1 promoter region and TCF7L2 activates the transcription of TDRD1 gene.
Collapse
|
13
|
Kulkarni P, Leite VBP, Roy S, Bhattacharyya S, Mohanty A, Achuthan S, Singh D, Appadurai R, Rangarajan G, Weninger K, Orban J, Srivastava A, Jolly MK, Onuchic JN, Uversky VN, Salgia R. Intrinsically disordered proteins: Ensembles at the limits of Anfinsen's dogma. BIOPHYSICS REVIEWS 2022; 3:011306. [PMID: 38505224 PMCID: PMC10903413 DOI: 10.1063/5.0080512] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/17/2022] [Indexed: 03/21/2024]
Abstract
Intrinsically disordered proteins (IDPs) are proteins that lack rigid 3D structure. Hence, they are often misconceived to present a challenge to Anfinsen's dogma. However, IDPs exist as ensembles that sample a quasi-continuum of rapidly interconverting conformations and, as such, may represent proteins at the extreme limit of the Anfinsen postulate. IDPs play important biological roles and are key components of the cellular protein interaction network (PIN). Many IDPs can interconvert between disordered and ordered states as they bind to appropriate partners. Conformational dynamics of IDPs contribute to conformational noise in the cell. Thus, the dysregulation of IDPs contributes to increased noise and "promiscuous" interactions. This leads to PIN rewiring to output an appropriate response underscoring the critical role of IDPs in cellular decision making. Nonetheless, IDPs are not easily tractable experimentally. Furthermore, in the absence of a reference conformation, discerning the energy landscape representation of the weakly funneled IDPs in terms of reaction coordinates is challenging. To understand conformational dynamics in real time and decipher how IDPs recognize multiple binding partners with high specificity, several sophisticated knowledge-based and physics-based in silico sampling techniques have been developed. Here, using specific examples, we highlight recent advances in energy landscape visualization and molecular dynamics simulations to discern conformational dynamics and discuss how the conformational preferences of IDPs modulate their function, especially in phenotypic switching. Finally, we discuss recent progress in identifying small molecules targeting IDPs underscoring the potential therapeutic value of IDPs. Understanding structure and function of IDPs can not only provide new insight on cellular decision making but may also help to refine and extend Anfinsen's structure/function paradigm.
Collapse
Affiliation(s)
- Prakash Kulkarni
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, California 91010, USA
| | - Vitor B. P. Leite
- Departamento de Física, Instituto de Biociências, Letras e Ciências Exatas, Universidade Estadual Paulista (UNESP), São José do Rio Preto, São Paulo 15054-000, Brazil
| | - Susmita Roy
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal 741246, India
| | - Supriyo Bhattacharyya
- Translational Bioinformatics, Center for Informatics, Department of Computational and Quantitative Medicine, City of Hope National Medical Center, Duarte, California 91010, USA
| | - Atish Mohanty
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, California 91010, USA
| | - Srisairam Achuthan
- Center for Informatics, Division of Research Informatics, City of Hope National Medical Center, Duarte, California 91010, USA
| | - Divyoj Singh
- Center for BioSystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India
| | - Rajeswari Appadurai
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, Karnataka, India
| | - Govindan Rangarajan
- Department of Mathematics, Indian Institute of Science, Bangalore 560012, India
| | - Keith Weninger
- Department of Physics, North Carolina State University, Raleigh, North Carolina 27695, USA
| | | | - Anand Srivastava
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, Karnataka, India
| | - Mohit Kumar Jolly
- Center for BioSystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India
| | - Jose N. Onuchic
- Center for Theoretical Biological Physics, Rice University, Houston, Texas 77005-1892, USA
| | | | - Ravi Salgia
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, California 91010, USA
| |
Collapse
|
14
|
Kwon OJ, Zhang B, Jia D, Zhang L, Wei X, Zhou Z, Liu D, Huynh KT, Zhang K, Zhang Y, Labhart P, Sboner A, Barbieri C, Haffner MC, Creighton CJ, Xin L. Elevated expression of the colony-stimulating factor 1 (CSF1) induces prostatic intraepithelial neoplasia dependent of epithelial-Gp130. Oncogene 2022; 41:1309-1323. [PMID: 34999736 PMCID: PMC8882147 DOI: 10.1038/s41388-021-02169-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 12/06/2021] [Accepted: 12/22/2021] [Indexed: 11/24/2022]
Abstract
Macrophages are increased in human benign prostatic hyperplasia and prostate cancer. We generate a Pb-Csf1 mouse model with prostate-specific overexpression of macrophage colony-stimulating factor (M-Csf/Csf1). Csf1 overexpression promotes immune cell infiltration into the prostate, modulates the macrophage polarity in a lobe-specific manner, and induces senescence and low-grade prostatic intraepithelial neoplasia (PIN). The Pb-Csf1 prostate luminal cells exhibit increased stem cell features and undergo an epithelial-to-mesenchymal transition. Human prostate cancer patients with high CSF-1 expression display similar transcriptional alterations with the Pb-Csf1 model. P53 knockout alleviates senescence but fails to progress PIN lesions. Ablating epithelial Gp130 but not Il1r1 substantially blocks PIN lesion formation. The androgen receptor (AR) is downregulated in Pb-Csf1 mice. ChIP-Seq analysis reveals altered AR binding in 2482 genes although there is no significant widespread change in global AR transcriptional activity. Collectively, our study demonstrates that increased macrophage infiltration causes PIN formation but fails to transform prostate cells.
Collapse
Affiliation(s)
- Oh-Joon Kwon
- Department of Urology, University of Washington, Seattle, WA, 98109, USA
| | - Boyu Zhang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Deyong Jia
- Department of Urology, University of Washington, Seattle, WA, 98109, USA
| | - Li Zhang
- Department of Urology, University of Washington, Seattle, WA, 98109, USA
| | - Xing Wei
- Department of Urology, University of Washington, Seattle, WA, 98109, USA
| | - Zhicheng Zhou
- Department of Urology, University of Washington, Seattle, WA, 98109, USA
| | - Deli Liu
- Sandra and Edward Meyer Cancer Center and Department of Urology, Weill Cornell Medicine, New York, NY, USA
| | - Khoi Trung Huynh
- Department of Biology, University of Washington, Seattle, WA, 98109, USA
| | - Kai Zhang
- Department of Urology, University of Washington, Seattle, WA, 98109, USA
| | - Yiqun Zhang
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | | | - Andrea Sboner
- Sandra and Edward Meyer Cancer Center and Department of Urology, Weill Cornell Medicine, New York, NY, USA
| | - Chris Barbieri
- Sandra and Edward Meyer Cancer Center and Department of Urology, Weill Cornell Medicine, New York, NY, USA
| | - Michael C Haffner
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98109, USA
| | - Chad J Creighton
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Li Xin
- Department of Urology, University of Washington, Seattle, WA, 98109, USA.
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, 98109, USA.
| |
Collapse
|
15
|
Ahram M, Abdullah MS, Mustafa SA, Alsafadi DB, Battah AH. Androgen down-regulates desmocollin 2 in association with induction of mesenchymal transition of breast MDA-MB-453 cancer cells. Cytoskeleton (Hoboken) 2022; 78:391-399. [PMID: 35023302 DOI: 10.1002/cm.21691] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 01/06/2022] [Accepted: 01/08/2022] [Indexed: 11/06/2022]
Abstract
Desmosomes are cellular structures that are critical in cell-cell adhesion and in maintaining tissue architecture. Changes in the expression of desmocollin-2 (DSC2) have been noted during tumor progression into an invasive phenotype and as cells undergo epithelial-mesenchymal transition. We have previously reported that breast MDA-MB-453 cancer cells, a luminal androgen receptor model of triple-negative breast cancer, acquire mesenchymal features when treated with the androgen receptor (AR) agonist, dihydrotestosterone (DHT). We have therefore investigated androgen regulation of the expression and cellular localization of DSC2 in MDA-MB-453 cells. Treatment of the cells with DHT resulted in a dose-dependent reduction in DSC2 protein levels and dispersion of its membrane localization concomitant with AR- and β-catenin-mediated mesenchymal transition of cells. A significant correlation was revealed between decreased expression of AR and increased expression of DSC2 in patient samples. In addition, whereas lower expression of AR was associated with a reduced overall and recurrence-free survival of breast cancer patients, higher expression of DSC2 was found in invasive breast tumors than in normal breast cells and was correlated with lower patient survival. Upon knocking down DSC2, the cells became elongated, mesenchymal-like, and slightly, but insignificantly, more migratory. The addition of DHT further stimulated cell elongation and migration. DSC2 siRNA-transfected cells reverted to a normal epithelial morphology upon inhibition of β-catenin. These results highlight the role of DSC2 in maintaining the epithelial morphology of MDA-MB-453 cells and the negative regulation of the desmosomal protein by DHT during stimulation of the androgen-induced, β-catenin-mediated mesenchymal transition of the cells. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Mamoun Ahram
- Department of Physiology and Biochemistry, School of Medicine, The University of Jordan, Amman, Jordan
| | - Mohammad S Abdullah
- Department of Physiology and Biochemistry, School of Medicine, The University of Jordan, Amman, Jordan
| | - Shahed A Mustafa
- Department of Microbiology, Pathology, and Forensic Medicine, School of Medicine, The University of Jordan, Amman, Jordan
| | - Dana B Alsafadi
- Department of Physiology and Biochemistry, School of Medicine, The University of Jordan, Amman, Jordan
| | - Abdelkader H Battah
- Department of Microbiology, Pathology, and Forensic Medicine, School of Medicine, The University of Jordan, Amman, Jordan
| |
Collapse
|
16
|
The Transcription Factors Zeb1 and Snail Induce Cell Malignancy and Cancer Stem Cell Phenotype in Prostate Cells, Increasing Androgen Synthesis Capacity and Therapy Resistance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1393:51-64. [PMID: 36587301 DOI: 10.1007/978-3-031-12974-2_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Prostate cancer (PCa) incidence has increased during the last decades, becoming one of the leading causes of death by cancer in men worldwide. During an extended period of prostate cancer, malignant cells are androgen-sensitive being testosterone the main responsible for tumor growth. Accordingly, treatments blocking production and action of testosterone are mostly used. However, during disease progression, PCa cells become androgen insensitive producing a castration-resistant stage with a worse prognosis. Overcoming castration-resistant prostate cancer (CRPC) has become a great challenge in the management of this disease. In the search for molecular pathways leading to therapy resistance, the epithelial-mesenchymal transition (EMT), and particularly the transcription factors zinc finger E-box-binding homeobox 1 (Zeb1) and zinc finger protein SNAI1 (Snail), master genes of the EMT, have shown to have pivotal roles. Also, the discovery that cancer stem cells (CSCs) can be generated de novo from their non-CSCs counterpart has led to the question whereas these EMT transcription factors could be implicated in this dynamic conversion between non-CSC and CSC. In this review, we analyze evidence supporting the idea that Zeb1 and Snail induce cell malignancy and cancer stem cell phenotype in prostate cells, increasing androgen synthesis capacity and therapy resistance.
Collapse
|
17
|
Kitz J, Lefebvre C, Carlos J, Lowes LE, Allan AL. Reduced Zeb1 Expression in Prostate Cancer Cells Leads to an Aggressive Partial-EMT Phenotype Associated with Altered Global Methylation Patterns. Int J Mol Sci 2021; 22:ijms222312840. [PMID: 34884649 PMCID: PMC8657557 DOI: 10.3390/ijms222312840] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/22/2021] [Accepted: 11/24/2021] [Indexed: 12/20/2022] Open
Abstract
Prostate cancer is the most common cancer in American men and the second leading cause of cancer-related death. Most of these deaths are associated with metastasis, a process involving the epithelial-to-mesenchymal (EMT) transition. Furthermore, growing evidence suggests that partial-EMT (p-EMT) may lead to more aggressive disease than complete EMT. In this study, the EMT-inducing transcription factor Zeb1 was knocked down in mesenchymal PC-3 prostate cancer cells (Zeb1KD) and resulting changes in cellular phenotype were assessed using protein and RNA analysis, invasion and migration assays, cell morphology assays, and DNA methylation chip analysis. Inducible knockdown of Zeb1 resulted in a p-EMT phenotype including co-expression of epithelial and mesenchymal markers, a mixed epithelial/mesenchymal morphology, increased invasion and migration, and enhanced expression of p-EMT markers relative to PC-3 mesenchymal controls (p ≤ 0.05). Treatment of Zeb1KD cells with the global de-methylating drug 5-azacytidine (5-aza) mitigated the observed aggressive p-EMT phenotype (p ≤ 0.05). DNA methylation chip analysis revealed 10 potential targets for identifying and/or targeting aggressive p-EMT prostate cancer in the future. These findings provide a framework to enhance prognostic and/or therapeutic options for aggressive prostate cancer in the future by identifying new p-EMT biomarkers to classify patients with aggressive disease who may benefit from 5-aza treatment.
Collapse
Affiliation(s)
- Jenna Kitz
- London Regional Cancer Program, London Health Sciences Centre, Department of Anatomy & Cell Biology, Western University, London, ON N6A 5W9, Canada; (J.K.); (C.L.)
| | - Cory Lefebvre
- London Regional Cancer Program, London Health Sciences Centre, Department of Anatomy & Cell Biology, Western University, London, ON N6A 5W9, Canada; (J.K.); (C.L.)
| | - Joselia Carlos
- Department of Medical Biophysics, Western University, London, ON N6A 5C1, Canada;
| | - Lori E. Lowes
- Flow Cytometry, London Health Sciences Centre, London, ON N6A 5W9, Canada;
| | - Alison L. Allan
- London Regional Cancer Program, London Health Sciences Centre, Department of Anatomy & Cell Biology, Western University, London, ON N6A 5W9, Canada; (J.K.); (C.L.)
- Department of Oncology, Western University, London, ON N6A 5W9, Canada
- Cancer Research Laboratory Program, Lawson Health Research Institute, London, ON N6C 2R5, Canada
- Correspondence: ; Tel.: +1-519-685-8600 (ext. 55134)
| |
Collapse
|
18
|
Carcinogenesis of Triple-Negative Breast Cancer and Sex Steroid Hormones. Cancers (Basel) 2021; 13:cancers13112588. [PMID: 34070471 PMCID: PMC8197527 DOI: 10.3390/cancers13112588] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 05/19/2021] [Accepted: 05/23/2021] [Indexed: 11/21/2022] Open
Abstract
Simple Summary Triple-negative breast cancer (TNBC) lacks all of three treatment targets (estrogen receptor-α, ER-α; progesterone receptor, PgR; and human epidermal growth factor receptor 2, HER2) and is usually associated with a poor clinical outcome; however, several sex steroid receptors, such as androgen receptor (AR), ER-β, and G-protein-coupled estrogen receptor, are frequently expressed and their biological and clinical importance has been suggested. Despite the structural similarity between sex steroid hormones (androgens and estrogens) or receptors (AR and ER-β), similar signaling mechanisms of these hormones, and the coexistence of these hormones and their receptors in TNBC in a clinical setting, most studies or reviews focused on only one of these receptors, and rarely reviewed them in a comprehensive way. In this review, the carcinogenic or pathobiological role of sex steroid hormones in TNBC is considered, focusing on common and differing features of hormone actions. Abstract Triple-negative breast cancer (TNBC) lacks an effective treatment target and is usually associated with a poor clinical outcome; however, hormone unresponsiveness, which is the most important biological characteristic of TNBC, only means the lack of nuclear estrogenic signaling through the classical estrogen receptor (ER), ER-α. Several sex steroid receptors other than ER-α: androgen receptor (AR), second ER, ER-β, and non-nuclear receptors represented by G-protein-coupled estrogen receptor (GPER), are frequently expressed in TNBC and their biological and clinical importance has been suggested by a large number of studies. Despite the structural similarity between each sex steroid hormone (androgens and estrogens) or each receptor (AR and ER-β), and similarity in the signaling mechanisms of these hormones, most studies or reviews focused on one of these receptors, and rarely reviewed them in a comprehensive way. Considering the coexistence of these hormones and their receptors in TNBC in a clinical setting, a comprehensive viewpoint would be important to correctly understand the association between the carcinogenic mechanism or pathobiology of TNBC and sex steroid hormones. In this review, the carcinogenic or pathobiological role of sex steroid hormones in TNBC is considered, focusing on the common and divergent features of the action of these hormones.
Collapse
|
19
|
Singh D, Bocci F, Kulkarni P, Jolly MK. Coupled Feedback Loops Involving PAGE4, EMT and Notch Signaling Can Give Rise to Non-genetic Heterogeneity in Prostate Cancer Cells. ENTROPY (BASEL, SWITZERLAND) 2021; 23:288. [PMID: 33652914 PMCID: PMC7996788 DOI: 10.3390/e23030288] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/18/2021] [Accepted: 02/23/2021] [Indexed: 12/12/2022]
Abstract
Non-genetic heterogeneity is emerging as a crucial factor underlying therapy resistance in multiple cancers. However, the design principles of regulatory networks underlying non-genetic heterogeneity in cancer remain poorly understood. Here, we investigate the coupled dynamics of feedback loops involving (a) oscillations in androgen receptor (AR) signaling mediated through an intrinsically disordered protein PAGE4, (b) multistability in epithelial-mesenchymal transition (EMT), and c) Notch-Delta-Jagged signaling mediated cell-cell communication, each of which can generate non-genetic heterogeneity through multistability and/or oscillations. Our results show how different coupling strengths between AR and EMT signaling can lead to monostability, bistability, or oscillations in the levels of AR, as well as propagation of oscillations to EMT dynamics. These results reveal the emergent dynamics of coupled oscillatory and multi-stable systems and unravel mechanisms by which non-genetic heterogeneity in AR levels can be generated, which can act as a barrier to most existing therapies for prostate cancer patients.
Collapse
Affiliation(s)
- Divyoj Singh
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India;
- Undergraduate Programme, Indian Institute of Science, Bangalore 560012, India
| | - Federico Bocci
- Center for Theoretical Biological Physics, Rice University, Houston, TX 77005, USA;
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA 92697, USA
| | - Prakash Kulkarni
- Department of Medical Oncology and Experimental Therapeutics, City of Hope National Medical Center, Duarte, CA 91010, USA;
| | - Mohit Kumar Jolly
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India;
| |
Collapse
|
20
|
Al-Othman N, Ahram M, Alqaraleh M. Role of androgen and microRNA in triple-negative breast cancer. Breast Dis 2020; 39:15-27. [PMID: 31839601 DOI: 10.3233/bd-190416] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Breast cancer (BC) is the most frequent type of malignancy affecting females worldwide. Molecular-based studies resulted in an identification of at least four subtypes of breast carcinoma, including luminal A and luminal B, Human growth factor receptor (HER-2)-enriched and triple-negative tumors (basal-like and normal breast-like). A proportion of BC cases are of the triple-negative breast cancer (TNBC) type. TNBC lacks the expression of estrogen receptor (ER), progesterone receptor (PR), and HER-2, and is known to express androgen receptor (AR) at considerable levels. AR has been shown to promote the progression of TNBC. However, the exact mechanisms have yet to be unraveled. One of these mechanisms could be through regulating the expression of microRNA (miRNA) molecules, which play an important regulatory role in BC through post-transcriptional gene silencing. Activation of AR controls the expression of miRNA molecules, which target selective mRNAs, consequently, affecting protein expression. In this review we attempt to elucidate the relations between AR and miRNA in TNBC.
Collapse
Affiliation(s)
- Nihad Al-Othman
- Division of Anatomy, Biochemistry and Genetic, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Mamoun Ahram
- Department of Physiology and Biochemistry, School of Medicine, The University of Jordan, Amman, Jordan
| | - Moath Alqaraleh
- Department of Biological Sciences, School of Science, The University of Jordan, Amman, Jordan
| |
Collapse
|
21
|
Gao J, Yuan Y, Zhang L, Yu S, Lu J, Feng J, Hu S. Inhibition of ZEB1-AS1 confers cisplatin sensitivity in breast cancer by promoting microRNA-129-5p-dependent ZEB1 downregulation. Cancer Cell Int 2020; 20:90. [PMID: 32210737 PMCID: PMC7092489 DOI: 10.1186/s12935-020-1164-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 03/04/2020] [Indexed: 11/22/2022] Open
Abstract
Background Breast cancer is the leading cause of cancer-related mortality in women worldwide. Long non-coding RNAs (lncRNAs) are of critical importance in tumor drug resistance. Herein, this study aims to determine the roles of lncRNA ZEB1-AS1 in drug resistance of breast cancer involving microRNA-129-5p (miR-129-5p) and ZEB1. Methods Microarray-based gene expression profiling of breast cancer was conducted to identify the differentially expressed lncRNAs. ZEB1 expression was measured in adjacent and cancerous tissues. Next, MCF-7 and MDA-MB-231 cells were treated with a series of inhibitor, mimic or siRNA to clarify the roles of lncRNA ZEB1-AS1 and miR-129-5p in drug resistance of breast cancer. Then the target relationship of miR-129-5p with lncRNA ZEB1-AS1 and ZEB1 was verified. The expression patterns of miR-129-5p, lncRNA ZEB1-AS1, Bcl-2, MDR-1, ZEB1 and corresponding proteins were evaluated. Moreover, the apoptosis and drug resistance of MCF-7 cell were detected by CCK-8 and flow cytometry respectively. Results LncRNA ZEB1-AS1 was observed to be an upregulated lncRNA in breast cancer, and ZEB1 overexpression was noted in breast cancerous tissues. MiR-129-5p was revealed to specifically bind to both ZEB1 and lncRNA ZEB1-AS1. Moreover, the expression levels of ZEB1-AS1, ZEB1, Bcl-2, MDR-1, and corresponding proteins were decreased, but the expression of miR-129-5p was increased with transfection of miR-129-5p mimic and lncRNA ZEB1-AS1 siRNA. Besides, drug resistance to cisplatin was inhibited, and cell apoptosis was promoted in breast cancer after transfection of miR-129-5p mimic, lncRNA ZEB1-AS1 siRNA, and ZEB1 siRNA. Conclusion In conclusion, the study provides evidence that lncRNA ZEB1-AS1 silencing protects against drug resistance in breast cancer by promoting miR-129-5p-dependent ZEB1 downregulation. It may serve as a novel therapeutic target in breast cancer treatment.
Collapse
Affiliation(s)
- Jin Gao
- Department of Medical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, No. 42, Baiziting, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Yuan Yuan
- Department of Medical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, No. 42, Baiziting, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Lili Zhang
- Department of Medical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, No. 42, Baiziting, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Shaorong Yu
- Department of Medical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, No. 42, Baiziting, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Jianwei Lu
- Department of Medical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, No. 42, Baiziting, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Jifeng Feng
- Department of Medical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, No. 42, Baiziting, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Sainan Hu
- Department of Medical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, No. 42, Baiziting, Nanjing, 210009, Jiangsu, People's Republic of China.
| |
Collapse
|
22
|
Soleymani Fard S, Sotoudeh M, Saliminejad K, Yazdanbod M, Mahmoodzadeh H, Kouchaki S, Yaghmaie M, Mousavi SA, Malekzadeh R, Alimoghaddam K, Ghaffari SH. Investigation of the Correlation between Androgen Receptor and ZEB1 and its Value in Progression of Gastric Cancer. Avicenna J Med Biotechnol 2020; 12:52-60. [PMID: 32153739 PMCID: PMC7035462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
BACKGROUND Zinc-finger Enhancer Binding protein (ZEB1) acts as a transcription factor to promote cancer progression through regulating Epithelial to Mesenchymal Transition (EMT). It is well-known that ZEB1 mRNA expression is directly induced by both Estrogen Receptor (ER) and Progesterone Receptor (PR). Moreover, Androgen Receptor (AR) and PR could bind to the same regulatory element. Since it has been shown that AR overexpresses in Gastric Cancer (GC) as a male-predominant tumor, the goal of this study was to evaluate whether AR could regulate ZEB1 expression in GC. METHODS The expression profile of ZEB1 in 60 fresh GC and adjacent non-tumor tissues and 50 normal gastric specimens was assessed by qRT-PCR, and the association of ZEB1 expression with clinicopathological features was investigated. Furthermore, possible correlation between ZEB1 and AR was evaluated to elucidate a novel prognostic marker using Kaplan-Meier method and Cox regression model. Finally, molecular interaction of ZEB1 and AR was assessed using a potent AR antagonist in GC cells. RESULTS Among GC patients, 70.2% (40/57) overexpressed ZEB1 and 64.91% (37/57) overexpressed AR relative to normal gastric tissues. ZEB1 overexpression was significantly correlated with the AR overexpression in GC patients. Moreover, ZEB1 overexpression was remarkably associated with lower overall survival; however, it was not an independent prognostic factor. Evidence shows that simultaneous evaluation of ZEB1 and AR expression could independently predict survival of GC patients (HR= 2.193, p=0.047). CONCLUSION These findings have clinical importance suggesting simultaneous evaluation of ZEB1 and AR expression as a potential prognostic marker. Moreover, AR may regulate ZEB1 expression in GC cells proposing a possible promising targeted therapy for GC patients.
Collapse
Affiliation(s)
- Shahrzad Soleymani Fard
- Hematology, Oncology and Stem Cell Transplantation Research Institute, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoud Sotoudeh
- Digestive Oncology Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Kioomars Saliminejad
- Reproductive Biotechnology Research Center, Avicenna Research Institute (ACECR), Tehran, Iran
| | | | - Habibollah Mahmoodzadeh
- Department of Surgical Oncology, Cancer Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Shaghayegh Kouchaki
- Hematology, Oncology and Stem Cell Transplantation Research Institute, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Marjan Yaghmaie
- Hematology, Oncology and Stem Cell Transplantation Research Institute, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Asadollah Mousavi
- Hematology, Oncology and Stem Cell Transplantation Research Institute, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Malekzadeh
- Digestive Oncology Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Kamran Alimoghaddam
- Hematology, Oncology and Stem Cell Transplantation Research Institute, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran,Corresponding authors: Kamran Alimoghaddam, Ph.D., Hematology, Oncology and Stem Cell Transplantation Research Institute, Shariati Hospital, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran, Seyed Hamidollah Ghaffari, Ph.D., Hematology, Oncology and Stem Cell Transplantation Research Institute, Shariati Hospital, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran, Tel: +98 21 84902665, Fax: +98 21 88004140, E-mail: ;,
| | - Seyed Hamidollah Ghaffari
- Hematology, Oncology and Stem Cell Transplantation Research Institute, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran,Corresponding authors: Kamran Alimoghaddam, Ph.D., Hematology, Oncology and Stem Cell Transplantation Research Institute, Shariati Hospital, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran, Seyed Hamidollah Ghaffari, Ph.D., Hematology, Oncology and Stem Cell Transplantation Research Institute, Shariati Hospital, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran, Tel: +98 21 84902665, Fax: +98 21 88004140, E-mail: ;,
| |
Collapse
|
23
|
Yang CW, Cao HH, Guo Y, Feng YM, Zhang N. Identification of Novel Breast Cancer Genes based on Gene Expression Profiles and PPI Data. CURR PROTEOMICS 2019. [DOI: 10.2174/1570164616666190126111354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:Breast cancer is one of the most common malignancies, and a threat to female health all over the world. However, the molecular mechanism of breast cancer has not been fully discovered yet.Objective:It is crucial to identify breast cancer-related genes, which could provide new biomarker for breast cancer diagnosis as well as potential treatment targets.Methods:Here we used the minimum redundancy-maximum relevance (mRMR) method to select significant genes, then mapped the transcripts of the genes on the Protein-Protein Interaction (PPI) network and traced the shortest path between each pair of two proteins.Results:As a result, we identified 24 breast cancer-related genes whose betweenness were over 700. The GO enrichment analysis indicated that the transcription and oxygen level are very important in breast cancer. And the pathway analysis indicated that most of these 24 genes are enriched in prostate cancer, endocrine resistance, and pathways in cancer.Conclusion:We hope these 24 genes might be useful for diagnosis, prognosis and treatment for breast cancer.
Collapse
Affiliation(s)
- Cheng-Wen Yang
- Tianjin Key Lab of BME Measurement, Department of Biomedical Engineering, Tianjin University, Tianjin, China
| | - Huan-Huan Cao
- Tianjin Key Lab of BME Measurement, Department of Biomedical Engineering, Tianjin University, Tianjin, China
| | - Yu Guo
- Tianjin Key Lab of BME Measurement, Department of Biomedical Engineering, Tianjin University, Tianjin, China
| | - Yuan-Ming Feng
- Tianjin Key Lab of BME Measurement, Department of Biomedical Engineering, Tianjin University, Tianjin, China
| | - Ning Zhang
- Tianjin Key Lab of BME Measurement, Department of Biomedical Engineering, Tianjin University, Tianjin, China
| |
Collapse
|
24
|
Song KA, Faber AC. Epithelial-to-mesenchymal transition and drug resistance: transitioning away from death. J Thorac Dis 2019; 11:E82-E85. [PMID: 31372302 DOI: 10.21037/jtd.2019.06.11] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Kyung-A Song
- Philips Institute for Oral Health Research, VCU School of Dentistry and Massey Cancer Center, Richmond, VA, USA
| | - Anthony C Faber
- Philips Institute for Oral Health Research, VCU School of Dentistry and Massey Cancer Center, Richmond, VA, USA
| |
Collapse
|
25
|
Siles L, Ninfali C, Cortés M, Darling DS, Postigo A. ZEB1 protects skeletal muscle from damage and is required for its regeneration. Nat Commun 2019; 10:1364. [PMID: 30910999 PMCID: PMC6434033 DOI: 10.1038/s41467-019-08983-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 02/12/2019] [Indexed: 12/31/2022] Open
Abstract
The mechanisms linking muscle injury and regeneration are not fully understood. Here we report an unexpected role for ZEB1 regulating inflammatory and repair responses in dystrophic and acutely injured muscles. ZEB1 is upregulated in the undamaged and regenerating myofibers of injured muscles. Compared to wild-type counterparts, Zeb1-deficient injured muscles exhibit enhanced damage that corresponds with a retarded p38-MAPK-dependent transition of their macrophages towards an anti-inflammatory phenotype. Zeb1-deficient injured muscles also display a delayed and poorer regeneration that is accounted by the retarded anti-inflammatory macrophage transition and their intrinsically deficient muscle satellite cells (MuSCs). Macrophages in Zeb1-deficient injured muscles show lower phosphorylation of p38 and its forced activation reverts the enhanced muscle damage and poorer regeneration. MuSCs require ZEB1 to maintain their quiescence, prevent their premature activation following injury, and drive efficient regeneration in dystrophic muscles. These data indicate that ZEB1 protects muscle from damage and is required for its regeneration. Following muscle damage, an inflammatory response is associated to activation of satellite cells, which drive muscle repair. Here, the authors show that upregulation of Zeb1 in macrophages and muscle fibres regulates inflammation, and also show a role for Zeb1 in maintenance of satellite cell quiescence.
Collapse
Affiliation(s)
- Laura Siles
- Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, 08036, Barcelona, Spain
| | - Chiara Ninfali
- Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, 08036, Barcelona, Spain
| | - Marlies Cortés
- Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, 08036, Barcelona, Spain
| | - Douglas S Darling
- Center for Genetics and Molecular Medicine and Department of Immunology and Infectious Diseases, University of Louisville, Louisville, KY, 40202, USA
| | - Antonio Postigo
- Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, 08036, Barcelona, Spain. .,Molecular Targets Program, James G. Brown Cancer Center, Louisville, KY, 40202, USA. .,ICREA, Barcelona, 08010, Spain.
| |
Collapse
|
26
|
Al-Marsoummi S, Vomhof-DeKrey E, Basson MD. Schlafen12 Reduces the Aggressiveness of Triple Negative Breast Cancer through Post-Transcriptional Regulation of ZEB1 That Drives Stem Cell Differentiation. Cell Physiol Biochem 2019; 53:999-1014. [PMID: 31838790 PMCID: PMC7007982 DOI: 10.33594/000000191] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/10/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND/AIMS Schlafen12 (SLFN12) promotes human intestinal and prostatic epithelial differentiation. We sought to determine whether SLFN12 reduces triple-negative breast cancer (TNBC) aggressiveness. METHODS We validated bioinformatics analyses of publicly available databases by staining human TNBC. After virally overexpressing or siRNA-reducing SLFN12 in TNBC cell lines, we measured proliferation by CCK-8 assay, invasion into basement-membrane-coated pores, mRNA by q-RT-PCR and protein by Western blotting. Flow cytometry assessed proliferation and stem cell marker expression, and sorted CD44+/CD24- cells. Stemness was also assessed by mammosphere formation, and translation by click-it-AHA chemistry. RESULTS SLFN12 expression was lower in TNBC tumors and correlated with survival. SLFN12 overexpression reduced TNBC MDA-MB-231, BT549, and Hs578T proliferation. In MDA-MB-231 cells, AdSLFN12 reduced invasion, promoted cell cycle arrest, increased E-cadherin promoter activity, mRNA, and protein, and reduced vimentin expression and protein. SLFN12 knockdown increased vimentin. AdSLFN12 reduced the proportion of MDA-MB-231 CD44+CD24- cells, with parallel differentiation changes. SLFN12 overexpression reduced MDA-MB-231 mammosphere formation. SLFN12 overexpression decreased ZEB1 and Slug protein despite increased ZEB1 and Slug mRNA in all three lines. SLFN12 overexpression accelerated MDA-MB-231 ZEB1 proteasomal degradation and slowed ZEB1 translation. SLFN12 knockdown increased ZEB1 protein. Coexpressing ZEB1 attenuated the SLFN12 effect on E-cadherin mRNA and proliferation in all three lines. CONCLUSION SLFN12 may reduce TNBC aggressiveness and improve survival in part by a post-transcriptional decrease in ZEB1 that promotes TNBC cancer stem cell differentiation.
Collapse
Affiliation(s)
- Sarmad Al-Marsoummi
- Department of Biomedical Sciences, University of North Dakota School of Medicine and the Health Sciences, Grand Forks, ND, USA
| | - Emilie Vomhof-DeKrey
- Department of Biomedical Sciences, University of North Dakota School of Medicine and the Health Sciences, Grand Forks, ND, USA
- Department of Surgery, University of North Dakota School of Medicine and the Health Sciences, Grand Forks, ND, USA
| | - Marc D Basson
- Department of Biomedical Sciences, University of North Dakota School of Medicine and the Health Sciences, Grand Forks, ND, USA,
- Department of Surgery, University of North Dakota School of Medicine and the Health Sciences, Grand Forks, ND, USA
- Department of Pathology, University of North Dakota School of Medicine and the Health Sciences, Grand Forks, ND, USA
| |
Collapse
|
27
|
Neural Transcription Factors in Disease Progression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1210:437-462. [PMID: 31900920 DOI: 10.1007/978-3-030-32656-2_19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Progression to the malignant state is fundamentally dependent on transcriptional regulation in cancer cells. Optimum abundance of cell cycle proteins, angiogenesis factors, immune evasion markers, etc. is needed for proliferation, metastasis or resistance to treatment. Therefore, dysregulation of transcription factors can compromise the normal prostate transcriptional network and contribute to malignant disease progression.The androgen receptor (AR) is considered to be a key transcription factor in prostate cancer (PCa) development and progression. Consequently, androgen pathway inhibitors (APIs) are currently the mainstay in PCa treatment, especially in castration-resistant prostate cancer (CRPC). However, emerging evidence suggests that with increased administration of potent APIs, prostate cancer can progress to a highly aggressive disease that morphologically resembles small cell carcinoma, which is referred to as neuroendocrine prostate cancer (NEPC), treatment-induced or treatment-emergent small cell prostate cancer. This chapter will review how neuronal transcription factors play a part in inducing a plastic stage in prostate cancer cells that eventually progresses to a more aggressive state such as NEPC.
Collapse
|
28
|
Bozickovic O, Skartveit L, Engelsen AST, Helland T, Jonsdottir K, Flågeng MH, Fenne IS, Janssen E, Lorens JB, Bjørkhaug L, Sagen JV, Mellgren G. A novel SRC-2-dependent regulation of epithelial-mesenchymal transition in breast cancer cells. J Steroid Biochem Mol Biol 2019; 185:57-70. [PMID: 30048685 DOI: 10.1016/j.jsbmb.2018.07.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 07/01/2018] [Accepted: 07/16/2018] [Indexed: 12/11/2022]
Abstract
Steroid receptor coactivator 2 (SRC-2) is a nuclear receptor coactivator, important for the regulation of estrogen receptor alpha (ERα)-mediated transcriptional activity in breast cancer cells. However, the transcriptional role of SRC-2 in breast cancer is still ambiguous. Here we aimed to unravel a more precise transcriptional role of SRC-2 and uncover unique target genes in MCF-7 breast cancer cells, as opposed to the known oncogene SRC-3. Gene expression analyses of cells depleted of either SRC-2 or SRC-3 showed that they transcriptionally regulate mostly separate gene sets. However, individual unique gene sets were implicated in some of the same major gene ontology biological processes, such as cellular structure and development. This finding was supported by three-dimensional cell cultures, demonstrating that depletion of SRC-2 and SRC-3 changed the morphology of the cells into epithelial-like hollow acinar structures, indicating that both SRC proteins are involved in maintaining the hybrid E/M phenotype. In clinical ER-positive, HER2-negative breast cancer samples the expression of SRC-2 was negatively correlated with the expression of MCF-7-related luminal, cell cycle and cellular morphogenesis genes. Finally, elucidating SRC-2 unique transcriptional effects, we identified Lyn kinase (an EMT biomarker) to be upregulated exclusively after SRC-2 depletion. In conclusion, we show that both SRC-2 and SRC-3 are essential for the EMT in breast cancer cells, controlling different transcriptional niches.
Collapse
Affiliation(s)
- Olivera Bozickovic
- Department of Clinical Science, University of Bergen, N-5021 Bergen, Norway; Hormone Laboratory, Haukeland University Hospital, N-5021 Bergen, Norway; KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5021 Bergen, Norway.
| | - Linn Skartveit
- Hormone Laboratory, Haukeland University Hospital, N-5021 Bergen, Norway.
| | - Agnete S T Engelsen
- Centre for Cancer Biomarkers (CCBIO), Department of Biomedicine, University of Bergen, N-5009 Bergen, Norway.
| | - Thomas Helland
- Hormone Laboratory, Haukeland University Hospital, N-5021 Bergen, Norway.
| | - Kristin Jonsdottir
- Department of Pathology, Stavanger University Hospital, N-4068 Stavanger, Norway.
| | | | - Ingvild S Fenne
- Hormone Laboratory, Haukeland University Hospital, N-5021 Bergen, Norway.
| | - Emiel Janssen
- Department of Mathematics and Natural Sciences, University of Stavanger, N-4036 Stavanger, Norway.
| | - James B Lorens
- Centre for Cancer Biomarkers (CCBIO), Department of Biomedicine, University of Bergen, N-5009 Bergen, Norway.
| | - Lise Bjørkhaug
- Hormone Laboratory, Haukeland University Hospital, N-5021 Bergen, Norway; KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5021 Bergen, Norway; Department of Biomedical Laboratory Sciences, Western Norway University of Applied Sciences, N-5020 Bergen, Norway.
| | - Jørn V Sagen
- Department of Clinical Science, University of Bergen, N-5021 Bergen, Norway; Hormone Laboratory, Haukeland University Hospital, N-5021 Bergen, Norway; KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5021 Bergen, Norway.
| | - Gunnar Mellgren
- Department of Clinical Science, University of Bergen, N-5021 Bergen, Norway; Hormone Laboratory, Haukeland University Hospital, N-5021 Bergen, Norway; KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5021 Bergen, Norway.
| |
Collapse
|
29
|
Effect of AR antagonist combined with PARP1 inhibitor on sporadic triple-negative breast cancer bearing AR expression and methylation-mediated BRCA1 dysfunction. Biomed Pharmacother 2018; 111:169-177. [PMID: 30580238 DOI: 10.1016/j.biopha.2018.11.136] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 11/22/2018] [Accepted: 11/27/2018] [Indexed: 11/23/2022] Open
Abstract
Triple-negative breast cancer (TNBC) patients usually present worse clinical outcomes due to their high heterogeneity. The purpose of our study is to investigate the prognostic role of AR and BRCA1 expression in sporadic TNBC patients, and effect of AR blockade and PARP1 inhibitor for TNBC patients who characterized by positive-AR expression and BRCA1 inactivation or dysfunction. In our present study, we found that AR is expressed in 43.6% and 34.0% of TNBC tissues, when 1% or 10% staining was used as the threshold for AR positivity, respectively. When 1% staining was used as the threshold, AR expression indicates a poor disease-free survival (DFS) of TNBC patients. TNBC patients with negative BRCA1 show a poor DFS, and BRCA1 suppression is associated with the methylation status of its promoter. Interestingly, BRCA1-/AR + TNBC patients have shorter DFS than other TNBC patients regardless of the threshold for AR positivity. AR antagonists MDV3100 enhances the PARP1 inhibitor Olaparib-mediated decrease of cell viability in AR-positive/BRCA1-inactivated cells in vitro and in vivo. Our results suggested that combination of AR blockade and PARP1 inhibitor may be a potential strategy for sporadic TNBC patients who characterized by positive-AR expression and BRCA1 inactivation or dysfunction.
Collapse
|
30
|
Klec C, Prinz F, Pichler M. Involvement of the long noncoding RNA NEAT1 in carcinogenesis. Mol Oncol 2018; 13:46-60. [PMID: 30430751 PMCID: PMC6322192 DOI: 10.1002/1878-0261.12404] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 10/25/2018] [Accepted: 11/07/2018] [Indexed: 12/14/2022] Open
Abstract
Altered expression levels of the long noncoding RNA (lncRNA) nuclear‐enriched abundant transcript 1 (NEAT1) have been reported in different types of cancer. More than half of the NEAT1 studies in cancer have been published within the last 2 years. In this review, we discuss very recent developments and insights into NEAT1 contribution to carcinogenesis. Summarizing the literature, it becomes obvious that NEAT1 is a lncRNA highly de‐/upregulated in a variety of cancer entities, in which it primarily acts as a competing endogenous RNA (ceRNA) which sponges tumor‐suppressive microRNA (miRNA). The sponged miRNA lose their ability to degrade, silence, or hamper translation of their downstream—mostly oncogenic—target transcripts, ultimately promoting carcinogenesis. This role of NEAT1 function in tumorigenesis suggests it may be a prognostic biomarker as well as potential therapeutic target, pending the completion of further studies into the underlying mechanisms.
Collapse
Affiliation(s)
- Christiane Klec
- Division of Oncology, Department of Internal Medicine, Medical University of Graz (MUG), Austria.,Research Unit for Non-coding RNAs and Genome Editing, Medical University of Graz (MUG), Austria
| | - Felix Prinz
- Division of Oncology, Department of Internal Medicine, Medical University of Graz (MUG), Austria.,Research Unit for Non-coding RNAs and Genome Editing, Medical University of Graz (MUG), Austria
| | - Martin Pichler
- Division of Oncology, Department of Internal Medicine, Medical University of Graz (MUG), Austria.,Research Unit for Non-coding RNAs and Genome Editing, Medical University of Graz (MUG), Austria.,Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
31
|
Shi Y, Yang F, Huang D, Guan X. Androgen blockade based clinical trials landscape in triple negative breast cancer. Biochim Biophys Acta Rev Cancer 2018; 1870:283-290. [DOI: 10.1016/j.bbcan.2018.05.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 05/22/2018] [Accepted: 05/22/2018] [Indexed: 01/12/2023]
|
32
|
Al-Othman N, Hammad H, Ahram M. Dihydrotestosterone regulates expression of CD44 via miR-328-3p in triple-negative breast cancer cells. Gene 2018; 675:128-135. [DOI: 10.1016/j.gene.2018.06.094] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 06/14/2018] [Accepted: 06/27/2018] [Indexed: 12/17/2022]
|
33
|
|
34
|
Androgen receptor in triple negative breast cancer: A potential target for the targetless subtype. Cancer Treat Rev 2018; 68:102-110. [PMID: 29940524 DOI: 10.1016/j.ctrv.2018.06.005] [Citation(s) in RCA: 171] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 06/08/2018] [Accepted: 06/09/2018] [Indexed: 12/13/2022]
Abstract
Triple negative breast cancer (TNBC) represents the 15-20% of all breast cancers (BC) and is characterized by a very aggressive behavior. Recent data suggest that TNBC is not a single disease, but it is rather an umbrella for different ontology-profiles such as basal like 1 and 2, mesenchymal, and the luminal androgen receptor (LAR). The LAR subtype is characterized by the expression of the Androgen Receptor (AR) and its downstream effects. Notwithstanding the role of the AR in several signaling pathways, its impact on a biological and clinical standpoint is still controversial. The LAR subtype has been associated with better prognosis, less chemotherapy responsiveness and lower pathologic complete response after neoadjuvant treatment. Clinical evidence suggests a role for anti-androgen therapies such as bicalutamide, enzalutamide and abiraterone, offering an interesting chemo-free alternative for chemo-unresponsive patients, and therefore potentially shifting current treatment strategies.
Collapse
|
35
|
Hunter I, Hay CW, Esswein B, Watt K, McEwan IJ. Tissue control of androgen action: The ups and downs of androgen receptor expression. Mol Cell Endocrinol 2018; 465:27-35. [PMID: 28789969 DOI: 10.1016/j.mce.2017.08.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 07/28/2017] [Accepted: 08/03/2017] [Indexed: 12/17/2022]
Abstract
The hormone testosterone plays crucial roles during male development and puberty and throughout life, as an anabolic regulator of muscle and bone structure and function. The actions of testosterone are mediated, primarily, through the androgen receptor, a member of the nuclear receptor superfamily. The androgen receptor gene is located on the X-chromosome and receptor levels are tightly controlled both at the level of transcription of the gene and post-translationally at the protein level. Sp1 has emerged as the major driver of expression of the androgen receptor gene, while auto-regulation by androgens is associated with both positive and negative regulation in a possible cell-selective manner. Research into the networks of positive and negative regulators of the androgen receptor gene are vital in order to understand the temporal and spatial control of receptor levels and the consequences for healthy aging and disease. A clear understanding of the multiple transcription factors participating in regulation of the androgen receptor gene will likely aid in the development and application of hormone therapies to boast or curb receptor activity.
Collapse
Affiliation(s)
- Irene Hunter
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, Scotland, UK
| | - Colin W Hay
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, Scotland, UK
| | - Bianca Esswein
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, Scotland, UK; Friedrich-Schiller-Universitat, Jena, Germany
| | - Kate Watt
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, Scotland, UK
| | - Iain J McEwan
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, Scotland, UK.
| |
Collapse
|
36
|
West DC, Kocherginsky M, Tonsing-Carter EY, Dolcen DN, Hosfield DJ, Lastra RR, Sinnwell JP, Thompson KJ, Bowie KR, Harkless RV, Skor MN, Pierce CF, Styke SC, Kim CR, de Wet L, Greene GL, Boughey JC, Goetz MP, Kalari KR, Wang L, Fleming GF, Györffy B, Conzen SD. Discovery of a Glucocorticoid Receptor (GR) Activity Signature Using Selective GR Antagonism in ER-Negative Breast Cancer. Clin Cancer Res 2018; 24:3433-3446. [PMID: 29636357 DOI: 10.1158/1078-0432.ccr-17-2793] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 02/14/2018] [Accepted: 04/04/2018] [Indexed: 12/30/2022]
Abstract
Purpose: Although high glucocorticoid receptor (GR) expression in early-stage estrogen receptor (ER)-negative breast cancer is associated with shortened relapse-free survival (RFS), how associated GR transcriptional activity contributes to aggressive breast cancer behavior is not well understood. Using potent GR antagonists and primary tumor gene expression data, we sought to identify a tumor-relevant gene signature based on GR activity that would be more predictive than GR expression alone.Experimental Design: Global gene expression and GR ChIP-sequencing were performed to identify GR-regulated genes inhibited by two chemically distinct GR antagonists, mifepristone and CORT108297. Differentially expressed genes from MDA-MB-231 cells were cross-evaluated with significantly expressed genes in GR-high versus GR-low ER-negative primary breast cancers. The resulting subset of GR-targeted genes was analyzed in two independent ER-negative breast cancer cohorts to derive and then validate the GR activity signature (GRsig).Results: Gene expression pathway analysis of glucocorticoid-regulated genes (inhibited by GR antagonism) revealed cell survival and invasion functions. GR ChIP-seq analysis demonstrated that GR antagonists decreased GR chromatin association for a subset of genes. A GRsig that comprised n = 74 GR activation-associated genes (also reversed by GR antagonists) was derived from an adjuvant chemotherapy-treated Discovery cohort and found to predict probability of relapse in a separate Validation cohort (HR = 1.9; P = 0.012).Conclusions: The GRsig discovered herein identifies high-risk ER-negative/GR-positive breast cancers most likely to relapse despite administration of adjuvant chemotherapy. Because GR antagonism can reverse expression of these genes, we propose that addition of a GR antagonist to chemotherapy may improve outcome for these high-risk patients. Clin Cancer Res; 24(14); 3433-46. ©2018 AACR.
Collapse
Affiliation(s)
- Diana C West
- Department of Medicine, The University of Chicago, Chicago, Illinois.,Department of Chemistry and Physics, Ave Maria University, Ave Maria, Florida
| | - Masha Kocherginsky
- Department of Preventive Medicine, Northwestern University, Chicago, Illinois
| | | | - D Nesli Dolcen
- Department of Medicine, The University of Chicago, Chicago, Illinois
| | - David J Hosfield
- Ben May Department for Cancer Research, The University of Chicago, Chicago, Illinois
| | - Ricardo R Lastra
- Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Jason P Sinnwell
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Kevin J Thompson
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Kathleen R Bowie
- Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Ryan V Harkless
- Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Maxwell N Skor
- Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Charles F Pierce
- Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Sarah C Styke
- Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Caroline R Kim
- Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Larischa de Wet
- Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Geoffrey L Greene
- Ben May Department for Cancer Research, The University of Chicago, Chicago, Illinois
| | - Judy C Boughey
- Department of Surgery, Mayo Clinic, Rochester, Minnesota
| | - Matthew P Goetz
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota.,Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | - Krishna R Kalari
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Gini F Fleming
- Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Balázs Györffy
- MTA-TTK Lendület Cancer Biomarker Research Group, Institute of Enzymology, Budapest, Hungary.,Semmelweis University, Second Department of Pediatrics, Budapest, Hungary
| | - Suzanne D Conzen
- Department of Medicine, The University of Chicago, Chicago, Illinois. .,Ben May Department for Cancer Research, The University of Chicago, Chicago, Illinois
| |
Collapse
|
37
|
Gene regulatory pattern analysis reveals essential role of core transcriptional factors' activation in triple-negative breast cancer. Oncotarget 2017; 8:21938-21953. [PMID: 28423538 PMCID: PMC5400636 DOI: 10.18632/oncotarget.15749] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 01/10/2017] [Indexed: 12/31/2022] Open
Abstract
Background Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype. Genome-scale molecular characteristics and regulatory mechanisms that distinguish TNBC from other subtypes remain incompletely characterized. Results By combining gene expression analysis and PANDA network, we defined three different TF regulatory patterns. A core TNBC-Specific TF Activation Driven Pattern (TNBCac) was specifically identified in TNBC by computational analysis. The essentialness of core TFs (ZEB1, MZF1, SOX10) in TNBC was highlighted and validated by cell proliferation analysis. Furthermore, 13 out of 35 co-targeted genes were also validated to be targeted by ZEB1, MZF1 and SOX10 in TNBC cell lines by real-time quantitative PCR. In three breast cancer cohorts, non-TNBC patients could be stratified into two subgroups by the 35 co-targeted genes along with 5 TFs, and the subgroup that more resembled TNBC had a worse prognosis. Methods We constructed gene regulatory networks in breast cancer by Passing Attributes between Networks for Data Assimilation (PANDA). Co-regulatory modules were specifically identified in TNBC by computational analysis, while the essentialness of core translational factors (TF) in TNBC was highlighted and validated by in vitro experiments. Prognostic effects of different factors were measured by Log-rank test and displayed by Kaplan-Meier plots. Conclusions We identified a core co-regulatory module specifically existing in TNBC, which enabled subtype re-classification and provided a biologically feasible view of breast cancer.
Collapse
|
38
|
Ahram M, Mustafa E, Zaza R, Abu Hammad S, Alhudhud M, Bawadi R, Zihlif M. Differential expression and androgen regulation of microRNAs and metalloprotease 13 in breast cancer cells. Cell Biol Int 2017; 41:1345-1355. [PMID: 28816390 DOI: 10.1002/cbin.10841] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Accepted: 08/14/2017] [Indexed: 01/11/2023]
Abstract
MicroRNA molecules (miRNAs) play important roles in regulating cell behavior. The expression of certain miRNAs has been shown to be regulated by the androgen receptor (AR), which seems to have a critical role in the tumorigenic process of breast cancer. The differential expression of 84 miRNAs was first examined in three breast cancer cell lines: the luminal MCF-7 and T47D cells and the molecular apocrine MDA-MB-453 cells. Analysis of basal expression of miRNAs revealed that each cell line had distinct miRNA expression where let-7a and -7b were markers of MDA-MB-453 cells, whereas miR-205 was a marker for the luminal cell lines. Treating the cells with the AR agonist, CI-4AS-1, resulted in unique alterations in the expression of specific miRNA among the three cell lines. Particularly, the expression of miR-100 and miR-125 was reduced in MDA-MB-453 cells by five and three folds, respectively. This effect was simultaneous with AR-induced increase in the expression and extracellular release of metalloprotease-13 (MMP13). Transfection of cells with either miR-100 or miR-125b negated the induction of MMP13 release. Additionally, AR activation induced a morphological alteration of MDA-MB-453 cells, which was blocked by miR-125b only. Collectively, these data indicate that AR may control the biological behavior of breast cancer cells and protein expression via miRNAs.
Collapse
Affiliation(s)
- Mamoun Ahram
- School of Medicine, Department of Physiology and Biochemistry, The University of Jordan, Amman, Jordan
| | - Ebtihal Mustafa
- School of Medicine, Department of Physiology and Biochemistry, The University of Jordan, Amman, Jordan
| | - Rand Zaza
- School of Medicine, Department of Physiology and Biochemistry, The University of Jordan, Amman, Jordan
| | - Shatha Abu Hammad
- School of Medicine, Department of Physiology and Biochemistry, The University of Jordan, Amman, Jordan
| | - Mariam Alhudhud
- School of Medicine, Department of Physiology and Biochemistry, The University of Jordan, Amman, Jordan
| | - Randa Bawadi
- School of Medicine, Department of Physiology and Biochemistry, The University of Jordan, Amman, Jordan
| | - Malek Zihlif
- School of Medicine, Department of Pharmacology, The University of Jordan, Amman, Jordan
| |
Collapse
|
39
|
Song W, Tang L, Xu Y, Sun Q, Yang F, Guan X. ERβ1 inhibits metastasis of androgen receptor-positive triple-negative breast cancer by suppressing ZEB1. J Exp Clin Cancer Res 2017; 36:75. [PMID: 28583190 PMCID: PMC5460479 DOI: 10.1186/s13046-017-0545-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 05/29/2017] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Increasing evidence has indicated an important role for estrogen receptor beta 1 (ERβ1) in breast cancer. However, the role of ERβ1 in the metastasis of androgen receptor (AR)-positive triple-negative breast cancer (TNBC) and the underlying mechanisms are still unknown. METHODS Stable ERβ1-expressing TNBC cell lines were generated for this study. We detected the abilities of cell migration and invasion by wound-healing and transwell assays and the expression of E-cadherin and N-cadherin by quantitative RT-PCR (qRT-PCR) and western blotting assays in TNBC cell lines. Chromatin immunoprecipitation (ChIP) analysis was performed to assess the effect of AR on ERβ1 promoter. Tumor metastasis was evaluated in vivo using a lung metastasis mouse model. Lastly, immunohistochemical expression of ERβ1 in TNBC tissues was analyzed and correlated with clinicopathological features. RESULTS ERβ1 suppressed the invasion and migration abilities of AR-positive TNBC cells and induced the downregulation of ZEB1. ZEB1 overexpression abrogated the increase in E-cadherin expression and the decrease in N-cadherin expression modulated by ERβ1. A lung metastasis mouse model showed that the incidence of metastasis was lower in ERβ1-expressing TNBC cells. Further, AR activation increased the anti-metastatic effect of ERβ1 in AR-positive TNBC cells, which accelerated ERβ1 transcription by functioning as a transcription factor that bound to the promoter of ERβ1. No significant change was observed in AR expression induced by ERβ1. Immunohistochemistry (IHC) analysis of TNBC clinical samples showed that ERβ1 and AR were positive in 31.7% and 23.2% of samples, respectively. ERβ1 expression was negatively correlated with ZEB1 expression and lymph node metastasis, and positively correlated with the expression of AR and E-cadherin. CONCLUSION Our findings suggested a potential role of ERβ1 in metastasis of AR-positive TNBC and provided novel insights into the mechanism of action of ERβ1 and the possible relationship between ERβ1 and AR.
Collapse
Affiliation(s)
- Wei Song
- Department of Medical Oncology, Jinling Hospital, Southern Medical University, Guangzhou, 510515 China
| | - Lin Tang
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002 China
| | - Yumei Xu
- Department of Medical Oncology, Jinling Hospital, Southern Medical University, Guangzhou, 510515 China
| | - Qian Sun
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002 China
| | - Fang Yang
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002 China
| | - Xiaoxiang Guan
- Department of Medical Oncology, Jinling Hospital, Southern Medical University, Guangzhou, 510515 China
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002 China
- Department of Medical Oncology, Jinling Hospital, Nanjing Medical University, Nanjing, 210029 China
| |
Collapse
|
40
|
AR Signaling in Breast Cancer. Cancers (Basel) 2017; 9:cancers9030021. [PMID: 28245550 PMCID: PMC5366816 DOI: 10.3390/cancers9030021] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 02/13/2017] [Accepted: 02/18/2017] [Indexed: 12/31/2022] Open
Abstract
Androgen receptor (AR, a member of the steroid hormone receptor family) status has become increasingly important as both a prognostic marker and potential therapeutic target in breast cancer. AR is expressed in up to 90% of estrogen receptor (ER) positive breast cancer, and to a lesser degree, human epidermal growth factor 2 (HER2) amplified tumors. In the former, AR signaling has been correlated with a better prognosis given its inhibitory activity in estrogen dependent disease, though conversely has also been shown to increase resistance to anti-estrogen therapies such as tamoxifen. AR blockade can mitigate this resistance, and thus serves as a potential target in ER-positive breast cancer. In HER2 amplified breast cancer, studies are somewhat conflicting, though most show either no effect or are associated with poorer survival. Much of the available data on AR signaling is in triple-negative breast cancer (TNBC), which is an aggressive disease with inferior outcomes comparative to other breast cancer subtypes. At present, there are no approved targeted therapies in TNBC, making study of the AR signaling pathway compelling. Gene expression profiling studies have also identified a luminal androgen receptor (LAR) subtype that is dependent on AR signaling in TNBC. Regardless, there seems to be an association between AR expression and improved outcomes in TNBC. Despite lower pathologic complete response (pCR) rates with neoadjuvant therapy, patients with AR-expressing TNBC have been shown to have a better prognosis than those that are AR-negative. Clinical studies targeting AR have shown somewhat promising results. In this paper we review the literature on the biology of AR in breast cancer and its prognostic and predictive roles. We also present our thoughts on therapeutic strategies.
Collapse
|
41
|
Ware KE, Somarelli JA, Schaeffer D, Li J, Zhang T, Park S, Patierno SR, Freedman J, Foo WC, Garcia MA, Armstrong AJ. Snail promotes resistance to enzalutamide through regulation of androgen receptor activity in prostate cancer. Oncotarget 2016; 7:50507-50521. [PMID: 27409172 PMCID: PMC5226599 DOI: 10.18632/oncotarget.10476] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 06/20/2016] [Indexed: 11/25/2022] Open
Abstract
Treatment with androgen-targeted therapies can induce upregulation of epithelial plasticity pathways. Epithelial plasticity is known to be important for metastatic dissemination and therapeutic resistance. The goal of this study is to elucidate the functional consequence of induced epithelial plasticity on AR regulation during disease progression to identify factors important for treatment-resistant and metastatic prostate cancer. We pinpoint the epithelial plasticity transcription factor, Snail, at the nexus of enzalutamide resistance and prostate cancer metastasis both in preclinical models of prostate cancer and in patients. In patients, Snail expression is associated with Gleason 9-10 high-risk disease and is strongly overexpressed in metastases as compared to localized prostate cancer. Snail expression is also elevated in enzalutamide-resistant prostate cancer cells compared to enzalutamide-sensitive cells, and downregulation of Snail re-sensitizes enzalutamide-resistant cells to enzalutamide. While activation of Snail increases migration and invasion, it is also capable of promoting enzalutamide resistance in enzalutamide-sensitive cells. This Snail-mediated enzalutamide resistance is a consequence of increased full-length AR and AR-V7 expression and nuclear localization. Downregulation of either full-length AR or AR-V7 re-sensitizes cells to enzalutamide in the presence of Snail, thus connecting Snail-induced enzalutamide resistance directly to AR biology. Finally, we demonstrate that Snail is capable of mediating-resistance through AR even in the absence of AR-V7. These findings imply that increased Snail expression during progression to metastatic disease may prime cells for resistance to AR-targeted therapies by promoting AR activity in prostate cancer.
Collapse
Affiliation(s)
- Kathryn E. Ware
- Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, NC, USA
- Department of Genitourinary Oncology, Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - Jason A. Somarelli
- Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, NC, USA
- Department of Genitourinary Oncology, Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - Daneen Schaeffer
- Department of Oncology, Translational Research, Janssen Research and Development, Spring House, PA, USA
| | - Jing Li
- Department of Hematology and Oncology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Tian Zhang
- Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, NC, USA
- Department of Genitourinary Oncology, Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - Sally Park
- Department of Genitourinary Oncology, Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - Steven R. Patierno
- Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, NC, USA
- Department of Genitourinary Oncology, Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - Jennifer Freedman
- Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, NC, USA
- Department of Genitourinary Oncology, Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - Wen-Chi Foo
- Department of Pathology, Duke University, Durham, NC, USA
| | - Mariano A. Garcia
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Andrew J. Armstrong
- Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, NC, USA
- Department of Genitourinary Oncology, Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| |
Collapse
|
42
|
Asano Y, Kashiwagi S, Onoda N, Kurata K, Morisaki T, Noda S, Takashima T, Ohsawa M, Kitagawa S, Hirakawa K. Clinical verification of sensitivity to preoperative chemotherapy in cases of androgen receptor-expressing positive breast cancer. Br J Cancer 2016; 114:14-20. [PMID: 26757422 PMCID: PMC4716546 DOI: 10.1038/bjc.2015.434] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Revised: 09/16/2015] [Accepted: 11/06/2015] [Indexed: 02/05/2023] Open
Abstract
Background: Triple-negative breast cancer (TNBC) patients testing positive for androgen receptor (AR) expression are thought to be chemotherapy resistant, similar to other hormone receptor-positive breast cancers; however, this has not been substantially validated in the clinic. In this study, we investigated the association between chemotherapy sensitivity and AR expression in patients treated with neoadjuvant chemotherapy (NAC) using standardised chemotherapy criteria and regimens. Methods: A total of 177 patients with resectable early-stage breast cancer were treated with NAC. Oestrogen receptor, progesterone receptor, HER2, Ki67 and AR status were assessed immunohistochemically. Results: Sixty-one patients were diagnosed with TNBC; AR expression was identified in 23 (37.7%), which was significantly less common than that found in non-TNBC patients (103 of 116; 88.8% P<0.001). The rate of pathological complete response after NAC was significantly lower (P=0.001), and disease recurrence was more common (P=0.008) in patients with AR-positive compared with those with AR-negative TNBC. In TNBC cases, as expected, the non-recurrence period in cases that were negative for AR expression was significantly extended (P=0.006, log-rank). Conclusions: Androgen receptor expressions may be useful as biomarkers to predict treatment responses to NAC in TNBC. Moreover, induction of a change in subtype to the AR-negative phenotype was observed after NAC.
Collapse
Affiliation(s)
- Yuka Asano
- Department of Surgical Oncology, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Japan
| | - Shinichiro Kashiwagi
- Department of Surgical Oncology, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Japan
| | - Naoyoshi Onoda
- Department of Surgical Oncology, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Japan
| | - Kento Kurata
- Department of Surgical Oncology, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Japan
| | - Tamami Morisaki
- Department of Surgical Oncology, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Japan
| | - Satoru Noda
- Department of Surgical Oncology, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Japan
| | - Tsutomu Takashima
- Department of Surgical Oncology, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Japan
| | - Masahiko Ohsawa
- Department of Diagnostic Pathology, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Japan
| | - Seiichi Kitagawa
- Department of Physiology, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Japan
| | - Kosei Hirakawa
- Department of Surgical Oncology, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Japan
| |
Collapse
|
43
|
Mooney SM, Parsana P, Hernandez JR, Liu X, Verdone JE, Torga G, Harberg CA, Pienta KJ. The presence of androgen receptor elements regulates ZEB1 expression in the absence of androgen receptor. J Cell Biochem 2016; 116:115-23. [PMID: 25160502 DOI: 10.1002/jcb.24948] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 08/22/2014] [Indexed: 01/17/2023]
Abstract
Zinc finger E-box binding homeobox 1 (ZEB1) is a transcription factor that plays a central role in the epithelial to mesenchymal transition (EMT) of cancer cell lines. Studies on its regulation have mostly focused on the negative 3'UTR binding of miR200c. Interestingly, it has been previously reported that androgen receptor (AR) regulates ZEB1 expression in breast and prostate cancers. In order to validate this, various ZEB1 promoter deletions were cloned into a luciferase reporter system to elucidate the contribution of two putative androgen response elements (AREs). The in vivo contribution of AR was also assessed in cell lines after R1881 treatment using qPCR with prostate specific antigen (PSA) as the positive control. We discovered that AR upregulates the levels of expression of ZEB1 10-fold on a luciferase promoter that only contains the distal ARE. However, when the proximal ARE is included, no additional activation is apparent with AR or its hormone independent variant, AR-V7. Furthermore, we demonstrate here that a promoter construct containing both AREs activates transcription of ZEB1 even in the AR-null cell lines DU145 and PC3. Incubation of the AR-positive cell line, LNCaP with R1881, failed to substantially increase the expression levels of ZEB1. Despite the presence of AREs in the promoter region, it appears that ZEB1 expression can be induced even without AR. In addition, the region around the distal ARE is a potent repressor in AR-null cell lines.
Collapse
Affiliation(s)
- Steven M Mooney
- The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Voutsadakis IA. Epithelial-Mesenchymal Transition (EMT) and Regulation of EMT Factors by Steroid Nuclear Receptors in Breast Cancer: A Review and in Silico Investigation. J Clin Med 2016; 5:E11. [PMID: 26797644 PMCID: PMC4730136 DOI: 10.3390/jcm5010011] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 12/23/2015] [Accepted: 12/30/2015] [Indexed: 12/20/2022] Open
Abstract
Steroid Nuclear Receptors (SNRs) are transcription factors of the nuclear receptor super-family. Estrogen Receptor (ERα) is the best-studied and has a seminal role in the clinic both as a prognostic marker but also as a predictor of response to anti-estrogenic therapies. Progesterone Receptor (PR) is also used in the clinic but with a more debatable prognostic role and the role of the four other SNRs, ERβ, Androgen Receptor (AR), Glucocorticoid Receptor (GR) and Mineralocorticoid Receptor (MR), is starting only to be appreciated. ERα, but also to a certain degree the other SNRs, have been reported to be involved in virtually every cancer-enabling process, both promoting and impeding carcinogenesis. Epithelial-Mesenchymal Transition (EMT) and the reverse Mesenchymal Epithelial Transition (MET) are such carcinogenesis-enabling processes with important roles in invasion and metastasis initiation but also establishment of tumor in the metastatic site. EMT is governed by several signal transduction pathways culminating in core transcription factors of the process, such as Snail, Slug, ZEB1 and ZEB2, and Twist, among others. This paper will discuss direct regulation of these core transcription factors by SNRs in breast cancer. Interrogation of publicly available databases for binding sites of SNRs on promoters of core EMT factors will also be included in an attempt to fill gaps where other experimental data are not available.
Collapse
Affiliation(s)
- Ioannis A Voutsadakis
- Division of Medical Oncology, Department of Internal Medicine, Sault Area Hospital, Sault Ste Marie, ON P6B 0A8, Canada.
- Division of Clinical Sciences, Northern Ontario School of Medicine, Sudbury, QC P3E 2C6, Canada.
| |
Collapse
|
45
|
Taliaferro-Smith L, Oberlick E, Liu T, McGlothen T, Alcaide T, Tobin R, Donnelly S, Commander R, Kline E, Nagaraju GP, Havel L, Marcus A, Nahta R, O'Regan R. FAK activation is required for IGF1R-mediated regulation of EMT, migration, and invasion in mesenchymal triple negative breast cancer cells. Oncotarget 2016; 6:4757-72. [PMID: 25749031 PMCID: PMC4467113 DOI: 10.18632/oncotarget.3023] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Accepted: 12/31/2014] [Indexed: 11/25/2022] Open
Abstract
Triple negative breast cancer (TNBC) is a highly metastatic disease that currently lacks effective prevention and treatment strategies. The insulin-like growth factor 1 receptor (IGF1R) and focal adhesion kinase (FAK) signaling pathways function in numerous developmental processes, and alterations in both are linked with a number of common pathological diseases. Overexpression of IGF1R and FAK are closely associated with metastatic breast tumors. The present study investigated the interrelationship between IGF1R and FAK signaling in regulating the malignant properties of TNBC cells. Using small hairpin RNA (shRNA)-mediated IGF1R silencing methods, we showed that IGF1R is essential for sustaining mesenchymal morphologies of TNBC cells and modulates the expression of EMT-related markers. We further showed that IGF1R overexpression promotes migratory and invasive behaviors of TNBC cell lines. Most importantly, IGF1R-driven migration and invasion is predominantly mediated by FAK activation and can be suppressed using pharmacological inhibitors of FAK. Our findings in TNBC cells demonstrate a novel role of the IGF1R/FAK signaling pathway in regulating critical processes involved in the metastatic cascade. These results may improve the current understanding of the basic molecular mechanisms of TNBC metastasis and provide a strong rationale for co-targeting of IGF1R and FAK as therapy for mesenchymal TNBCs.
Collapse
Affiliation(s)
- LaTonia Taliaferro-Smith
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322 USA
| | - Elaine Oberlick
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322 USA.,Graduate Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA, 02115 USA
| | - Tongrui Liu
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322 USA
| | - Tanisha McGlothen
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322 USA
| | - Tiffanie Alcaide
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322 USA
| | - Rachel Tobin
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322 USA
| | - Siobhan Donnelly
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, 30322 USA
| | - Rachel Commander
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, 30322 USA
| | - Erik Kline
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322 USA
| | - Ganji Purnachandra Nagaraju
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322 USA
| | - Lauren Havel
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322 USA
| | - Adam Marcus
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322 USA
| | - Rita Nahta
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, 30322 USA
| | - Ruth O'Regan
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322 USA.,Georgia Cancer Center for Excellence at Grady Memorial Hospital, Atlanta, GA, 30303 USA
| |
Collapse
|
46
|
Broster SA, Kyprianou N. Epithelial–mesenchymal transition in prostatic disease. Future Oncol 2015; 11:3197-206. [DOI: 10.2217/fon.15.253] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
A fully differentiated epithelium of the normal prostate gland allows epithelial cells to de-differentiate into mesenchymal-like derivatives via the process of epithelial–mesenchymal transition (EMT) and redifferentiate via the reverse process, mesenchymal–epithelial transition. This review discusses the phenotypic changes associated with EMT and its programming in the development of the two growth disorders of the aging prostate gland, benign prostatic hyperplasia and prostate adenocarcinoma. Considering the cellular heterogeneity that characterizes both conditions, identifying the transcriptional programming of the phenotypic framework defining EMT and its reverse process mesenchymal–epithelial transition in their pathological landscape will enable novel platforms for biomarker-driven therapeutics and their implementation in benign prostatic hyperplasia and prostate cancer.
Collapse
Affiliation(s)
- Seth A Broster
- Departments of Urology, Molecular Biochemistry, Pathology, Toxicology & Cancer Biology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Natasha Kyprianou
- Departments of Urology, Molecular Biochemistry, Pathology, Toxicology & Cancer Biology, University of Kentucky College of Medicine, Lexington, KY, USA
| |
Collapse
|
47
|
Barton VN, D'Amato NC, Gordon MA, Christenson JL, Elias A, Richer JK. Androgen Receptor Biology in Triple Negative Breast Cancer: a Case for Classification as AR+ or Quadruple Negative Disease. Discov Oncol 2015. [PMID: 26201402 DOI: 10.1007/s12672-015-0232-3] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Triple negative breast cancer (TNBC) is an aggressive breast cancer subtype that lacks estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 (HER2) amplification. Due to the absence of these receptors, TNBC does not respond to traditional endocrine or HER2-targeted therapies that improve patient prognosis in other breast cancer subtypes. TNBC has a poor prognosis, and currently, there are no effective targeted therapies. Some TNBC tumors express androgen receptor (AR) and may benefit from AR-targeted therapies. Here, we review the literature on AR in TNBC and propose that TNBC be further sub-classified as either AR+ TNBC or quadruple negative breast cancer since targeting AR may represent a viable therapeutic option for a subset of TNBC.
Collapse
Affiliation(s)
- Valerie N Barton
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora CO, RC1 North P18-5127 Mail Stop 8104, 12800 E. 19th Ave, Aurora, CO, 80015, USA
| | - Nicholas C D'Amato
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora CO, RC1 North P18-5127 Mail Stop 8104, 12800 E. 19th Ave, Aurora, CO, 80015, USA
| | - Michael A Gordon
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora CO, RC1 North P18-5127 Mail Stop 8104, 12800 E. 19th Ave, Aurora, CO, 80015, USA
| | - Jessica L Christenson
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora CO, RC1 North P18-5127 Mail Stop 8104, 12800 E. 19th Ave, Aurora, CO, 80015, USA
| | - Anthony Elias
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora CO, RC1 North P18-5127 Mail Stop 8104, 12800 E. 19th Ave, Aurora, CO, 80015, USA
| | - Jennifer K Richer
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora CO, RC1 North P18-5127 Mail Stop 8104, 12800 E. 19th Ave, Aurora, CO, 80015, USA.
| |
Collapse
|
48
|
Lu YJ, Liu YC, Lin MC, Chen YT, Lin LY. Coordinative modulation of human zinc transporter 2 gene expression through active and suppressive regulators. J Nutr Biochem 2015; 26:351-9. [PMID: 25618524 DOI: 10.1016/j.jnutbio.2014.11.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 10/29/2014] [Accepted: 11/17/2014] [Indexed: 12/31/2022]
Abstract
Zinc transporter 2 (ZnT2) is one of the cellular factors responsible for Zn homeostasis. Upon Zn overload, ZnT2 reduces cellular Zn by transporting it into excretory vesicles. We investigated the molecular mechanism that regulates human ZnT2 (hZnT2) gene expression. Zn induces hZnT2 expression in dose- and time-dependent manners. Overexpression of metal-responsive transcription factor 1 (MTF-1) increases hZnT2 transcription, whereas depletion of MTF-1 reduces hZnT2 expression. There are five putative metal response elements (MREs) within 1kb upstream of the hZnT2 gene. A serial deletion of the hZnT2 promoter region (from 5' to 3') shows that the two MREs proximal to the gene are essential for Zn-induced promoter activity. Further mutation analysis concludes that the penultimate MRE (MREb) supports the metal-induced promoter activity. The hZnT2 promoter has also a zinc finger E-box binding homeobox (ZEB) binding element. Mutation or deletion of this ZEB binding element elevates the basal and Zn-induced hZnT2 promoter activities. Knockdown of ZEB1 mRNA enhances the hZnT2 transcript level in HEK-293 cells. In MCF-7 (ZEB-deficient) cells, expression of ZEB proteins attenuates the Zn-induced hZnT2 expression. However, expressions of MTF-1 target genes such as human ZnT1 and metallothionein IIA were not affected. Our study shows the expression of the hZnT2 gene is coordinately regulated via active and suppressive modulators.
Collapse
Affiliation(s)
- Yu-Ju Lu
- Institute of Molecular and Cellular Biology and Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Ya-Chuan Liu
- Institute of Molecular and Cellular Biology and Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Meng-Chieh Lin
- Institute of Molecular and Cellular Biology and Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Yi-Ting Chen
- Institute of Molecular and Cellular Biology and Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Lih-Yuan Lin
- Institute of Molecular and Cellular Biology and Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
49
|
O'Reilly EA, Gubbins L, Sharma S, Tully R, Guang MHZ, Weiner-Gorzel K, McCaffrey J, Harrison M, Furlong F, Kell M, McCann A. The fate of chemoresistance in triple negative breast cancer (TNBC). BBA CLINICAL 2015; 3:257-75. [PMID: 26676166 PMCID: PMC4661576 DOI: 10.1016/j.bbacli.2015.03.003] [Citation(s) in RCA: 280] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 03/03/2015] [Accepted: 03/05/2015] [Indexed: 12/21/2022]
Abstract
BACKGROUND Treatment options for women presenting with triple negative breast cancer (TNBC) are limited due to the lack of a therapeutic target and as a result, are managed with standard chemotherapy such as paclitaxel (Taxol®). Following chemotherapy, the ideal tumour response is apoptotic cell death. Post-chemotherapy, cells can maintain viability by undergoing viable cellular responses such as cellular senescence, generating secretomes which can directly enhance the malignant phenotype. SCOPE OF REVIEW How tumour cells retain viability in response to chemotherapeutic engagement is discussed. In addition we discuss the implications of this retained tumour cell viability in the context of the development of recurrent and metastatic TNBC disease. Current adjuvant and neo-adjuvant treatments available and the novel potential therapies that are being researched are also reviewed. MAJOR CONCLUSIONS Cellular senescence and cytoprotective autophagy are potential mechanisms of chemoresistance in TNBC. These two non-apoptotic outcomes in response to chemotherapy are inextricably linked and are neglected outcomes of investigation in the chemotherapeutic arena. Cellular fate assessments may therefore have the potential to predict TNBC patient outcome. GENERAL SIGNIFICANCE Focusing on the fact that cancer cells can bypass the desired cellular apoptotic response to chemotherapy through cellular senescence and cytoprotective autophagy will highlight the importance of targeting non-apoptotic survival pathways to enhance chemotherapeutic efficacy.
Collapse
Affiliation(s)
- Elma A O'Reilly
- UCD Conway Institute of Biomolecular and Biomedical Research, UCD School of Medicine and Medical Science (SMMS), Belfield, Dublin 4, Ireland ; Department of Surgery, Mater Misericordiae Hospital, Dublin 7, Ireland
| | - Luke Gubbins
- UCD Conway Institute of Biomolecular and Biomedical Research, UCD School of Medicine and Medical Science (SMMS), Belfield, Dublin 4, Ireland
| | - Shiva Sharma
- UCD Conway Institute of Biomolecular and Biomedical Research, UCD School of Medicine and Medical Science (SMMS), Belfield, Dublin 4, Ireland ; Department of Surgery, Mater Misericordiae Hospital, Dublin 7, Ireland
| | - Riona Tully
- UCD Conway Institute of Biomolecular and Biomedical Research, UCD School of Medicine and Medical Science (SMMS), Belfield, Dublin 4, Ireland
| | - Matthew Ho Zhing Guang
- UCD Conway Institute of Biomolecular and Biomedical Research, UCD School of Medicine and Medical Science (SMMS), Belfield, Dublin 4, Ireland
| | - Karolina Weiner-Gorzel
- UCD Conway Institute of Biomolecular and Biomedical Research, UCD School of Medicine and Medical Science (SMMS), Belfield, Dublin 4, Ireland
| | - John McCaffrey
- Department of Oncology, Mater Misericordiae Hospital, Dublin 7, Ireland
| | - Michele Harrison
- Department of Pathology, Mater Misericordiae Hospital, Dublin 7, Ireland
| | - Fiona Furlong
- School of Pharmacy, Queens University Belfast, Belfast BT7 1NN, UK
| | - Malcolm Kell
- Department of Surgery, Mater Misericordiae Hospital, Dublin 7, Ireland
| | - Amanda McCann
- UCD Conway Institute of Biomolecular and Biomedical Research, UCD School of Medicine and Medical Science (SMMS), Belfield, Dublin 4, Ireland
| |
Collapse
|
50
|
miR-205 acts as a tumour radiosensitizer by targeting ZEB1 and Ubc13. Nat Commun 2014; 5:5671. [PMID: 25476932 PMCID: PMC4377070 DOI: 10.1038/ncomms6671] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 10/24/2014] [Indexed: 12/14/2022] Open
Abstract
Tumor cells associated with therapy resistance (radioresistance and drug resistance) are likely to give rise to local recurrence and distant metastatic relapse. Recent studies revealed microRNA (miRNA)-mediated regulation of metastasis and epithelial-mesenchymal transition; however, whether specific miRNAs regulate tumor radioresistance and can be exploited as radiosensitizing agents remains unclear. Here we find that miR-205 promotes radiosensitivity and is downregulated in radioresistant subpopulations of breast cancer cells, and that loss of miR-205 is highly associated with poor distant relapse-free survival in breast cancer patients. Notably, therapeutic delivery of miR-205 mimics via nanoliposomes can sensitize the tumor to radiation in a xenograft model. Mechanistically, radiation suppresses miR-205 expression through ataxia telangiectasia mutated (ATM) and zinc finger E-box binding homeobox 1 (ZEB1). Moreover, miR-205 inhibits DNA damage repair by targeting ZEB1 and the ubiquitin-conjugating enzyme Ubc13. These findings identify miR-205 as a radiosensitizing miRNA and reveal a new therapeutic strategy for radioresistant tumors.
Collapse
|