1
|
Le Y, Liu Q, Yang Y, Wu J. The emerging role of nuclear receptor coactivator 4 in health and disease: a novel bridge between iron metabolism and immunity. Cell Death Discov 2024; 10:312. [PMID: 38961066 PMCID: PMC11222541 DOI: 10.1038/s41420-024-02075-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 07/05/2024] Open
Abstract
Nuclear receptor coactivator 4 (NCOA4) has recently been recognized as a selective cargo receptor of ferritinophagy participating in ferroptosis. However, NCOA4 is also a coactivator that modulates the transcriptional activity of many vital nuclear receptors. Recent novel studies have documented the role of NCOA4 in healthy and pathogenic conditions via its modulation of iron- and non-iron-dependent metabolic pathways. NCOA4 exhibits non-ferritinophagic and iron-independent features such as promoting tumorigenesis and erythropoiesis, immunomodulation, regulating autophagy, and participating in DNA replication and mitosis. Full-length human-NCOA4 is composed of 614 amino acids, of which the N-terminal (1-237) contains nuclear-receptor-binding domains, while the C-terminal (238-614) principally contains a ferritin-binding domain. The exploration of the protein structure of NCOA4 suggests that NCOA4 possesses additional significant and complex functions based on its structural domains. Intriguingly, another three isoforms of NCOA4 that are produced by alternative splicing have been identified, which may also display disparate activities in physiological and pathological processes. Thus, NCOA4 has become an important bridge that encompasses interactions between immunity and metabolism. In this review, we outline the latest advances in the important regulating mechanisms underlying NCOA4 actions in health and disease conditions, providing insights into potential therapeutic interventions.
Collapse
Affiliation(s)
- Yue Le
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Qinjie Liu
- Department of General Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Yi Yang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China.
| | - Jie Wu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China.
- Research Center of Surgery, BenQ Medical Center, the Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, 210021, China.
| |
Collapse
|
2
|
Li X, Xiong H, Mou X, Huang C, Thomas ER, Yu W, Jiang Y, Chen Y. Androgen receptor cofactors: A potential role in understanding prostate cancer. Biomed Pharmacother 2024; 173:116338. [PMID: 38417290 DOI: 10.1016/j.biopha.2024.116338] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 02/19/2024] [Accepted: 02/23/2024] [Indexed: 03/01/2024] Open
Abstract
Prostate cancer (PCa) is witnessing a concerning rise in incidence annually, with the androgen receptor (AR) emerging as a pivotal contributor to its growth and progression. Mounting evidence underscores the AR's ability to recruit cofactors, influencing downstream gene transcription and thereby fueling the proliferation and metastasis of PCa cells. Although, clinical strategies involving AR antagonists provide some relief, managing castration resistant prostate cancer (CRPC) remains a formidable challenge. Thus, the need of the hour lies in unearthing new drugs or therapeutic targets to effectively combat PCa. This review encapsulates the pivotal roles played by coactivators and corepressors of AR, notably androgen receptor-associated protein (ARA) and steroid receptor Coactivators (SRC) in PCa. Our data unveils how these cofactors intricately modulate histone modifications, cell cycling, SUMOylation, and apoptosis through their interactions with AR. Among the array of cofactors scrutinised, such as ARA70β, ARA24, ARA160, ARA55, ARA54, PIAS1, PIAS3, SRC1, SRC2, SRC3, PCAF, p300/CBP, MED1, and CARM1, several exhibit upregulation in PCa. Conversely, other cofactors like ARA70α, PIASy, and NCoR/SMRT demonstrate downregulation. This duality underscores the complexity of AR cofactor dynamics in PCa. Based on our findings, we propose that manipulating cofactor regulation to modulate AR function holds promise as a novel therapeutic avenue against advanced PCa. This paradigm shift offers renewed hope in the quest for effective treatments in the face of CRPC's formidable challenges.
Collapse
Affiliation(s)
- Xiang Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Haojun Xiong
- Department of Dermatology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xingzhu Mou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Department of Dermatology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Cancan Huang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Department of Dermatology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | | | - Wenjing Yu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Yu Jiang
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China.
| | - Yan Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Department of Dermatology, The Affiliated Hospital, Southwest Medical University, Luzhou, China.
| |
Collapse
|
3
|
Gioukaki C, Georgiou A, Gkaralea LE, Kroupis C, Lazaris AC, Alamanis C, Thomopoulou GE. Unravelling the Role of P300 and TMPRSS2 in Prostate Cancer: A Literature Review. Int J Mol Sci 2023; 24:11299. [PMID: 37511059 PMCID: PMC10379122 DOI: 10.3390/ijms241411299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/26/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023] Open
Abstract
Prostate cancer is one of the most common malignant diseases in men, and it contributes significantly to the increased mortality rate in men worldwide. This study aimed to review the roles of p300 and TMPRSS2 (transmembrane protease, serine 2) in the AR (androgen receptor) pathway as they are closely related to the development and progression of prostate cancer. This paper represents a library-based study conducted by selecting the most suitable, up-to-date scientific published articles from online journals. We focused on articles that use similar techniques, particularly those that use prostate cancer cell lines and immunohistochemical staining to study the molecular impact of p300 and TMPRSS2 in prostate cancer specimens. The TMPRSS2:ERG fusion is considered relevant to prostate cancer, but its association with the development and progression as well as its clinical significance have not been fully elucidated. On the other hand, high p300 levels in prostate cancer biopsies predict larger tumor volumes, extraprostatic extension of disease, and seminal vesicle involvement at prostatectomy, and may be associated with prostate cancer progression after surgery. The inhibition of p300 has been shown to reduce the proliferation of prostate cancer cells with TMPRSS2:ETS (E26 transformation-specific) fusions, and combining p300 inhibitors with other targeted therapies may increase their efficacy. Overall, the interplay between the p300 and TMPRSS2 pathways is an active area of research.
Collapse
Affiliation(s)
- Charitomeni Gioukaki
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Alexandros Georgiou
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | | | - Christos Kroupis
- Department of Clinical Biochemistry, Attikon University Hospital, National and Kapodistrian University of Athens, 12461 Athens, Greece
| | - Andreas C Lazaris
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Christos Alamanis
- 1st Urology Department, Laiko Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Georgia Eleni Thomopoulou
- Cytopathology Department, Attikon University Hospital, National and Kapodistrian University of Athens, 12461 Athens, Greece
| |
Collapse
|
4
|
Li T, Yang WY, Liu TT, Li Y, Liu L, Zheng X, Zhao L, Zhang F, Hu Y. Advances in the Diagnosis and Treatment of a Driving Target: RET Rearrangements in non-Small-Cell Lung Cancer (NSCLC) Especially in China. Technol Cancer Res Treat 2023; 22:15330338221148802. [PMID: 36628459 PMCID: PMC9837270 DOI: 10.1177/15330338221148802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
In the era of precision medicine, with the deepening of the research on malignant tumor driving genes, clinical oncology has fully entered the era of targeted therapy. For non-small-cell lung cancer (NSCLC), the development of targeted drugs targeting driver genes, such as epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK), has successfully opened up a new model of targeted therapy. At present, proto-oncogene rearranged during transfection (RET) fusion gene is an important novel oncogenic driving target, and specific receptor tyrosine kinase inhibitors (TKIs) targeting RET fusion have been approved. This article will review the latest research about the molecular characteristics, pathogenesis, detection, and clinical treatment strategies of RET rearrangements especially in China.
Collapse
Affiliation(s)
- Tao Li
- Department of Oncology, The First Medical Center of PLA General Hospital, Graduate School, Medical College of Chinese PLA, Beijing, China,Chinese PLA Key Laboratory of Oncology, Key Laboratory for Tumor Targeting Therapy and Antibody Drugs (Ministry of Education), China,Tao Li, MD, Department of Oncology, The First Medical Center of PLA General Hospital, Graduate School, Medical College of Chinese PLA, Beijing 100029, China.
| | - Wen-Yu Yang
- Department of Oncology, The First Medical Center of PLA General Hospital, Graduate School, Medical College of Chinese PLA, Beijing, China,Chinese PLA Key Laboratory of Oncology, Key Laboratory for Tumor Targeting Therapy and Antibody Drugs (Ministry of Education), China,School of Medicine, Nankai University, Tianjin, China
| | - Ting-Ting Liu
- Department of Pulmonary and Critical Care Medicine, The Second Medical Center of PLA General Hospital, Beijing, China,Graduate School, Medical College of Chinese PLA, Beijing, China
| | - Yao Li
- Department of Oncology, The First Medical Center of PLA General Hospital, Graduate School, Medical College of Chinese PLA, Beijing, China,Chinese PLA Key Laboratory of Oncology, Key Laboratory for Tumor Targeting Therapy and Antibody Drugs (Ministry of Education), China
| | - Lu Liu
- Department of Nutriology, The First Medical Center of PLA General Hospital, Graduate School, Medical College of Chinese PLA, Beijing, China
| | - Xuan Zheng
- Department of Oncology, The First Medical Center of PLA General Hospital, Graduate School, Medical College of Chinese PLA, Beijing, China,Chinese PLA Key Laboratory of Oncology, Key Laboratory for Tumor Targeting Therapy and Antibody Drugs (Ministry of Education), China
| | - Lei Zhao
- Institute of Translational Medicine, PLA General Hospital, Beijing, China
| | - Fan Zhang
- Department of Oncology, The First Medical Center of PLA General Hospital, Graduate School, Medical College of Chinese PLA, Beijing, China,Chinese PLA Key Laboratory of Oncology, Key Laboratory for Tumor Targeting Therapy and Antibody Drugs (Ministry of Education), China
| | - Yi Hu
- Department of Oncology, The First Medical Center of PLA General Hospital, Graduate School, Medical College of Chinese PLA, Beijing, China,Chinese PLA Key Laboratory of Oncology, Key Laboratory for Tumor Targeting Therapy and Antibody Drugs (Ministry of Education), China
| |
Collapse
|
5
|
Santana-Codina N, del Rey MQ, Kapner KS, Zhang H, Gikandi A, Malcolm C, Poupault C, Kuljanin M, John KM, Biancur DE, Chen B, Das NK, Lowder KE, Hennessey CJ, Huang W, Yang A, Shah YM, Nowak JA, Aguirre AJ, Mancias JD. NCOA4-Mediated Ferritinophagy Is a Pancreatic Cancer Dependency via Maintenance of Iron Bioavailability for Iron-Sulfur Cluster Proteins. Cancer Discov 2022; 12:2180-2197. [PMID: 35771492 PMCID: PMC9437572 DOI: 10.1158/2159-8290.cd-22-0043] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 05/02/2022] [Accepted: 06/07/2022] [Indexed: 11/16/2022]
Abstract
Pancreatic ductal adenocarcinomas (PDAC) depend on autophagy for survival; however, the metabolic substrates that autophagy provides to drive PDAC progression are unclear. Ferritin, the cellular iron storage complex, is targeted for lysosomal degradation (ferritinophagy) by the selective autophagy adaptor NCOA4, resulting in release of iron for cellular utilization. Using patient-derived and murine models of PDAC, we demonstrate that ferritinophagy is upregulated in PDAC to sustain iron availability, thereby promoting tumor progression. Quantitative proteomics reveals that ferritinophagy fuels iron-sulfur cluster protein synthesis to support mitochondrial homeostasis. Targeting NCOA4 leads to tumor growth delay and prolonged survival but with the development of compensatory iron acquisition pathways. Finally, enhanced ferritinophagy accelerates PDAC tumorigenesis, and an elevated ferritinophagy expression signature predicts for poor prognosis in patients with PDAC. Together, our data reveal that the maintenance of iron homeostasis is a critical function of PDAC autophagy, and we define NCOA4-mediated ferritinophagy as a therapeutic target in PDAC. SIGNIFICANCE Autophagy and iron metabolism are metabolic dependencies in PDAC. However, targeted therapies for these pathways are lacking. We identify NCOA4-mediated selective autophagy of ferritin ("ferritinophagy") as upregulated in PDAC. Ferritinophagy supports PDAC iron metabolism and thereby tumor progression and represents a new therapeutic target in PDAC. See related commentary by Jain and Amaravadi, p. 2023. See related article by Ravichandran et al., p. 2198. This article is highlighted in the In This Issue feature, p. 2007.
Collapse
Affiliation(s)
- Naiara Santana-Codina
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Maria Quiles del Rey
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Kevin S. Kapner
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Huan Zhang
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Ajami Gikandi
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Callum Malcolm
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Clara Poupault
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Miljan Kuljanin
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Kristen M. John
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Douglas E. Biancur
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Brandon Chen
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Nupur K. Das
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Kristen E. Lowder
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Connor J. Hennessey
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Wesley Huang
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Annan Yang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Yatrik M. Shah
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, Michigan
| | - Jonathan A. Nowak
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Andrew J. Aguirre
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Joseph D. Mancias
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
- Department of Radiation Oncology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
6
|
Anguiano B, Montes de Oca C, Delgado-González E, Aceves C. Prostate gland as a target organ of thyroid hormones: advances and controversies. Endocr Connect 2022; 11:e210581. [PMID: 35041618 PMCID: PMC8859956 DOI: 10.1530/ec-21-0581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 01/17/2022] [Indexed: 12/02/2022]
Abstract
Thyroid hormones (THs) are involved in the development and function of the male reproductive system, but their effects on the prostate have been poorly studied. This work reviews studies related to the interrelationship between the thyroid and the prostate. The information presented here is based upon bibliographic searches in PubMed using the following search terms: prostate combined with thyroid hormone or triiodothyronine, thyroxine, hypothyroidism, hyperthyroidism, or deiodinase. We identified and searched 49 articles directly related to the issue, and discarded studies related to endocrine disruptors. The number of publications has grown in the last 20 years, considering that one of the first studies was published in 1965. This review provides information based on in vitro studies, murine models, and clinical protocols in patients with thyroid disorders. Studies indicate that THs regulate different aspects of growth, metabolism, and prostate pathology, whose global effect depends on total and/or free concentrations of THs in serum, local bioavailability, and the endocrine androgen/thyronine context.
Collapse
Affiliation(s)
- Brenda Anguiano
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
- Correspondence should be addressed to B Anguiano:
| | - Carlos Montes de Oca
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| | - Evangelina Delgado-González
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| | - Carmen Aceves
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| |
Collapse
|
7
|
Jonnalagadda B, Arockiasamy S, Krishnamoorthy S. Cellular growth factors as prospective therapeutic targets for combination therapy in androgen independent prostate cancer (AIPC). Life Sci 2020; 259:118208. [PMID: 32763294 DOI: 10.1016/j.lfs.2020.118208] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 07/27/2020] [Accepted: 08/02/2020] [Indexed: 12/21/2022]
Abstract
Cancer is the second leading cause of death worldwide, with prostate cancer, the second most commonly diagnosed cancer among men. Prostate cancer develops in the peripheral zone of the prostate gland, and the initial progression largely depends on androgens, the male reproductive hormone that regulates the growth and development of the prostate gland and testis. The currently available treatments for androgen dependent prostate cancer are, however, effective for a limited period, where the patients show disease relapse, and develop androgen-independent prostate cancer (AIPC). Studies have shown various intricate cellular processes such as, deregulation in multiple biochemical and signaling pathways, intra-tumoral androgen synthesis; AR over-expression and mutations and AR activation via alternative growth pathways are involved in progression of AIPC. The currently approved treatment strategies target a single cellular protein or pathway, where the cells slowly develop resistance and adapt to proliferate via other cellular pathways over a period of time. Therefore, an increased research aims to understand the efficacy of combination therapy, which targets multiple interlinked pathways responsible for acquisition of resistance and survival. The combination therapy is also shown to enhance efficacy as well as reduce toxicity of the drugs. Thus, the present review focuses on the signaling pathways involved in the progression of AIPC, comprising a heterogeneous population of cells and the advantages of combination therapy. Several clinical and pre-clinical studies on a variety of combination treatments have shown beneficial outcomes, yet further research is needed to understand the potential of combination therapy and its diverse strategies.
Collapse
Affiliation(s)
- Bhavana Jonnalagadda
- Department of Biomedical Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - Sumathy Arockiasamy
- Department of Biomedical Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai, India.
| | - Sriram Krishnamoorthy
- Department of Urology, Sri Ramachandra Medical Centre, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| |
Collapse
|
8
|
Rockfield S, Raffel J, Mehta R, Rehman N, Nanjundan M. Iron overload and altered iron metabolism in ovarian cancer. Biol Chem 2017; 398:995-1007. [PMID: 28095368 DOI: 10.1515/hsz-2016-0336] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 01/09/2017] [Indexed: 12/28/2022]
Abstract
Iron is an essential element required for many processes within the cell. Dysregulation in iron homeostasis due to iron overload is detrimental. This nutrient is postulated to contribute to the initiation of cancer; however, the mechanisms by which this occurs remain unclear. Defining how iron promotes the development of ovarian cancers from precursor lesions is essential for developing novel therapeutic strategies. In this review, we discuss (1) how iron overload conditions may initiate ovarian cancer development, (2) dysregulated iron metabolism in cancers, (3) the interplay between bacteria, iron, and cancer, and (4) chemotherapeutic strategies targeting iron metabolism in cancer patients.
Collapse
|
9
|
Hu J, Wang G, Sun T. Dissecting the roles of the androgen receptor in prostate cancer from molecular perspectives. Tumour Biol 2017; 39:1010428317692259. [PMID: 28475016 DOI: 10.1177/1010428317692259] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Androgen receptor plays a pivotal role in prostate cancer progression, and androgen deprivation therapy to intercept androgen receptor signal pathway is an indispensable treatment for most advanced prostate cancer patients to delay cancer progression. However, the emerging of castration-resistant prostate cancer reminds us the alteration of androgen receptor, which includes androgen receptor mutation, the formation of androgen receptor variants, and androgen receptor distribution in cancer cells. In this review, we introduce the process of androgen receptor and also its variants' formation, translocation, and function alteration by protein modification or interaction with other pathways. We dissect the roles of androgen receptor in prostate cancer from molecular perspective to provide clues for battling prostate cancer, especially castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Jieping Hu
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Gongxian Wang
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ting Sun
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
10
|
Culig Z. Androgen Receptor Coactivators in Regulation of Growth and Differentiation in Prostate Cancer. J Cell Physiol 2016. [PMID: 26201947 DOI: 10.1002/jcp.25099] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Androgen receptor (AR) is a key factor in regulation of growth and differentiation in normal and malignant prostate. Endocrine therapies for prostate cancer include inhibition of androgen production either by analogs of luteinizing hormone releasing hormone or abiraterone acetate and/or use of anti-androgens such as hydroxyflutamide, bicalutamide, and enzalutamide. Castration therapy-resistant cancer develops inevitably in patients who undergo treatment. AR coactivators are proteins which interact with one or more regions of the AR thus enhancing its function. Although several functions of AR coactivators may be redundant, specific functions have been identified and analyzed. The p160 group of coactivators, SRC-1, -2, and -3 not only potentiate the activation of the AR, but are also implicated in potentiation of function of insulin-like growth factor-I and activation of the Akt pathway. Transcriptional integrators p300 and CBP are up-regulated by androgen ablation and may influence antagonist/agonist balance of non-steroidal anti-androgens. A therapy approach designed to target p300 in prostate cancer revealed its role in regulation of proliferation of migration of androgen-sensitive and -insensitive prostate cancer cells. Coactivators p300 and SRC-1 are required for AR activation by interleukin-6 (IL-6), a cytokine that is overexpressed in castration therapy-resistant prostate cancer. Some coactivators, such as Vav3, are involved in regulation of transcriptional activity of truncated AR, which emerge during endocrine thrapy. Stimulation of cellular migration and invasion by AR coactivators has also been described. Translational studies with aim to introduce anti-AR coactivator therapy have not been successfully implemented in the clinic so far.
Collapse
Affiliation(s)
- Zoran Culig
- Experimental Urology, Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
11
|
Delgado-González E, Sánchez-Tusie AA, Morales G, Aceves C, Anguiano B. Triiodothyronine Attenuates Prostate Cancer Progression Mediated by β-Adrenergic Stimulation. Mol Med 2016; 22:1-11. [PMID: 26928389 DOI: 10.2119/molmed.2015.00047] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 02/19/2016] [Indexed: 01/10/2023] Open
Abstract
Prostate cancer cells are responsive to adrenergic and thyroid stimuli. It is well established that β-adrenergic activation (protein kinase A [PKA]/cAMP response element binding protein [CREB]) promotes cancer progression, but the role of thyroid hormones is poorly understood. We analyzed the effects of β-adrenergic stimulation (isoproterenol [ISO]) and/or thyroid hormone on neuroendocrine (NE) differentiation and cell invasion, using in vivo (LNCaP tumor) and in vitro models (LNCaP and DU145 human cells). Nude mice were inoculated with LNCaP cells and were treated for 6 wks with ISO (200 μg/d), triiodothyronine (T3, 2.5 μg/d) or both. ISO alone reduced tumor growth but increased tumor expression of cAMP response element (CRE)-dependent genes (real-time polymerase chain reaction, chromogranin A, neuron-specific enolase, survivin, vascular endothelial growth factor [VEGF], urokinase plasmin activator [uPA] and metalloproteinase-9 [MMP-9]) and some proteins related to NE differentiation and/or invasiveness (synaptophysin, VEGF, pCREB). T3 reduced tumor growth and prevented the overexpression of ISO-stimulated factors through a pCREB-independent mechanism. In low invasive LNCaP cells, 50 μmol/L ISO or 100 nmol/L thyroxine (T4) induced the acquisition of NE-like morphology (phase-contrast microscopy), increased VEGF secretion (ELISA) and invasive capacity (Transwell assay), but no synergistic effects were observed after the coadministration of ISO + T4. In contrast, 10 nmol/L T3 alone had no effect, but it prevented the NE-like morphology and invasiveness stimulated by ISO. None of these treatments had any effect on highly invasive DU145 cells. In summary, this study showed that ISO and T4 increase cancer progression, and T3 attenuates ISO-stimulated progression. Further studies are required to determine if changes in the ratio of T4/T3 could be relevant for prostate cancer progression.
Collapse
Affiliation(s)
- Evangelina Delgado-González
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| | - Ana Alicia Sánchez-Tusie
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| | - Giapsy Morales
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| | - Carmen Aceves
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| | - Brenda Anguiano
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| |
Collapse
|
12
|
Rawal RM, Joshi MN, Bhargava P, Shaikh I, Pandit AS, Patel RP, Patel S, Kothari K, Shah M, Saxena A, Bagatharia SB. Tobacco habituated and non-habituated subjects exhibit different mutational spectrums in head and neck squamous cell carcinoma. 3 Biotech 2015; 5:685-696. [PMID: 28324520 PMCID: PMC4569615 DOI: 10.1007/s13205-014-0267-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 11/15/2014] [Indexed: 12/28/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the sixth most common non-skin cancer in the world. Tobacco chewing is implicated with most of the cases of HNSCC but this type of cancer is increasing in non-tobacco chewers as well. This study was instigated to provide comprehensive variant and gene-level data in HNSCC subjects of the Indian population and fill the gap in the literature on comparative assessment of gene mutations in cancer subjects with a habit of tobacco and those without any habit using targeted amplicon sequencing. We performed targeted Amplicon sequencing of 409 tumor suppressor genes and oncogenes, frequently mutated across many cancer types, including head and neck. DNA from primary tumor tissues and matched blood was analyzed for HNSCC patients with a habit of tobacco and those without any habit. PDE4DIP, SYNE1, and NOTCH1 emerged as the highly mutated genes in HNSCC. A total of 39 candidate causal variants in 22 unique cancer driver genes were identified in non-habitual (WoH) and habitual (WH) subjects. Comparison of genes from both the subjects, showed seven unique cancer driver genes (KIT, ATM, RNF213, GATA2, DST, RET, CYP2C19) in WoH, while WH showed five (IL7R, PKHD1, MLL3, PTPRD, MAPK8) and 10 genes (SETD2, ATR, CDKN2A, NCOA4, TP53, SYNE1, KAT6B, THBS1, PTPRT, and FGFR3) were common to both subjects. In addition to this NOTCH1, NOTCH2, and NOTCH4 gene were found to be mutated only in habitual subjects. These findings strongly support a causal role for tobacco, acting via PI3K and MAPK pathway inhibition and stimulation of various genes leading to oncogenic transformations in case of tobacco chewers. In case of non-tobacco chewers it appears that mutations in the pathway affecting the squamous epithelial lineage and DNA repair genes lead to HNSCC. Somatic mutation in CYP2C19 gene in the non-habitual subjects suggests that this gene may have a tobacco independent role in development and progression of HNSCC. In addition to sharing high mutation rate, NOTCH gene family was found to be mutated only in habitual sample. Further, presence of mutated genes not earlier reported to be involved in HNSCC, suggest that the Indian sub-continent may have different sets of genes, as compared to other parts of the world, involved in the development and progression of HNSCC.
Collapse
Affiliation(s)
- Rakesh M Rawal
- Gujarat Cancer and Research Institute, Gujarat Cancer Society, Civil Hospital Campus, Asarwa, Ahmedabad, 380 016, Gujarat, India
| | - Madhvi N Joshi
- Gujarat State Biotechnology Mission, Department of Science and Technology, Government of Gujarat, 11th Block, 9th Floor, Udyog Bhavan, Gandhinagar, 382 011, Gujarat, India
| | - Poonam Bhargava
- Gujarat State Biotechnology Mission, Department of Science and Technology, Government of Gujarat, 11th Block, 9th Floor, Udyog Bhavan, Gandhinagar, 382 011, Gujarat, India
| | - Inayat Shaikh
- Gujarat State Biotechnology Mission, Department of Science and Technology, Government of Gujarat, 11th Block, 9th Floor, Udyog Bhavan, Gandhinagar, 382 011, Gujarat, India
| | - Aanal S Pandit
- Gujarat State Biotechnology Mission, Department of Science and Technology, Government of Gujarat, 11th Block, 9th Floor, Udyog Bhavan, Gandhinagar, 382 011, Gujarat, India
| | - Riddhi P Patel
- Gujarat State Biotechnology Mission, Department of Science and Technology, Government of Gujarat, 11th Block, 9th Floor, Udyog Bhavan, Gandhinagar, 382 011, Gujarat, India
| | - Shanaya Patel
- Gujarat Cancer and Research Institute, Gujarat Cancer Society, Civil Hospital Campus, Asarwa, Ahmedabad, 380 016, Gujarat, India
| | - Kiran Kothari
- Gujarat Cancer and Research Institute, Gujarat Cancer Society, Civil Hospital Campus, Asarwa, Ahmedabad, 380 016, Gujarat, India
| | - Manoj Shah
- Gujarat Cancer and Research Institute, Gujarat Cancer Society, Civil Hospital Campus, Asarwa, Ahmedabad, 380 016, Gujarat, India
| | - Akshay Saxena
- Gujarat State Biotechnology Mission, Department of Science and Technology, Government of Gujarat, 11th Block, 9th Floor, Udyog Bhavan, Gandhinagar, 382 011, Gujarat, India
| | - Snehal B Bagatharia
- Gujarat State Biotechnology Mission, Department of Science and Technology, Government of Gujarat, 11th Block, 9th Floor, Udyog Bhavan, Gandhinagar, 382 011, Gujarat, India.
| |
Collapse
|
13
|
Wang C, Sun H, Zou R, Zhou T, Wang S, Sun S, Tong C, Luo H, Li Y, Li Z, Wang E, Chen Y, Cao L, Li F, Zhao Y. MDC1 functionally identified as an androgen receptor co-activator participates in suppression of prostate cancer. Nucleic Acids Res 2015; 43:4893-908. [PMID: 25934801 PMCID: PMC4446443 DOI: 10.1093/nar/gkv394] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 04/14/2015] [Indexed: 02/06/2023] Open
Abstract
Mediator of DNA damage checkpoint protein 1 (MDC1) is essential for DNA damage response. However, the role of MDC1 in modulating gene transcription independently of DNA damage and the underlying mechanisms have not been fully defined. Androgen receptor (AR) is the central signaling pathway in prostate cancer (PCa) and its target genes are involved in both promotion and suppression of PCa. Here, we functionally identified MDC1 as a co-activator of AR. We demonstrate that MDC1 facilitates the association between AR and histone acetyltransferase GCN5, thereby increasing histone H3 acetylation level on cis-regulatory elements of AR target genes. MDC1 knockdown promotes PCa cells growth and migration. Moreover, depletion of MDC1 results in decreased expression of a subset of the endogenous androgen-induced target genes, including cell cycle negative regulator p21 and PCa metastasis inhibitor Vinculin, in AR positive PCa cell lines. Finally, the expression of MDC1 and p21 correlates negatively with aggressive phenotype of clinical PCa. These studies suggest that MDC1 as an epigenetic modifier regulates AR transcriptional activity and MDC1 may function as a tumor suppressor of PCa, and provide new insight into co-factor-AR-signaling pathway mechanism and a better understanding of the function of MDC1 on PCa.
Collapse
Affiliation(s)
- Chunyu Wang
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Hongmiao Sun
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Renlong Zou
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Tingting Zhou
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Shengli Wang
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Shiying Sun
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Changci Tong
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Hao Luo
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Yanshu Li
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Zhenhua Li
- Department of Urology, the First Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, China
| | - Enhua Wang
- Department of Pathology, the First Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, China
| | - Yuhua Chen
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Liu Cao
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Feng Li
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Yue Zhao
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| |
Collapse
|
14
|
Ligr M, Wu X, Daniels G, Zhang D, Wang H, Hajdu C, Wang J, Pan R, Pei Z, Zhang L, Melis M, Pincus MR, Saunders JK, Lee P, Xu R. Imbalanced expression of Tif1γ inhibits pancreatic ductal epithelial cell growth. Am J Cancer Res 2014; 4:196-210. [PMID: 24959375 PMCID: PMC4065401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 03/30/2014] [Indexed: 06/03/2023] Open
Abstract
Transcriptional intermediary factor 1 gamma (Tif1γ) (Ectodermin/PTC7/RFG7/TRIM33) is a transcriptional cofactor with an important role in the regulation of the TGFβ pathway. It has been suggested that it competes with Smad2/Smad3 for binding to Smad4, or alternatively that it may target Smad4 for degradation, although its role in carcinogenesis is unclear. In this study, we showed that Tif1γ interacts with Smad1/Smad4 complex in vivo, using both yeast two-hybrid and coimmunoprecipitation assays. We demonstrated that Tif1γ inhibits transcriptional activity of the Smad1/Smad4 complex through its PHD domain or bromo-domainin pancreatic cells by luciferase assay. Additionally, there is a dynamic inverse relationship between the levels of Tif1γ and Smad4 in benign and malignant pancreatic cell lines. Overexpression of Tif1γ resulted in decreased level of Smad4. Both overexpression and knockdown of Tif1γ resulted in growth inhibition in both benign and cancerous pancreatic cell lines, attributable to a G2-phase cell cycle arrest, but only knockdown of Tif1γ reduces tumor cell invasiveness in vitro. Our study demonstrated that imbalanced expression of Tif1γ results in inhibition of pancreatic ductal epithelial cell growth. In addition, knockdown of Tif1γ may inhibit tumor invasion. These data suggest that Tif1γ might serve as a potential therapeutic target for pancreatic cancer.
Collapse
Affiliation(s)
- Martin Ligr
- Department of Pathology, New York University Langone Medical CenterNew York, NY, USA
| | - Xinyu Wu
- Department of Pathology, New York University Langone Medical CenterNew York, NY, USA
| | - Garrett Daniels
- Department of Pathology, New York University Langone Medical CenterNew York, NY, USA
| | - David Zhang
- Department of Pathology, Mount Sinai School of MedicineNew York, NY, USA
| | - Huamin Wang
- Department of Pathology, The University of Texas M.D. Anderson Cancer CenterHouston, TX, USA
| | - Cristina Hajdu
- Department of Pathology, New York University Langone Medical CenterNew York, NY, USA
| | - Jinhua Wang
- NYU Cancer Institute, New York University Langone Medical CenterNew York, NY, USA
| | - Ruimin Pan
- Department of Pathology, New York University Langone Medical CenterNew York, NY, USA
| | - Zhiheng Pei
- Department of Pathology, New York University Langone Medical CenterNew York, NY, USA
| | - Lanjing Zhang
- Department of Pathology, The University of Texas M.D. Anderson Cancer CenterHouston, TX, USA
| | - Marcovalerio Melis
- Department of Pathology, Mount Sinai School of MedicineNew York, NY, USA
| | | | - John K Saunders
- Department of Surgery, New York University Langone Medical CenterNew York, NY, USA
| | - Peng Lee
- Department of Pathology, New York University Langone Medical CenterNew York, NY, USA
- Department of Urology, New York University Langone Medical CenterNew York, NY, USA
- NYU Cancer Institute, New York University Langone Medical CenterNew York, NY, USA
- New York Harbor Healthcare SystemNew York, NY, USA
| | - Ruliang Xu
- Department of Pathology, New York University Langone Medical CenterNew York, NY, USA
| |
Collapse
|
15
|
Daniels G, Jha R, Shen Y, Logan SK, Lee P. Androgen receptor coactivators that inhibit prostate cancer growth. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2014; 2:62-70. [PMID: 25374906 PMCID: PMC4219292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 03/26/2014] [Indexed: 06/04/2023]
Abstract
It is well documented that androgen receptor (AR), a steroid hormone receptor, is important for prostate cancer (PCa) growth. Conversely, however, there is increasing evidence that activation of AR by androgens can also lead to growth suppression in prostate cells. AR mediated transcription is regulated by a number of different transcriptional coactivators. Changes in expression level or cellular localization of specific coactivators may play a crucial role in this switch between proliferative and anti- proliferative processes regulated by AR target gene programs. In this review, we discuss the expression and function of several AR coactivators exhibiting growth suppressive function in PCa, including ARA70/ELE1/NCOA4, androgen receptor coactivator p44/MEP50/WDR77, TBLR1, and ART-27. In luciferase reporter assays, they all have been shown to activate AR mediated transcriptional activation. ARA70 exists in two forms, the full length nuclear ARA70α and internally spliced cytoplasmic ARA70β. For p44 and TBLR1, we identified nuclear and cytoplasmic forms with distinct expression and function. In comparison of their expression (ARA70α, p44, TBLR1 and ART-27) in prostate, these coactivators are expressed in the nucleus of benign prostate epithelial cells while they are more predominantly expressed in cytoplasmic form (ARA70β, cytoplasmic p44 and TBLR1) in PCa. Consistent with their nuclear expression in benign prostate, the nuclear form of these coactivators inhibit PCa growth targeting a subset of AR target genes. In contrast, the cytoplasmic versions of these proteins enhance PCa growth and invasion. Interestingly, first characterized as an AR coactivator in luciferase assays, ART-27 functions as corepressor for endogenous AR target genes. Importantly, the growth inhibitions by these nuclear proteins are androgen-dependent processes and the regulation of invasion is androgen-independent. Understanding the molecular switches involved in the transition from AR dependent growth promotion to growth suppression and dysregulation of these coactivator proteins promoting androgen-independent invasion may lead to identification of novel therapeutic targets for PCa.
Collapse
Affiliation(s)
- Garrett Daniels
- Department of Pathology, New York University School of MedicineNew York, NY
| | - Ruchi Jha
- Department of Pathology, New York University School of MedicineNew York, NY
| | - Ying Shen
- Department of Pathology, New York University School of MedicineNew York, NY
| | - Susan K Logan
- Department of Pathology, New York University School of MedicineNew York, NY
- Department of Urology, New York University School of MedicineNew York, NY
- Department of Biochemistry and Clinical Pharmacology, New York University School of MedicineNew York, NY
| | - Peng Lee
- Department of Pathology, New York University School of MedicineNew York, NY
- Department of Urology, New York University School of MedicineNew York, NY
- Department of New York Harbor Healthcare System, New York University School of MedicineNew York, NY
| |
Collapse
|
16
|
Daniels G, Li Y, Gellert LL, Zhou A, Melamed J, Wu X, Zhang X, Zhang D, Meruelo D, Logan SK, Basch R, Lee P. TBLR1 as an androgen receptor (AR) coactivator selectively activates AR target genes to inhibit prostate cancer growth. Endocr Relat Cancer 2014; 21:127-42. [PMID: 24243687 PMCID: PMC3947037 DOI: 10.1530/erc-13-0293] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Androgen receptor (AR), a steroid hormone receptor, is critical for prostate cancer growth. However, activation of AR by androgens can also lead to growth suppression and differentiation. Transcriptional cofactors play an important role in this switch between proliferative and anti-proliferative AR target gene programs. Transducin β-like-related protein 1 (TBLR1), a core component of the nuclear receptor corepressor complex, shows both corepressor and coactivator activities on nuclear receptors, but little is known about its effects on AR and prostate cancer. We characterized TBLR1 as a coactivator of AR in prostate cancer cells and determined that the activation is dependent on both phosphorylation and 19S proteosome. We showed that TBLR1 physically interacts with AR and directly occupies the androgen-response elements of the affected AR target genes in an androgen-dependent manner. TBLR1 is primarily localized in the nucleus in benign prostate cells and nuclear expression is significantly reduced in prostate cancer cells in culture. Similarly, in human tumor samples, the expression of TBLR1 in the nucleus is significantly reduced in the malignant glands compared with the surrounding benign prostatic glands (P<0.005). Stable ectopic expression of nuclear TBLR1 leads to androgen-dependent growth suppression of prostate cancer cells in vitro and in vivo by selective activation of androgen-regulated genes associated with differentiation (e.g. KRT18) and growth suppression (e.g. NKX3-1), but not cell proliferation of the prostate cancer. Understanding the molecular switches involved in the transition from AR-dependent growth promotion to AR-dependent growth suppression will lead to more successful treatments for prostate cancer.
Collapse
Affiliation(s)
- Garrett Daniels
- Department of Pathology, New York University School of Medicine, New York, NY
| | - Yirong Li
- Department of Pathology, New York University School of Medicine, New York, NY
| | - Lan Lin Gellert
- Department of Pathology, New York University School of Medicine, New York, NY
| | - Albert Zhou
- Department of Pathology, New York University School of Medicine, New York, NY
| | - Jonathan Melamed
- Department of Pathology, New York University School of Medicine, New York, NY
| | - Xinyu Wu
- Department of Pathology, New York University School of Medicine, New York, NY
| | - Xinming Zhang
- Department of Pathology, New York University School of Medicine, New York, NY
| | - David Zhang
- Department of Pathology, Mount Sinai School of Medicine, New York, NY
| | - Daniel Meruelo
- Department of Pathology, New York University School of Medicine, New York, NY
- NYU Cancer Institute, New York University School of Medicine, New York, NY
| | - Susan K. Logan
- NYU Cancer Institute, New York University School of Medicine, New York, NY
- Department of Urology, New York University School of Medicine, New York, NY
- Department of Pharmacology, New York University School of Medicine, New York, NY
| | - Ross Basch
- Department of Pathology, New York University School of Medicine, New York, NY
| | - Peng Lee
- Department of Pathology, New York University School of Medicine, New York, NY
- NYU Cancer Institute, New York University School of Medicine, New York, NY
- Department of Pharmacology, New York University School of Medicine, New York, NY
- New York Harbor Healthcare System, New York University School of Medicine, New York, NY
| |
Collapse
|
17
|
Santoro M, Carlomagno F. Central role of RET in thyroid cancer. Cold Spring Harb Perspect Biol 2013; 5:a009233. [PMID: 24296167 DOI: 10.1101/cshperspect.a009233] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
RET (rearranged during transfection) is a receptor tyrosine kinase involved in the development of neural crest derived cell lineages, kidney, and male germ cells. Different human cancers, including papillary and medullary thyroid carcinomas, lung adenocarcinomas, and myeloproliferative disorders display gain-of-function mutations in RET. Accordingly, RET protein has become a promising molecular target for cancer treatment.
Collapse
Affiliation(s)
- Massimo Santoro
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Universita' degli Studi di Napoli Federico II, 80131 Napoli, Italy
| | | |
Collapse
|
18
|
Culig Z, Santer FR. Molecular aspects of androgenic signaling and possible targets for therapeutic intervention in prostate cancer. Steroids 2013; 78:851-9. [PMID: 23643785 DOI: 10.1016/j.steroids.2013.04.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 04/05/2013] [Accepted: 04/16/2013] [Indexed: 01/18/2023]
Abstract
The androgen axis is of crucial importance in the development of novel therapeutic approaches for non-organ-confined prostate cancer. Recent studies revealed that tumor cells have the ability to synthesize androgenic hormones in an intracrine manner. This recognition opened the way for the development of a novel drug, abiraterone acetate, which shows benefits in clinical trials. A novel anti-androgen enzalutamide that inhibits androgen receptor (AR) nuclear translocation has also been developed and tested in the clinic. AR coactivators exert specific cellular regulatory functions, however it is difficult to improve the treatment because of a large number of coregulators overexpressed in prostate cancer. AR itself is a target of several miRNAs which may cause its increased degradation, inhibition of proliferation, and increased apoptosis. Truncated AR occur in prostate cancer as a consequence of alternative splicing. They exhibit ligand-independent transcriptional activity. Although there has been an improvement of endocrine therapy in prostate cancer, increased intracrine ligand synthesis and appearance of variant receptors may facilitate the development of resistance.
Collapse
Affiliation(s)
- Zoran Culig
- Division of Experimental Urology, Department of Urology, Innsbruck Medical University, Anichstrasse 35, A-6020 Innsbruck, Austria.
| | | |
Collapse
|
19
|
Functional domains of androgen receptor coactivator p44/Mep50/WDR77and its interaction with Smad1. PLoS One 2013; 8:e64663. [PMID: 23734213 PMCID: PMC3667176 DOI: 10.1371/journal.pone.0064663] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2012] [Accepted: 04/17/2013] [Indexed: 11/20/2022] Open
Abstract
p44/MEP50/WDR77 has been identified as a coactivator of androgen receptor (AR), with distinct growth suppression and promotion function in gender specific endocrine organs and their malignancies. We dissected the functional domains of p44 for protein interaction with transcription factors, transcriptional activation, as well as the functional domains in p44 related to its growth inhibition in prostate cancer. Using a yeast two-hybrid screen, we identified a novel transcription complex AR-p44-Smad1, confirmed for physical interaction by co-immunoprecipitaion and functional interaction with luciferase assays in human prostate cancer cells. Yeast two-hybrid assay revealed that the N-terminal region of p44, instead of the traditional WD40 domain at the C-terminus, mediates the interaction among p44, N-terminus of AR and full length Smad1. Although both N and C terminal domains of p44 are necessary for maximum AR transcriptional activation, the N terminal fragment of p44 alone maintains the basic effect on AR transcriptional activation. Cell proliferation assays with N- and C- terminal deletion mutations indicated that the central portion of p44 is required for nuclear p44 mediated prostate cancer growth inhibition.
Collapse
|
20
|
Furman D, Jojic V, Kidd B, Shen-Orr S, Price J, Jarrell J, Tse T, Huang H, Lund P, Maecker HT, Utz PJ, Dekker CL, Koller D, Davis MM. Apoptosis and other immune biomarkers predict influenza vaccine responsiveness. Mol Syst Biol 2013; 9:659. [PMID: 23591775 PMCID: PMC3658270 DOI: 10.1038/msb.2013.15] [Citation(s) in RCA: 155] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 03/07/2013] [Indexed: 12/17/2022] Open
Abstract
Despite the importance of the immune system in many diseases, there are currently no objective benchmarks of immunological health. In an effort to identifying such markers, we used influenza vaccination in 30 young (20-30 years) and 59 older subjects (60 to >89 years) as models for strong and weak immune responses, respectively, and assayed their serological responses to influenza strains as well as a wide variety of other parameters, including gene expression, antibodies to hemagglutinin peptides, serum cytokines, cell subset phenotypes and in vitro cytokine stimulation. Using machine learning, we identified nine variables that predict the antibody response with 84% accuracy. Two of these variables are involved in apoptosis, which positively associated with the response to vaccination and was confirmed to be a contributor to vaccine responsiveness in mice. The identification of these biomarkers provides new insights into what immune features may be most important for immune health.
Collapse
Affiliation(s)
- David Furman
- Department of Microbiology and Immunology, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Vladimir Jojic
- Department of Computer Science, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Brian Kidd
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Shai Shen-Orr
- Department of Immunology, Faculty of Medicine, Technion, Technion City, Haifa, Israel
| | - Jordan Price
- Department of Microbiology and Immunology, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Justin Jarrell
- Division of Immunology and Rheumatology, Department of Medicine, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Tiffany Tse
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Huang Huang
- Department of Microbiology and Immunology, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Peder Lund
- Department of Microbiology and Immunology, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Holden T Maecker
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Paul J Utz
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Palo Alto, CA, USA
- Division of Immunology and Rheumatology, Department of Medicine, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Cornelia L Dekker
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Palo Alto, CA, USA
- Department of Pediatrics, Division of Infectious Diseases, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Daphne Koller
- Department of Computer Science, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Mark M Davis
- Department of Microbiology and Immunology, School of Medicine, Stanford University, Palo Alto, CA, USA
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Palo Alto, CA, USA
- The Howard Hughes Medical Institute, Chevy Chase, MD, USA
| |
Collapse
|
21
|
FitzGerald LM, Zhang X, Kolb S, Kwon EM, Liew YC, Hurtado-Coll A, Knudsen BS, Ostrander EA, Stanford JL. Investigation of the relationship between prostate cancer and MSMB and NCOA4 genetic variants and protein expression. Hum Mutat 2012; 34:149-56. [PMID: 22887727 DOI: 10.1002/humu.22176] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2012] [Accepted: 07/19/2012] [Indexed: 12/21/2022]
Abstract
Two genome-wide association studies (GWAS) identified the β-microseminoprotein (MSMB) promoter SNP, rs10993994:C>T, as significantly associated with prostate cancer (PC) risk. Follow-up studies demonstrate that the variant allele directly affects expression of the MSMB-encoded protein, PSP94, and also suggest that it affects mRNA expression levels of an adjacent gene, NCOA4, which is involved in androgen receptor transactivation. In a population-based study of 1,323 cases and 1,268 age-matched controls, we found the NCOA4 SNP, rs7350420:T>C, was associated with a 15% reduction in PC risk, but the association was not significant after adjustment for the rs10993994:C>T genotype. Tumor tissue microarrays of 519 radical prostatectomy patients were used to measure PSP94 and NCOA4 protein expression. Taken together, these data confirm that the rs10993994:C>T variant allele is associated with decreased PSP94 expression, and the association is stronger in tumor compared to normal prostate tissue. No association was observed between rs10993994:C>T and NCOA4 expression, and only moderate associations were seen between two NCOA4 SNPs, rs10761618:T>C and rs7085433:G>A, and NCOA4 protein expression. These data indicate that the increase in PC risk associated with rs10993994:C>T is likely mediated by the variant's effect on PSP94 expression; however, this effect does not extend to NCOA4 in the data presented here.
Collapse
Affiliation(s)
- Liesel M FitzGerald
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Ligr M, Li Y, Logan SK, Taneja S, Melamed J, Lepor H, Garabedian MJ, Lee P. Mifepristone inhibits GRβ coupled prostate cancer cell proliferation. J Urol 2012; 188:981-8. [PMID: 22819113 DOI: 10.1016/j.juro.2012.04.102] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Indexed: 10/28/2022]
Abstract
PURPOSE The GR gene produces GRα and GRβ isoforms by alternative splicing of a C-terminal exon. GRα binds glucocorticoids, modulates transcription in a glucocorticoid dependent manner and has a growth inhibitory role in prostate cells. Due to this role glucocorticoids are often used to treat androgen independent prostate cancer. In contrast, GRβ has intrinsic transcriptional activity and binds mifepristone (RU486) but not glucocorticoids to control gene expression. To our knowledge the role of GRβ in prostate cell proliferation is unknown. MATERIALS AND METHODS We determined GRβ levels in various prostate cancer cell lines by reverse transcriptase-polymerase chain reaction and Western blot. The effect of GRβ on the kinetics of prostate cancer cell growth was determined by cell counting and flow cytometry upon mifepristone and dexamethasone treatment. Cell proliferation was also examined after siRNA mediated knockdown and over expression of GRβ. RESULTS GRβ mRNA and protein were up-regulated in LNCaP cells that over expressed the androgen receptor co-factor ARA70β. Treatment of LNCaP-ARA70β with mifepristone or siRNA targeting GRβ inhibited proliferation compared to that of parental LNCaP cells. The immortal but nontumorigenic RC165 prostate cell line and the tumorigenic DU145 prostate cell line with endogenous GRβ also showed partial growth reduction upon GRβ depletion but to a lesser extent than LNCaP-ARA70β cells. The growth stimulatory effect of ARA70β on LNCaP cells was partly GRβ dependent, as was the proliferation of RC165 cells and to a lesser extent of DU145 cells. CONCLUSIONS Results suggest that patients with a primary tumor that expresses GRβ and ARA70β may benefit from mifepristone.
Collapse
Affiliation(s)
- Martin Ligr
- Department of Pathology, New York University School of Medicine, New York, New York 10010, USA
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Kollara A, Brown TJ. Expression and function of nuclear receptor co-activator 4: evidence of a potential role independent of co-activator activity. Cell Mol Life Sci 2012; 69:3895-909. [PMID: 22562579 PMCID: PMC3492700 DOI: 10.1007/s00018-012-1000-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Revised: 04/13/2012] [Accepted: 04/17/2012] [Indexed: 12/22/2022]
Abstract
Nuclear receptor coactivator 4 (NcoA4), also known as androgen receptor-associated protein 70 (ARA70), was initially discovered as a component of Ret-Fused Gene expressed in a subset of papillary thyroid carcinomas. Subsequent studies have established NcoA4 as a coactivator for a variety of nuclear receptors, including peroxisome proliferator activated receptors α and γ, and receptors for steroid hormones, vitamins D and A, thyroid hormone, and aryl hydrocarbons. While human NcoA4 has both LXXLL and FXXLF motifs that mediate p160 coactivator nuclear receptor interactions, this ubiquitously expressed protein lacks clearly defined functional domains. Several studies indicate that NcoA4 localizes predominantly to the cytoplasm and affects ligand-binding specificity of the androgen receptor, which has important implications for androgen-independent prostate cancer. Two NcoA4 variants, which may exert differential activities, have been identified in humans. Recent studies suggest that NcoA4 may play a role in development, carcinogenesis, inflammation, erythrogenesis, and cell cycle progression that may be independent of its role as a receptor coactivator. This review summarizes what is currently known of the structure, expression, regulation, and potential functions of this unique protein in cancerous and non-cancerous pathologies.
Collapse
Affiliation(s)
- Alexandra Kollara
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, 25 Orde Street, 6-1001TB, Toronto, ON, M5T 3H7, Canada
| | | |
Collapse
|
24
|
Abstract
Specific thyroid cancer histotypes, such as papillary and medullary thyroid carcinoma, display genetic rearrangements or point mutations of the RET gene, resulting in its oncogenic conversion. The molecular mechanisms mediating RET rearrangement with other genes and the role of partner genes in tumorigenesis have been described. In addition, the RET protein has become a molecular target for medullary thyroid carcinoma treatment.
Collapse
Affiliation(s)
- Francesca Carlomagno
- *Francesca Carlomagno, Dipartimento di Biologia e Patologia Cellulare e Molecolare L. Califano, Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, IT–80123 Napoli (Italy), Tel. +39 081 746 3603, E-Mail
| |
Collapse
|
25
|
Androgen receptor co-activators in the regulation of cellular events in prostate cancer. World J Urol 2011; 30:297-302. [PMID: 22105110 DOI: 10.1007/s00345-011-0797-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Accepted: 11/07/2011] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVES Androgen receptor (AR) action in benign and malignant tissue is potentiated by a number of co-regulatory proteins that may interact with one or more receptor domains. With improvement of research methodologies, it became possible to detect a number of co-activators whose expression is increased in prostate cancer tissue. METHODS Manuscripts describing prostate cancer-relevant regulation of cellular events by co-activators are selected and summarized. RESULTS AR co-activators may regulate histone modification, proteasomal degradation, chaperones, sumoylation, chromatin remodeling, and cytoskeleton. Some of them (TIF-2) are up-regulated by androgens, whereas the expression of others increases during androgen ablation (p300, CBP, and Tip60). Most co-factors are important for the stimulation of cellular proliferation, although in some cases (ART-27), they act as tumor suppressors and are deleted in prostate cancer tissue. In addition to stimulating AR, some co-activators suppress apoptosis in prostate cancer cells that do not express the AR (p300 and SRC-3). It was recently shown that the inhibition of p300 slows down proliferation, stimulates apoptosis, and inhibits migration and invasion. CONCLUSIONS Co-factors whose down-regulation results in the alterations of multiple cellular functions may be valid targets for novel therapies in advanced prostate cancer.
Collapse
|
26
|
Kollara A, Ringuette MJ, Brown TJ. Dynamic distribution of nuclear coactivator 4 during mitosis: association with mitotic apparatus and midbodies. PLoS One 2011; 6:e22257. [PMID: 21814571 PMCID: PMC3144208 DOI: 10.1371/journal.pone.0022257] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Accepted: 06/21/2011] [Indexed: 11/19/2022] Open
Abstract
The cytoplasmic localization of Nuclear Receptor Coactivator 4 (NcoA4), also referred to as androgen receptor associated protein 70 (ARA70), indicates it may possess activities in addition to its role within the nucleus as a transcriptional enhancer. Towards identifying novel functions of NcoA4, we performed an in silico analysis of its amino acid sequence to identify potential functional domains and related proteins, and examined its subcellular distribution throughout the cell cycle. NcoA4 has no known or predicted functional or structural domains with the exception of an LxxLL and FxxLF nuclear receptor interaction motif and an N-terminal putative coiled-coil domain. Phylogenetic analysis indicated that NcoA4 has no paralogs and that a region referred to as ARA70-I family domain, located within the N-terminus and overlapping with the coiled-coil domain, is evolutionarily conserved in metazoans ranging from cnidarians to mammals. An adjacent conserved region, designated ARA70-II family domain, with no significant sequence similarity to the ARA70-I domain, is restricted to vertebrates. We demonstrate NcoA4 co-localizes with microtubules and microtubule organizing centers during prophase. Strong NcoA4 accumulation at the centrosomes was detected during interphase and telophase, with decreased levels at metaphase and anaphase. NcoA4 co-localized with tubulin and acetylated tubulin to the mitotic spindles during metaphase and anaphase, and to midbodies during telophase. Consistent with these observations, we demonstrated an interaction between NcoA4 and α-tubulin. Co-localization was not observed with microfilaments. These findings indicate a dynamic distribution of NcoA4 with components of the mitotic apparatus that is consistent with a potential non-transcriptional regulatory function(s) during cell division, which may be evolutionarily conserved.
Collapse
Affiliation(s)
- Alexandra Kollara
- Department of Obstetrics and Gynecology, Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Maurice J. Ringuette
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Theodore J. Brown
- Department of Obstetrics and Gynecology, Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
27
|
Wu X, Chen F, Sahin A, Albarracin C, Pei Z, Zou X, Singh B, Xu R, Daniels G, Li Y, Wei J, Blake M, Schneider RJ, Cowin P, Lee P. Distinct function of androgen receptor coactivator ARA70α and ARA70β in mammary gland development, and in breast cancer. Breast Cancer Res Treat 2011; 128:391-400. [PMID: 20814820 DOI: 10.1007/s10549-010-1131-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Accepted: 08/13/2010] [Indexed: 12/23/2022]
Abstract
Steroid receptor coactivators are important in regulating the function of the receptors in endocrine organ development and in cancers, including breast. Androgen receptor (AR) coactivator ARA70, was first identified as a gene fused to the ret oncogene and later characterized as an AR coactivator. We previously reported that the full length ARA70α functions as a tumor suppressor gene and that ARA70β functions as an oncogene in prostate cancer. Here we show that both ARA70α and ARA70β function as AR and estrogen receptor (ER) coactivators in breast cancer cells. However, ARA70α and ARA70β serve different functions in mammary gland development and breast cancer tumorigenesis. We observed hypoplastic development of mammary glands in MMTV driven ARA70α transgenic mice and overgrowth of mammary glands in ARA70β transgenic mice at virgin and pregnant stages. We determined that ARA70α inhibited cell proliferation, and that ARA70β promotes proliferation in MCF7 breast cancer cells. These effects were observed in hormone-free media, or in media with androgen or estrogen, though to varying degrees. Additionally, we observed that ARA70β strongly enhanced the invasive ability of MCF7 breast cancer cells in in vitro Matrigel assays. Significantly, decreased ARA70α expression is associated with increased tendency of breast cancer metastasis. In summary, ARA70α and ARA70β have distinct effects in mammary gland development and in the progression of breast cancer.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Breast Neoplasms/prevention & control
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/prevention & control
- Carcinoma, Ductal, Breast/secondary
- Cell Adhesion
- Cell Movement
- Cell Proliferation
- Female
- Humans
- Luciferases/metabolism
- Mammary Glands, Animal/cytology
- Mammary Glands, Animal/metabolism
- Mice
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Mice, Transgenic
- Nuclear Receptor Coactivators/genetics
- Nuclear Receptor Coactivators/metabolism
- Promoter Regions, Genetic/genetics
- RNA, Messenger/genetics
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Receptors, Estrogen/genetics
- Receptors, Estrogen/metabolism
- Response Elements
- Reverse Transcriptase Polymerase Chain Reaction
- Transfection
Collapse
Affiliation(s)
- Xinyu Wu
- Department of Pathology, New York University School of Medicine, 423 E. 23rd street, Room6139 N, New York, NY 10010, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Kauffman EC, Robinson BD, Downes MJ, Powell LG, Lee MM, Scherr DS, Gudas LJ, Mongan NP. Role of androgen receptor and associated lysine-demethylase coregulators, LSD1 and JMJD2A, in localized and advanced human bladder cancer. Mol Carcinog 2011; 50:931-44. [PMID: 21400613 DOI: 10.1002/mc.20758] [Citation(s) in RCA: 168] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Revised: 01/20/2011] [Accepted: 02/02/2011] [Indexed: 12/18/2022]
Abstract
Bladder cancer is approximately three times more common in men as compared to women. We and others have previously investigated the contribution of androgens and the androgen receptor (AR) to bladder cancer. JMJD2A and LSD1 are recently discovered AR coregulator proteins that mediate AR-dependent transcription via recently described histone lysine-demethylation (KDM) mechanisms. We used immunohistochemistry to examine JMJD2A, LSD1, and AR expression in 72 radical cystectomy specimens, resulting in evaluation of 129 tissue samples (59 urothelial carcinoma, 70 benign). We tested levels of these proteins for statistical association with clinicopathologic variables and patient survival. Expression of these markers was also assessed in human bladder cancer cell lines. The effects of pharmacological inhibition of LSD1 on the proliferation of these bladder cancer cells was determined. JMJD2A and AR levels were significantly lower in malignant versus benign urothelium, while increased LSD1 levels were observed in malignant urothelium relative to benign. A significant reduction in all three proteins occurred with cancer stage progression, including muscle invasion (JMJD2A/LSD1/AR), extravesical extension (JMJD2A/LSD1), and lymph node metastasis (JMJD2A/AR). Lower JMJD2A intensity correlated with additional poor prognostic features, including lymphovascular invasion, concomitant carcinoma in situ and tobacco usage, and predicted significantly worse overall survival. Pharmacological inhibition of LSD1 suppressed bladder cancer cell proliferation and androgen-induced transcription. Our results support a novel role for the AR-KDM complex in bladder cancer initiation and progression, identify JMJD2A as a promising prognostic biomarker, and demonstrate targeting of the KDM activity as an effective potential approach for bladder cancer growth inhibition.
Collapse
Affiliation(s)
- Eric C Kauffman
- Department of Urology, Weill Cornell Medical College, New York, New York 10065, USA
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Analysis of the 10q11 cancer risk locus implicates MSMB and NCOA4 in human prostate tumorigenesis. PLoS Genet 2010; 6:e1001204. [PMID: 21085629 PMCID: PMC2978684 DOI: 10.1371/journal.pgen.1001204] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2010] [Accepted: 10/13/2010] [Indexed: 12/21/2022] Open
Abstract
Genome-wide association studies (GWAS) have established a variant, rs10993994, on chromosome 10q11 as being associated with prostate cancer risk. Since the variant is located outside of a protein-coding region, the target genes driving tumorigenesis are not readily apparent. Two genes nearest to this variant, MSMB and NCOA4, are strong candidates for mediating the effects of rs109939934. In a cohort of 180 individuals, we demonstrate that the rs10993994 risk allele is associated with decreased expression of two MSMB isoforms in histologically normal and malignant prostate tissue. In addition, the risk allele is associated with increased expression of five NCOA4 isoforms in histologically normal prostate tissue only. No consistent association with either gene is observed in breast or colon tissue. In conjunction with these findings, suppression of MSMB expression or NCOA4 overexpression promotes anchorage-independent growth of prostate epithelial cells, but not growth of breast epithelial cells. These data suggest that germline variation at chromosome 10q11 contributes to prostate cancer risk by influencing expression of at least two genes. More broadly, the findings demonstrate that disease risk alleles may influence multiple genes, and associations between genotype and expression may only be observed in the context of specific tissue and disease states.
Collapse
|