1
|
Gilad Y, Shimon O, Han SJ, Lonard DM, O’Malley BW. Steroid receptor coactivators in Treg and Th17 cell biology and function. Front Immunol 2024; 15:1389041. [PMID: 38698860 PMCID: PMC11063348 DOI: 10.3389/fimmu.2024.1389041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 03/29/2024] [Indexed: 05/05/2024] Open
Abstract
Steroid receptor coactivators (SRCs) are master regulators of transcription that play key roles in human physiology and pathology. SRCs are particularly important for the regulation of the immune system with major roles in lymphocyte fate determination and function, macrophage activity, regulation of nuclear factor κB (NF-κB) transcriptional activity and other immune system biology. The three members of the p160 SRC family comprise a network of immune-regulatory proteins that can function independently or act in synergy with each other, and compensate for - or moderate - the activity of other SRCs. Recent evidence indicates that the SRCs are key participants in governing numerous aspects of CD4+ T cell biology. Here we review findings that establish the SRCs as essential regulators of regulatory T cells (Tregs) and T helper 17 (Th17) cells, with a focus on their crucial roles in Treg immunity in cancer and Treg-Th17 cell phenotypic plasticity.
Collapse
Affiliation(s)
- Yosi Gilad
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
- CoRegen, Inc., Baylor College of Medicine, Houston, TX, United States
| | - Ortal Shimon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
- CoRegen, Inc., Baylor College of Medicine, Houston, TX, United States
| | - Sang Jun Han
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
- CoRegen, Inc., Baylor College of Medicine, Houston, TX, United States
- Nuclear Receptor, Transcription and Chromatin Biology Program, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, United States
| | - David M. Lonard
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
- CoRegen, Inc., Baylor College of Medicine, Houston, TX, United States
- Nuclear Receptor, Transcription and Chromatin Biology Program, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, United States
| | - Bert W. O’Malley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
- CoRegen, Inc., Baylor College of Medicine, Houston, TX, United States
- Nuclear Receptor, Transcription and Chromatin Biology Program, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
2
|
Kiliti AJ, Sharif GM, Martin MB, Wellstein A, Riegel AT. AIB1/SRC-3/NCOA3 function in estrogen receptor alpha positive breast cancer. Front Endocrinol (Lausanne) 2023; 14:1250218. [PMID: 37711895 PMCID: PMC10498919 DOI: 10.3389/fendo.2023.1250218] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 08/14/2023] [Indexed: 09/16/2023] Open
Abstract
The estrogen receptor alpha (ERα) is a steroid receptor that is pivotal in the initiation and progression of most breast cancers. ERα regulates gene transcription through recruitment of essential coregulators, including the steroid receptor coactivator AIB1 (Amplified in Breast Cancer 1). AIB1 itself is an oncogene that is overexpressed in a subset of breast cancers and is known to play a role in tumor progression and resistance to endocrine therapy through multiple mechanisms. Here we review the normal and pathological functions of AIB1 in regard to its ERα-dependent and ERα-independent actions, as well as its genomic conservation and protein evolution. We also outline the efforts to target AIB1 in the treatment of breast cancer.
Collapse
Affiliation(s)
- Amber J. Kiliti
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, United States
- Department of Biochemistry, Molecular and Cellular Biology, Georgetown University, Washington, DC, United States
| | - Ghada M. Sharif
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, United States
| | - Mary Beth Martin
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, United States
- Department of Biochemistry, Molecular and Cellular Biology, Georgetown University, Washington, DC, United States
| | - Anton Wellstein
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, United States
| | - Anna T. Riegel
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, United States
| |
Collapse
|
3
|
Gilad Y, Lonard DM, O’Malley BW. Steroid receptor coactivators - their role in immunity. Front Immunol 2022; 13:1079011. [PMID: 36582250 PMCID: PMC9793089 DOI: 10.3389/fimmu.2022.1079011] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 11/18/2022] [Indexed: 12/14/2022] Open
Abstract
Steroid Receptor Coactivators (SRCs) are essential regulators of transcription with a wide range of impact on human physiology and pathology. In immunology, SRCs play multiple roles; they are involved in the regulation of nuclear factor-κB (NF-κB), macrophage (MΦ) activity, lymphoid cells proliferation, development and function, to name just a few. The three SRC family members, SRC-1, SRC-2 and SRC-3, can exert their immunological function either in an independent manner or act in synergy with each other. In certain biological contexts, one SRC family member can compensate for lack of activity of another member, while in other cases one SRC can exert a biological function that competes against the function of another family counterpart. In this review we illustrate the diverse biological functionality of the SRCs with regard to their role in immunity. In the light of recent development of SRC small molecule inhibitors and stimulators, we discuss their potential relevance as modulators of the immunological activity of the SRCs for therapeutic purposes.
Collapse
Affiliation(s)
- Yosi Gilad
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States,CoRegen, Inc., Baylor College of Medicine, Houston, TX, United States,*Correspondence: Yosi Gilad, ; David M. Lonard, ; Bert W. O’Malley,
| | - David M. Lonard
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States,CoRegen, Inc., Baylor College of Medicine, Houston, TX, United States,*Correspondence: Yosi Gilad, ; David M. Lonard, ; Bert W. O’Malley,
| | - Bert W. O’Malley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States,CoRegen, Inc., Baylor College of Medicine, Houston, TX, United States,*Correspondence: Yosi Gilad, ; David M. Lonard, ; Bert W. O’Malley,
| |
Collapse
|
4
|
Diao L, Li Y, Mei Q, Han W, Hu J. Retracted: AIB1 induces epithelial-mesenchymal transition in gastric cancer via the PI3K/AKT signaling. J Cell Biochem 2021; 122:926-933. [PMID: 31692102 DOI: 10.1002/jcb.29530] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 10/10/2019] [Indexed: 01/25/2023]
Abstract
Amplified in breast cancer 1 (AIB1) is overexpression in various cancers and promotes tumor cell proliferation, survival, and invasiveness. However, the role of AIB1 in the regulation of gastric cancer (GC) cell epithelial-mesenchymal transition (EMT) is still largely unclear. In the present study, immunohistochemistry showed that AIB1 was upregulated in our cohort of patients with GC and correlated with poor survival. Knockdown of AIB1 reduced the invasive ability of GC cells, downregulated the expression of epithelial cell marker E-cadherin, and upregulated mesenchymal cell marker vimentin. AIB1 overexpression elicited the opposite effect. PI-103, the inhibitor of the PI3K/AKT signaling, partially reversed AIB1 overexpression mediated a decrease in E-cadherin and an increase in vimentin. The present data demonstrated that AIB1 augmented the EMT via activation of PI3K/AKT signaling. In conclusion, our results suggested a novel role of AIB1 in GC invasion and EMT and raised the possibility of using this molecule as an indicator for GC treatment.
Collapse
Affiliation(s)
- Lei Diao
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yang Li
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Qiao Mei
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wei Han
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jing Hu
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
5
|
Kumar R. Role of conformational dynamics and flexibilities in the steroid receptor-coregulator protein complex formation. Gen Comp Endocrinol 2021; 309:113780. [PMID: 33882296 DOI: 10.1016/j.ygcen.2021.113780] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/21/2021] [Accepted: 04/09/2021] [Indexed: 10/21/2022]
Abstract
Understanding of the mechanisms of actions of the steroid hormone receptor (SHR)-coregulator (CoR) protein complexes in the gene regulations has revolutionized the field of molecular endocrinology and endocrine-related oncology. The discovery and characterization of steroid receptor coactivators (SRCs) and their ability to bind various transcription factors including SHRs to coordinate the regulation of multiple target genes highlights their importance as key coregulators in various cellular signaling crosstalks as well as therapeutic target for various endocrine-related disorders specifically endocrine cancers. The dynamic nature of the SHR-CoR multi-protein complexes indicate the critical role of conformational flexibilities within specific protein(s). In recent years, the importance of conformational dynamics of the SHRs in the intramolecular and intermolecular allosteric regulations mediated via their intrinsically disordered (ID) surfaces has been highlighted. In this review article, we have discussed the importance of ID conformations within the SRCs that may also be playing an important role in the formation/deformation of multi protein complexes involving SHRs and CoRs and subsequent target gene regulation.
Collapse
Affiliation(s)
- Raj Kumar
- Department of Biomedical Sciences, University of Houston - College of Medicine, Houston, TX, United States.
| |
Collapse
|
6
|
SRC-3, a Steroid Receptor Coactivator: Implication in Cancer. Int J Mol Sci 2021; 22:ijms22094760. [PMID: 33946224 PMCID: PMC8124743 DOI: 10.3390/ijms22094760] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/23/2021] [Accepted: 04/27/2021] [Indexed: 02/07/2023] Open
Abstract
Steroid receptor coactivator-3 (SRC-3), also known as amplified in breast cancer 1 (AIB1), is a member of the SRC family. SRC-3 regulates not only the transcriptional activity of nuclear receptors but also many other transcription factors. Besides the essential role of SRC-3 in physiological functions, it also acts as an oncogene to promote multiple aspects of cancer. This review updates the important progress of SRC-3 in carcinogenesis and summarizes its mode of action, which provides clues for cancer therapy.
Collapse
|
7
|
Palma AG, Soares Machado M, Lira MC, Rosa F, Rubio MF, Marino G, Kotsias BA, Costas MA. Functional relationship between CFTR and RAC3 expression for maintaining cancer cell stemness in human colorectal cancer. Cell Oncol (Dordr) 2021; 44:627-641. [PMID: 33616840 DOI: 10.1007/s13402-021-00589-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2021] [Indexed: 11/29/2022] Open
Abstract
PURPOSE CFTR mutations not only cause cystic fibrosis, but also increase the risk of colorectal cancer. A putative role of CFTR in colorectal cancer patients without cystic fibrosis has so far, however, not been investigated. RAC3 is a nuclear receptor coactivator that has been found to be overexpressed in several human tumors, and to be required for maintaining cancer stemness. Here, we investigated the functional relationship between CFTR and RAC3 for maintaining cancer stemness in human colorectal cancer. METHODS Cancer stemness was investigated by analysing the expression of stem cell markers, clonogenic growth and selective retention of fluorochrome, using stable transfection of shCFTR or shRAC3 in HCT116 colorectal cancer cells. In addition, we performed pathway enrichment and network analyses in both primary human colorectal cancer samples (TCGA, Xena platform) and Caco-2 colorectal cancer cells including (1) CD133+ or CD133- side populations and (2) CFTRwt or CFTRmut cells (ConsensusPathDB, STRING, Cytoscape, GeneMANIA). RESULTS We found that the CD133+ side population expresses higher levels of RAC3 and CFTR than the CD133- side population. RAC3 overexpression increased CFTR expression, whereas CFTR downregulation inhibited the cancer stem phenotype. CFTR mRNA levels were found to be increased in colorectal cancer samples from patients without cystic fibrosis compared to those with CFTR mutations, and this correlated with an increased expression of RAC3. The expression pattern of a gene set involved in inflammatory response and nuclear receptor modulation in CD133+ Caco-2 cells was found to be shared with that in CFTRwt Caco-2 cells. These genes may contribute to colorectal cancer development. CONCLUSIONS CFTR may play a non-tumor suppressor role in colorectal cancer development and maintenance involving enhancement of the expression of a set of genes related to cancer stemness and development in patients without CFTR mutations.
Collapse
Affiliation(s)
- Alejandra Graciela Palma
- Laboratorio de Biología Molecular y Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150. Cuerpo II, Piso 1, C1427ARO, Buenos Aires, Argentina
| | - Mileni Soares Machado
- Laboratorio de Biología Molecular y Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150. Cuerpo II, Piso 1, C1427ARO, Buenos Aires, Argentina
| | - María Cecilia Lira
- Laboratorio de Biología Molecular y Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150. Cuerpo II, Piso 1, C1427ARO, Buenos Aires, Argentina
| | - Francisco Rosa
- Laboratorio de Biología Molecular y Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150. Cuerpo II, Piso 1, C1427ARO, Buenos Aires, Argentina
| | - María Fernanda Rubio
- Laboratorio de Biología Molecular y Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150. Cuerpo II, Piso 1, C1427ARO, Buenos Aires, Argentina.,CONICET, Buenos Aires, Argentina
| | - Gabriela Marino
- CONICET, Buenos Aires, Argentina.,Laboratorio de Canales Iónicos, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150, C1427ARO, Buenos Aires, Argentina
| | - Basilio Aristidis Kotsias
- CONICET, Buenos Aires, Argentina.,Laboratorio de Canales Iónicos, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150, C1427ARO, Buenos Aires, Argentina
| | - Mónica Alejandra Costas
- Laboratorio de Biología Molecular y Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150. Cuerpo II, Piso 1, C1427ARO, Buenos Aires, Argentina. .,CONICET, Buenos Aires, Argentina.
| |
Collapse
|
8
|
Jahangiri R, Mosaffa F, Emami Razavi A, Teimoori-Toolabi L, Jamialahmadi K. PAX2 promoter methylation and AIB1 overexpression promote tamoxifen resistance in breast carcinoma patients. J Oncol Pharm Pract 2021; 28:310-325. [PMID: 33509057 DOI: 10.1177/1078155221989404] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
INTRODUCTION Disease recurrence is an important obstacle in estrogen receptor positive (ER+) tamoxifen treated breast carcinoma patients. Tamoxifen resistance-related molecular mechanisms are not fully understood. Alteration in DNA methylation which contributes to transcriptional regulation of cancer-related genes plays a crucial role in tamoxifen response. In the present study, the contribution of promoter methylation and mRNA expression of PAX2 and AIB1 in the development of breast carcinoma and tamoxifen refractory was assessed. METHODS Methylation specific-high resolution melting (MS-HRM) analysis and Real-time quantitative PCR (RT-qPCR) experiment were performed to analyze the promoter methylation and mRNA expression levels of PAX2 and AIB1 genes in 102 breast tumors and adjacent normal breast specimens. RESULTS We indicated that PAX2 expression is decreased in breast tissues due to hypermethylation in its promoter region. Compared to the adjacent normal tissues, the tumors exhibited significantly lower relative mRNA levels of PAX2 and increased expression of AIB1. Aberrant promoter methylation of PAX2 and overexpression of AIB1 was observed in tamoxifen resistance patients compared to the sensitive ones. Cox regression analysis exhibited that the increased promoter methylation status of PAX2 and overexpression of AIB1 remained as unfavorable identifiers which influence patients' survival independently. CONCLUSIONS Our results revealed that the aberration in PAX2 promoter methylation and AIB1 overexpression are associated with the tamoxifen response in breast carcinoma patients. Further research is needed to demonstrate the potential of using PAX2 and AIB1 expression and their methylation-mediated regulation as predictive or prognostic biomarkers or as a new target therapy for better disease management.
Collapse
Affiliation(s)
- Rosa Jahangiri
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Mosaffa
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | | | - Khadijeh Jamialahmadi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
9
|
Li L, Bao J, Wang H, Lei JH, Peng C, Zeng J, Hao W, Zhang X, Xu X, Yu C, Deng CX, Chen Q. Upregulation of amplified in breast cancer 1 contributes to pancreatic ductal adenocarcinoma progression and vulnerability to blockage of hedgehog activation. Theranostics 2021; 11:1672-1689. [PMID: 33408774 PMCID: PMC7778610 DOI: 10.7150/thno.47390] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 10/19/2020] [Indexed: 12/13/2022] Open
Abstract
Background: Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive and devastating cancers without effective treatments. Amplified in breast cancer 1 (AIB1) is a member of the steroid receptor coactivator family that mediates the transcriptional activities of nuclear receptors. While AIB1 is associated with the initiation and progression of multiple cancers, the mechanism by which AIB1 contributes to PDAC progression remains unknown. In this study, we aimed to explore the role of AIB1 in the progression of PDAC and elucidate the underlying mechanisms. Methods: The clinical significance and mRNA level of AIB1 in PDAC were studied by database analysis. To demonstrate whether AIB1 mediates the malignant features of PDAC cells, namely, proliferation, migration, invasion, we performed real-time PCR and Western blot analysis, established xenograft models and used in vivo metastasis assay. With insights into the mechanism of AIB1, we performed RNA sequencing (Seq), ChIP-Seq, luciferase reporter assays and pull-down assays. Furthermore, we analyzed the relationship between AIB1 expression and its target expression in PDAC cells and patients and explored whether PDAC cells with high AIB1 levels are sensitive to inhibitors of its target. Results: We found that AIB1 was significantly upregulated in PDAC and associated with its malignancy. Silencing AIB1 impaired hedgehog (Hh) activation by reducing the expression of smoothened (SMO), leading to cell cycle arrest and the inhibition of PDAC cell proliferation. In addition, AIB1, via upregulation of integrin αv (ITGAV) expression, promoted extracellular matrix (ECM) signaling, which played an important role in PDAC progression. Further studies showed that AIB1 preferably bound to AP-1 related elements and served as a coactivator for enhancing the transcriptional activity of MafB, which promoted the expression of SMO and ITGAV. PDAC cells with high AIB1 levels were sensitive to Hh signaling inhibitors, suggesting that blocking Hh activation is an effective treatment against PDAC with high AIB1 expression. Conclusions: These findings reveal that AIB1 is a crucial oncogenic regulator associated with PDAC progression via Hh and ECM signaling and suggest potential therapeutic targets for PDAC treatment.
Collapse
Affiliation(s)
- Licen Li
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Jiaolin Bao
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Haitao Wang
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Josh Haipeng Lei
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Cheng Peng
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Jianming Zeng
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Wenhui Hao
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Xu Zhang
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Xiaoling Xu
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Chundong Yu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361012, China
| | - Chu-Xia Deng
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Qiang Chen
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| |
Collapse
|
10
|
Gromisch C, Qadan M, Machado MA, Liu K, Colson Y, Grinstaff MW. Pancreatic Adenocarcinoma: Unconventional Approaches for an Unconventional Disease. Cancer Res 2020; 80:3179-3192. [PMID: 32220831 PMCID: PMC7755309 DOI: 10.1158/0008-5472.can-19-2731] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 02/08/2020] [Accepted: 03/24/2020] [Indexed: 12/16/2022]
Abstract
This review highlights current treatments, limitations, and pitfalls in the management of pancreatic cancer and discusses current research in novel targets and drug development to overcome these clinical challenges. We begin with a review of the clinical landscape of pancreatic cancer, including genetic and environmental risk factors, as well as limitations in disease diagnosis and prevention. We next discuss current treatment paradigms for pancreatic cancer and the shortcomings of targeted therapy in this disease. Targeting major driver mutations in pancreatic cancer, such as dysregulation in the KRAS and TGFβ signaling pathways, have failed to improve survival outcomes compared with nontargeted chemotherapy; thus, we describe new advances in therapy such as Ras-binding pocket inhibitors. We then review next-generation approaches in nanomedicine and drug delivery, focusing on preclinical advancements in novel optical probes, antibodies, small-molecule agents, and nucleic acids to improve surgical outcomes in resectable disease, augment current therapies, expand druggable targets, and minimize morbidity. We conclude by summarizing progress in current research, identifying areas for future exploration in drug development and nanotechnology, and discussing future prospects for management of this disease.
Collapse
Affiliation(s)
- Christopher Gromisch
- Departments of Pharmacology and Experimental Therapeutics, Biomedical Engineering, and Chemistry, Boston University, Boston, Massachusetts
| | - Motaz Qadan
- Division of Surgical Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Mariana Albuquerque Machado
- Departments of Pharmacology and Experimental Therapeutics, Biomedical Engineering, and Chemistry, Boston University, Boston, Massachusetts
| | - Kebin Liu
- Department of Biochemistry and Molecular Biology and Georgia Cancer Center, Medical College of Georgia, Augusta, Georgia
| | - Yolonda Colson
- Division of Thoracic Surgery, Massachusetts General Hospital, Boston, Massachusetts
| | - Mark W Grinstaff
- Departments of Pharmacology and Experimental Therapeutics, Biomedical Engineering, and Chemistry, Boston University, Boston, Massachusetts.
| |
Collapse
|
11
|
Beltrán-Gastélum M, Esteban-Fernández de Ávila B, Gong H, Venugopalan PL, Hianik T, Wang J, Subjakova V. Rapid Detection of AIB1 in Breast Cancer Cells Based on Aptamer-Functionalized Nanomotors. Chemphyschem 2019; 20:3177-3180. [PMID: 31639248 DOI: 10.1002/cphc.201900844] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 10/15/2019] [Indexed: 01/29/2023]
Abstract
Herein, we report ultrasound-propelled graphene-oxide coated gold nanowire motors, functionalized with fluorescein-labeled DNA aptamers (FAM-AIB1-apt), for qualitative detection of overexpressed AIB1 oncoproteins in MCF-7 breast cancer cells. The movement of nanomotors under the ultrasound field facilitated intracellular uptake and resulted in a faster aptamer binding with the target protein and thus faster fluorescence recovery. The propulsion behavior of the aptamer functionalized nanomotors greatly enhanced the fluorescence intensity compared to static conditions. The new aptamer@nanomotor-based strategy offers considerable potential for further development of sensing methodologies towards diagnosis of breast cancer.
Collapse
Affiliation(s)
- Mara Beltrán-Gastélum
- Department of Nanoengineering, University of California San Diego, La Jolla, California, 92093, United States
| | | | - Hua Gong
- Department of Nanoengineering, University of California San Diego, La Jolla, California, 92093, United States
| | - Pooyath Lekshmy Venugopalan
- Department of Nanoengineering, University of California San Diego, La Jolla, California, 92093, United States
| | - Tibor Hianik
- Department of Nuclear Physics and Biophysics Faculty of Mathematics, Physics and Informatics, Comenius University, Mlynská dolina F1, 842 48, Bratislava, Slovakia
| | - Joseph Wang
- Department of Nanoengineering, University of California San Diego, La Jolla, California, 92093, United States
| | - Veronika Subjakova
- Department of Nanoengineering, University of California San Diego, La Jolla, California, 92093, United States
- Department of Nuclear Physics and Biophysics Faculty of Mathematics, Physics and Informatics, Comenius University, Mlynská dolina F1, 842 48, Bratislava, Slovakia
| |
Collapse
|
12
|
Bijou I, Wang J. Evolving trends in pancreatic cancer therapeutic development. ANNALS OF PANCREATIC CANCER 2019; 2:17. [PMID: 33089149 PMCID: PMC7575122 DOI: 10.21037/apc.2019.09.01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Despite advances in translational research, the overall 5-year survival for pancreatic cancer remains dismal and with rising incidence pancreatic cancer is predicted to be the second leading cause of cancer death for many developed countries. Surgical intervention followed by cytotoxic chemotherapy are currently the best options for treatment, but disease recurrence is very common. Efforts to develop new therapeutic agents and delivery systems are necessary to achieve better clinical efficacy with less toxicity. Promising prospects are arising with new preclinical and clinical therapeutic strategies using small molecule targeted therapies, RNAi, stromal therapies, and immunotherapies. With a better understanding of the biology to aid target selection and discovery of biomarkers to aid precision medicine, better opportunities will evolve to shape the therapeutic landscape, enhance patient quality of life and increase overall survival.
Collapse
Affiliation(s)
- Imani Bijou
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | - Jin Wang
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| |
Collapse
|
13
|
Song X, Chen H, Zhang C, Yu Y, Chen Z, Liang H, Van Buren G, McElhany AL, Fisher WE, Lonard DM, O'Malley BW, Wang J. SRC-3 inhibition blocks tumor growth of pancreatic ductal adenocarcinoma. Cancer Lett 2019; 442:310-319. [PMID: 30423406 PMCID: PMC6311429 DOI: 10.1016/j.canlet.2018.11.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 11/06/2018] [Accepted: 11/07/2018] [Indexed: 12/28/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant and lethal disease with few treatment options. Steroid receptor coactivator-3 (SRC-3, also known as NCOA3, AIB1, pCIP, ACTR, RAC3, TRAM1) sits at the nexus of many growth signaling pathways and has been pursued as a therapeutic target for breast, prostate and lung cancers. In this study, we find that SRC-3 is overexpressed in PDAC and inversely correlates with patient overall survival. Knockdown of SRC-3 reduces pancreatic cancer cell proliferation, migration and invasion in vitro. Additionally, inhibition of SRC-3 using either shRNA or a small molecule inhibitor can significantly inhibit tumor growth in orthotopic pancreatic cancer mouse models. Collectively, this study establishes SRC-3 as a promising therapeutic target for pancreatic cancer treatment.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/pathology
- Carcinoma, Pancreatic Ductal/therapy
- Cell Line, Tumor
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Mice, Nude
- Mice, SCID
- Neoplasm Invasiveness
- Nuclear Receptor Coactivator 3/antagonists & inhibitors
- Nuclear Receptor Coactivator 3/genetics
- Nuclear Receptor Coactivator 3/metabolism
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/therapy
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- RNAi Therapeutics
- Signal Transduction
- Tumor Burden/drug effects
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Xianzhou Song
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Hui Chen
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Chengwei Zhang
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yang Yu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Zhongyuan Chen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA; Department of Statistics, Rice University, Houston, TX, 77030, USA
| | - Han Liang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA; Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - George Van Buren
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Amy L McElhany
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - William E Fisher
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - David M Lonard
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.
| | - Bert W O'Malley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.
| | - Jin Wang
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX, 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
14
|
Hao Y, Gao Y, Wu Y, An C. The AIB1siRNA-loaded hyaluronic acid-assembled PEI/heparin/Ca2+ nanocomplex as a novel therapeutic strategy in lung cancer treatment. Int J Mol Med 2018; 43:861-867. [PMID: 30535446 PMCID: PMC6317651 DOI: 10.3892/ijmm.2018.4014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 11/16/2018] [Indexed: 12/29/2022] Open
Abstract
In the present study, AIB1siRNA‑loaded polyethyleneimine (PEI)/heparin/Ca2+ nanoparticles (NPs) were successfully prepared and evaluated for their efficacy in lung cancer cells. The results demonstrated that the PEI and heparin complex reduced the toxic effect in cancer cells while maintaining its transfection efficiency. A nanosized particle of ~25 nm was formulated and siRNA was demonstrated to possess excellent binding efficiency in the particles. Confocal microscopy revealed that fluorescein‑labeled (FAM)‑small interfering (si)RNA dissociated from the HA‑PEI/heparin/Ca2+/siRNA (CPH‑siH) NPs and exhibited maximum fluorescence in the cytoplasm, which was important in elucidating its post‑transcriptional activity. CPH‑siH NPs exhibited a typical concentration‑dependent toxicity in cancer cells. Blank PEI/heparin/Ca2+ did not induce any toxicity in cancer cells, indicating its safety and lack of side effects. CPH‑siH (100 nm) induced the maximum apoptosis of cancer cells with nearly ~35% of cells in the early and late apoptosis stages. The expression of the nuclear receptor coactivator 3 (NCOA3, also known as AIB1) protein was knocked down in a concentration‑dependent manner, demonstrating the potent activity of AIB1siRNA in cancer cells. Together, these results indicated that HA‑PEI/heparin/Ca2+ NPs may be a promising carrier for the anticancer activity of AIB1siRNA in lung cancer cells.
Collapse
Affiliation(s)
- Ying Hao
- Department of Pathology, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, Xinjiang 832003, P.R. China
| | - Yongsheng Gao
- Department of Pathology, Shandong Cancer Hospital Affiliated to Shandong University, Jinan, Shandong 250117, P.R. China
| | - Yedan Wu
- Department of Respiratory Medicine, Yanbian University Hospital, Yanji, Jilin 133000, P.R. China
| | - Changshan An
- Department of Respiratory Medicine, Yanbian University Hospital, Yanji, Jilin 133000, P.R. China
| |
Collapse
|
15
|
Machado MS, Rosa FD, Lira MC, Urtreger AJ, Rubio MF, Costas MA. The inflammatory cytokine TNF contributes with RAC3-induced malignant transformation. EXCLI JOURNAL 2018; 17:1030-1042. [PMID: 30585274 PMCID: PMC6298201 DOI: 10.17179/excli2018-1759] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 10/16/2018] [Indexed: 12/16/2022]
Abstract
RAC3 is a coactivator of steroid receptors and NF-κB. It is usually overexpressed in several tumors, contributes to maintain cancer stem cells and also to induce them when is overexpressed in non-tumoral cells. In this work, we investigated whether the inflammatory cytokine TNF may contribute to the transforming effects of RAC3 overexpression in the non-tumoral HEK293 cell line. The study model included the HEK293 tumoral transformed cell line constitutively overexpressing RAC3 by stable transfection and control non-tumoral cells transfected with an empty vector. The HeLa and T47D tumoral cells that naturally overexpress RAC3 were used as positive control. We found that TNF potentiated RAC3-induced mesenchymal transition, involving an increased E-Cadherin downregulation, Vimentin and SNAIL upregulation and enhanced migratory behavior. Moreover, concerning the molecular mechanisms by which TNF potentiates the RAC3 transforming action, they involve the IKK activation, which in addition induced the β-Catenin transactivation. Our results demonstrate that although RAC3 overexpression could be a signal strong enough to induce cancer stem cells, the inflammatory microenvironment may be playing a key role contributing to the migratory and invasive phenotype required for metastasis and cancer persistence.
Collapse
Affiliation(s)
- Mileni Soares Machado
- Laboratorio de Biología Molecular y Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150, C1427ARO Buenos Aires, Argentina
| | - Francisco D Rosa
- Laboratorio de Biología Molecular y Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150, C1427ARO Buenos Aires, Argentina
| | - María C Lira
- Laboratorio de Biología Molecular y Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150, C1427ARO Buenos Aires, Argentina
| | - Alejandro J Urtreger
- Universidad de Buenos Aires, Instituto de Oncología Ángel H. Roffo, Área Investigación, Av. San Martín 5481, C1417DTB Buenos Aires, Argentina.,Member of the Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)
| | - María F Rubio
- Laboratorio de Biología Molecular y Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150, C1427ARO Buenos Aires, Argentina.,Member of the Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)
| | - Mónica A Costas
- Laboratorio de Biología Molecular y Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150, C1427ARO Buenos Aires, Argentina.,Member of the Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)
| |
Collapse
|
16
|
Li S, Zhang H, Yu Y, Liu M, Guo D, Zhang X, Zhang J. Imbalanced expression pattern of steroid receptor coactivator-1 and -3 in liver cancer compared with normal liver: An immunohistochemical study with tissue microarray. Oncol Lett 2018; 16:6339-6348. [PMID: 30405769 PMCID: PMC6202514 DOI: 10.3892/ol.2018.9443] [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] [Received: 09/29/2016] [Accepted: 08/10/2018] [Indexed: 12/02/2022] Open
Abstract
Steroids affect normal and pathological functions of the liver through receptors, which require coactivators for their transcriptional activation. Steroid receptor coactivator (SRC)-1 and SRC-3 have been demonstrated to be regulated in numerous cancers; however, their expression profiles in liver cancer including hepatocellular carcinoma (HCC) and cholangiocellular carcinoma (CCC) remain unclear. Using tissue microarray immunohistochemistry, normal liver tissue and HCC tissue exhibited immunoreactivity of SRC-1, which were predominantly localized within extranuclear components; in CCC, they were detected within the cell nuclei; SRC-3 was also detected in the cell nuclei. Furthermore, no altered expression of SRC-1 and SRC-3 was observed in liver cancer compared with normal liver tissue; however, in CCC, the expression of SRC-3 was significantly increased compared with that detected in HCC. Importantly, although expression of SRC-1 and SRC-3 did not reveal any significant differences (30 vs. 40%) in normal liver tissue, HCC and CCC expression of SRC-1 was significantly decreased compared with that of SRC-3 (9.3 vs. 36%, and 6.7 vs. 67.7% for HCC and CCC, respectively). Further comparative analysis revealed that this discrepancy was detected in males with liver cancer, across all ages of HCC cases, younger CCC cases and all stages of liver cancer. The results suggested the presence of an imbalanced expression pattern of SRC-1 and SRC-3 from normal liver tissue to liver cancer (decreased SRC-1 and increased SRC-3), which may affect hepatic function and therefore promote liver carcinogenesis.
Collapse
Affiliation(s)
- Shan Li
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, P.R. China.,Cadet Brigade, Third Military Medical University, Chongqing 400038, P.R. China
| | - Huiyan Zhang
- Department of Infectious Diseases, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Yanlan Yu
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, P.R. China.,Cadet Brigade, Third Military Medical University, Chongqing 400038, P.R. China
| | - Mengying Liu
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, P.R. China
| | - Deyu Guo
- Institute of Pathology, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Xuqing Zhang
- Department of Infectious Diseases, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Jiqiang Zhang
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, P.R. China
| |
Collapse
|
17
|
Sikdar N, Saha G, Dutta A, Ghosh S, Shrikhande SV, Banerjee S. Genetic Alterations of Periampullary and Pancreatic Ductal Adenocarcinoma: An Overview. Curr Genomics 2018; 19:444-463. [PMID: 30258276 PMCID: PMC6128383 DOI: 10.2174/1389202919666180221160753] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 01/31/2018] [Accepted: 02/01/2018] [Indexed: 02/06/2023] Open
Abstract
Pancreatic Ductal AdenoCarcinoma (PDAC) is one of the most lethal malignancies of all solid cancers. Precancerous lesions for PDAC include PanIN, IPMNs and MCNs. PDAC has a poor prognosis with a 5-year survival of approximately 6%. Whereas Periampulary AdenoCarcinoma (PAC) having four anatomic subtypes, pancreatic, Common Bile Duct (CBD), ampullary and duodenum shows relative better prognosis. The highest incidence of PDAC has been reported with black with respect to white population. Similarly, incidence rate of PAC also differs with different ethnic populations. Several lifestyle, environmental and occupational exposures including long-term diabetes, obesity, and smoking, have been linked to PDAC, however, for PAC the causal risk factors were poorly described. It is now clear that PDAC and PAC are a multi-stage process resulting from the accumulation of genomic alterations in the somatic DNA of normal cells as well as inherited mutations. Approximately 10% of PDAC have a familial inheritance. Germline mutations in CDKN2A, BRCA2, STK11, PALB2, PRSS1, etc., as well as certain syndromes have been well associated with predisposition to PDAC. KRAS, CDKN2A, TP53 and SMAD4 are the 4 "mountains" (high-frequency driver genes) which have been known to earliest somatic alterations for PDAC while relatively less frequent in PAC. Our understanding of the molecular carcinogenesis has improved in the last few years due to extensive research on PDAC which was not well explored in case of PAC. The genetic alterations that have been identified in PDAC and different subgroups of PAC are important implications for the development of genetic screening test, early diagnosis, and prognostic genetic markers. The present review will provide a brief overview of the incidence and prevalence of PDAC and PAC, mainly, increased risk in India, the several kinds of risk factors associated with the diseases as well as required genetic alterations for disease initiation and progression.
Collapse
Affiliation(s)
- Nilabja Sikdar
- Address correspondence to this author at the Human Genetics Unit, Indian Statistical Institute, 203, B.T. Road Kolkata 700108, India; Tel (1): +91-33
-25773240 (L); (2): +91-9830780397 (M); Fax: +91 33 35773049;, E-mail:
| | | | | | | | | | | |
Collapse
|
18
|
Panelo LC, Machado MS, Rubio MF, Jaworski F, Alvarado CV, Paz LA, Urtreger AJ, Vazquez E, Costas MA. High RAC3 expression levels are required for induction and maintaining of cancer cell stemness. Oncotarget 2018; 9:5848-5860. [PMID: 29464039 PMCID: PMC5814179 DOI: 10.18632/oncotarget.23635] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 12/04/2017] [Indexed: 01/10/2023] Open
Abstract
RAC3 is a transcription coactivator, usually overexpressed in several tumors and required to maintain the pluripotency in normal stem cells. In this work we studied the association between RAC3 overexpression on cancer cell stemness and the capacity of this protein to induce cancer stem properties in non tumoral cells. We performed in vitro and in vivo experiments using two strategies: by overexpressing RAC3 in the non tumoral cell line HEK293 and by silencing RAC3 in the human colorectal epithelial cell line HCT116 by transfection. Furthermore, we analysed public repository microarrays data from human colorectal tumors in different developmental stages. We found that RAC3 overexpression was mainly associated to CD133+ side-population of colon cancer cells and also to early and advanced stages of colon cancer, involving increased expression of mesenchymal and stem markers. In turn, RAC3 silencing induced diminished tumoral properties and cancer stem cells as determined by Hoechst efflux, tumorspheres and clonogenic growth, which correlated with decreased Nanog and OCT4 expression. In non tumoral cells, RAC3 overexpression induced tumoral transformation; mesenchymal phenotype and stem markers expression. Moreover, these transformed cells generated tumors in vivo. Our results demonstrate that RAC3 is required for maintaining and induction of cancer cell stemness.
Collapse
Affiliation(s)
- Laura C Panelo
- Laboratorio de Biología Moleculary Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, C1427ARO Buenos Aires, Argentina
| | - Mileni Soares Machado
- Laboratorio de Biología Moleculary Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, C1427ARO Buenos Aires, Argentina
| | - María F Rubio
- Laboratorio de Biología Moleculary Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, C1427ARO Buenos Aires, Argentina.,Laboratorio de Inflamación y Cancer, IQUIBICEN-CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, C1428EGA Buenos Aires, Argentina.,Argentine National Research Council (CONICET), C1425FQB Godoy Cruz (CABA), República Argentina
| | - Felipe Jaworski
- Laboratorio de Inflamación y Cancer, IQUIBICEN-CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, C1428EGA Buenos Aires, Argentina
| | - Cecilia V Alvarado
- Laboratorio de Biología Moleculary Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, C1427ARO Buenos Aires, Argentina
| | - Leonardo A Paz
- Laboratorio de Anatomía Patológica, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires, C1427ARO Buenos Aires, Argentina
| | - Alejandro J Urtreger
- Laboratorio de Anatomía Patológica, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires, C1427ARO Buenos Aires, Argentina.,Universidad de Buenos Aires, Instituto de Oncología "Angel H Roffo", Area de Investigación, C1417DTB Buenos Aires, Argentina.,Argentine National Research Council (CONICET), C1425FQB Godoy Cruz (CABA), República Argentina
| | - Elba Vazquez
- Laboratorio de Inflamación y Cancer, IQUIBICEN-CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, C1428EGA Buenos Aires, Argentina.,Argentine National Research Council (CONICET), C1425FQB Godoy Cruz (CABA), República Argentina
| | - Mónica A Costas
- Laboratorio de Biología Moleculary Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, C1427ARO Buenos Aires, Argentina.,Argentine National Research Council (CONICET), C1425FQB Godoy Cruz (CABA), República Argentina
| |
Collapse
|
19
|
Chen R, Zhang Y, Zhang C, Wu H, Yang S. miR-137 inhibits the proliferation of human non-small cell lung cancer cells by targeting SRC3. Oncol Lett 2017; 13:3905-3911. [PMID: 28521488 DOI: 10.3892/ol.2017.5904] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 01/17/2017] [Indexed: 12/21/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is the most common type of lung cancer. The results of the present study demonstrate that high expression of microRNA (miR)-137 and low expression of steroid receptor coactivator-3 (SRC3) had a significant negative correlation in 40 NSCLC tissue samples. In addition, cell colony formation and proliferation was significantly reduced in miR-137-transfected A549 and NCI-H838 cells compared with scramble-transfected NSCLC cell lines. miR-137 was identified to induce G1/S cell cycle arrest and dysregulate the mRNA expression of cell cycle-associated proteins (proliferating cell nuclear antigen, cyclin E, cyclin A1, cyclin A2 and p21) in NSCLC cells. Notably, miR-137 could significantly suppress SRC3 3' untranslated region (UTR) luciferase-reporter activity, an effect that was not detectable when the putative 3'-UTR target-site was mutated, further clarifying the molecular mechanisms underlying the role of miR-137 in NSCLC. In conclusion, the results of the present study suggest that miR-137 suppresses NSCLC cell proliferation by partially targeting SRC3.
Collapse
Affiliation(s)
- Ruilin Chen
- Department of Respiratory Medicine, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Yongqing Zhang
- Department of Respiratory Medicine, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Chengcheng Zhang
- Department of Respiratory Medicine, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Hua Wu
- Department of Respiratory Medicine, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Shumei Yang
- Department of Respiratory Medicine, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| |
Collapse
|
20
|
Tumas J, Kvederaviciute K, Petrulionis M, Kurlinkus B, Rimkus A, Sakalauskaite G, Cicenas J, Sileikis A. Metabolomics in pancreatic cancer biomarkers research. Med Oncol 2016; 33:133. [PMID: 27807722 DOI: 10.1007/s12032-016-0853-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 10/27/2016] [Indexed: 12/14/2022]
Abstract
Pancreatic cancer is one of the worst prognoses of all malignancies. More than 40,000 deaths a year from this disease are observed in European Union alone. The only possibly curative treatment of pancreatic cancer is surgery, yet only 15-20% of patients have operable disease and even patients, which go through surgery and adjuvant chemotherapy, survival is less than 30%. The sensitive and specific biomarkers which could be used for the advance of early diagnostics are needed and constantly researched. Metabolomics is a technology which analyzes the concentrations of low-molecular-weight metabolites (the metabolome) has lately effectively developed due to the improvements in analytical technology. Metabolome analysis can be a one of the useful approaches for the biomarker discovery and disease diagnosis. Here we discuss recent discoveries in the field of pancreatic cancer metabolomics as well as the most promising biomarkers for diagnostics, prognosis and prediction.
Collapse
Affiliation(s)
- Jaroslav Tumas
- Center of Abdominal Surgery, Vilnius University Hospital, Santariskiu Klinikos Santariskiu str. 2, 08661, Vilnius, Lithuania
| | - Kotryna Kvederaviciute
- Institute of Biotechnology, Vilnius University, Saulėtekio ave. 7, 01222, Vilnius, Lithuania
| | - Marius Petrulionis
- Center of Abdominal Surgery, Vilnius University Hospital, Santariskiu Klinikos Santariskiu str. 2, 08661, Vilnius, Lithuania
| | - Benediktas Kurlinkus
- Center of Hepatology, Gastroenterology and Dietology, Vilnius University Hospital, Santariskiu Klinikos Santariskiu str. 2, 08661, Vilnius, Lithuania
| | - Arnas Rimkus
- Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | | | - Jonas Cicenas
- Vetsuisse Faculty, Institute of Animal Pathology, University of Bern, 3012, Bern, Switzerland. .,MAP Kinase Resource, Bioinformatics, Melchiorstrasse 9, 3027, Bern, Switzerland. .,Proteomics Centre, Institute of Biochemistry, Vilnius University, 08662, Vilnius, Lithuania.
| | - Audrius Sileikis
- Center of Abdominal Surgery, Vilnius University Hospital, Santariskiu Klinikos Santariskiu str. 2, 08661, Vilnius, Lithuania. .,Center of Hepatology, Gastroenterology and Dietology, Vilnius University Hospital, Santariskiu Klinikos Santariskiu str. 2, 08661, Vilnius, Lithuania.
| |
Collapse
|
21
|
Xu FP, Liu YH, Luo XL, Zhang F, Zhou HY, Ge Y, Liu C, Chen J, Luo DL, Yan LX, Mei P, Xu J, Zhuang HG. Overexpression of SRC-3 promotes esophageal squamous cell carcinoma aggressiveness by enhancing cell growth and invasiveness. Cancer Med 2016; 5:3500-3511. [PMID: 27781415 PMCID: PMC5224859 DOI: 10.1002/cam4.884] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Revised: 08/02/2016] [Accepted: 08/04/2016] [Indexed: 02/06/2023] Open
Abstract
Steroid receptor coactivator‐3 (SRC‐3), a transcriptional coactivator for nuclear receptors and other transcription factors, plays an important role in the genesis and progression of several cancers. However, studies investigated the role of SRC‐3 in esophageal squamous cell carcinomas (ESCCs) are limited, and the role of SRC‐3 in tumor progression remains unclear. We examined the expression of SRC‐3 in 8 ESCC cell lines and 302 human ESCC tissues by qPCR, Western blot, and immunohistochemistry. In addition, ESCC cell lines were subjected to proliferation and invasion assays, tumorigenicity assay, flow cytometry assay, qPCR, Western blot, and Chromatin Immunoprecipitation assay to investigate the role of SRC‐3 in cancer progression. SRC‐3 was overexpressed in 48% of cases and correlated with poor overall (P = 0.0076) and progression‐free (P = 0.0069) survival of surgically resected ESCC patient. Cox regression analysis revealed that SRC‐3 is an independent prognostic marker. Furthermore, we found that activation of insulin‐like growth factor (IGF)/AKT) was involved in the SRC‐3 on the cell growth and invasiveness in two ESCC cell lines, Eca109 and EC18 cells. SRC‐3 overexpression is clinically and functionally relevant to the progression of human ESCC, and might be a useful molecular target for ESCC prognosis and treatment.
Collapse
Affiliation(s)
- Fang-Ping Xu
- Department of Pathology and Laboratory Medicine, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yan-Hui Liu
- Department of Pathology and Laboratory Medicine, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xin-Lan Luo
- Department of Pathology and Laboratory Medicine, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Fen Zhang
- Department of Pathology and Laboratory Medicine, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Hai-Yu Zhou
- Department of Thoracic Surgery, Cancer Center, Guangdong General Hospital, Guangzhou, China
| | - Yan Ge
- Department of Pathology and Laboratory Medicine, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Chao Liu
- Department of Pathology and Laboratory Medicine, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jie Chen
- Department of Pathology and Laboratory Medicine, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Dong-Lan Luo
- Department of Pathology and Laboratory Medicine, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Li-Xu Yan
- Department of Pathology and Laboratory Medicine, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Ping Mei
- Department of Pathology and Laboratory Medicine, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jie Xu
- Department of Pathology and Laboratory Medicine, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Heng-Guo Zhuang
- Department of Pathology and Laboratory Medicine, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
22
|
Lonard DM, O'Malley BW. Molecular Pathways: Targeting Steroid Receptor Coactivators in Cancer. Clin Cancer Res 2016; 22:5403-5407. [PMID: 27654711 DOI: 10.1158/1078-0432.ccr-15-1958] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 08/24/2016] [Accepted: 09/01/2016] [Indexed: 11/16/2022]
Abstract
Coactivators represent a large class of proteins that partner with nuclear receptors and other transcription factors to regulate gene expression. Given their pleiotropic roles in the control of transcription, coactivators have been implicated in a broad range of human disease states, including cancer. This is best typified by the three members of the steroid receptor coactivator (SRC) family, each of which integrates steroid hormone signaling and growth factor pathways to drive oncogenic gene expression programs in breast, endometrial, ovarian, prostate, and other cancers. Because of this, coactivators represent emerging targets for cancer therapeutics, and efforts are now being made to develop SRC-targeting agents, such as the SI-2 inhibitor and the novel SRC stimulator, MCB-613, that are able to block cancer growth in cell culture and animal model systems. Here, we will discuss the mechanisms through which coactivators drive cancer progression and how targeting coactivators represent a novel conceptual approach to combat tumor growth that is distinct from the use of other targeted therapeutic agents. We also will describe efforts to develop next-generation SRC inhibitors and stimulators that can be taken into the clinic for the treatment of recurrent, drug-resistant cancers. Clin Cancer Res; 22(22); 5403-7. ©2016 AACR.
Collapse
Affiliation(s)
- David M Lonard
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Bert W O'Malley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
23
|
Shi J, Liu W, Sui F, Lu R, He Q, Yang Q, Lv H, Shi B, Hou P. Frequent amplification of AIB1, a critical oncogene modulating major signaling pathways, is associated with poor survival in gastric cancer. Oncotarget 2016; 6:14344-59. [PMID: 25970779 PMCID: PMC4546471 DOI: 10.18632/oncotarget.3852] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 04/17/2015] [Indexed: 11/26/2022] Open
Abstract
Amplified in breast cancer 1 (AIB1) is a member of p160 steroid receptor coactivator (SRC) family that mediates the transcriptional activities of nuclear receptors and other transcription factors. It acts as a major oncogene in diverse cancers, whereas biological function of AIB1 in gastric cancer remains largely unclear. This study was designed to explore the role of AIB1 in gastric tumorigenesis and its potential as a useful prognostic marker and therapeutic target in this cancer. Our data demonstrated that AIB1 was significantly up-regulated in gastric cancer tissues as compared with control subjects. Moreover, AIB1 amplification was found in 47 of 133 (35.3%) gastric cancer cases, but not in control subjects. AIB1 amplification was positively associated with its protein expression, and was significantly correlated with poor patient survival. AIB1 knockdown in gastric cancer cells dramatically inhibited cell proliferation, invasiveness and tumorigenic potential in nude mice, and induced cell cycle arrest and apoptosis. Mechanically, AIB1 promotes gastric cancer cell proliferation, survival and invasiveness through modulating major signaling pathways such as ErbB and Wnt/β-catenin pathways. Collectively, these findings suggest that AIB1 plays an important role in the pathogenesis of gastric cancer and represents a potential prognostic marker and therapeutic target for this cancer.
Collapse
Affiliation(s)
- Jing Shi
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an 710061, The People's Republic of China
| | - Wei Liu
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an 710061, The People's Republic of China
| | - Fang Sui
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an 710061, The People's Republic of China
| | - Rong Lu
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an 710061, The People's Republic of China
| | - Qingyuan He
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an 710061, The People's Republic of China
| | - Qi Yang
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an 710061, The People's Republic of China
| | - Hongjun Lv
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an 710061, The People's Republic of China
| | - Bingyin Shi
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an 710061, The People's Republic of China
| | - Peng Hou
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an 710061, The People's Republic of China
| |
Collapse
|
24
|
Nikolai BC, Lanz RB, York B, Dasgupta S, Mitsiades N, Creighton CJ, Tsimelzon A, Hilsenbeck SG, Lonard DM, Smith CL, O'Malley BW. HER2 Signaling Drives DNA Anabolism and Proliferation through SRC-3 Phosphorylation and E2F1-Regulated Genes. Cancer Res 2016; 76:1463-75. [PMID: 26833126 PMCID: PMC4794399 DOI: 10.1158/0008-5472.can-15-2383] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 11/22/2015] [Indexed: 12/29/2022]
Abstract
Approximately 20% of early-stage breast cancers display amplification or overexpression of the ErbB2/HER2 oncogene, conferring poor prognosis and resistance to endocrine therapy. Targeting HER2(+) tumors with trastuzumab or the receptor tyrosine kinase (RTK) inhibitor lapatinib significantly improves survival, yet tumor resistance and progression of metastatic disease still develop over time. Although the mechanisms of cytosolic HER2 signaling are well studied, nuclear signaling components and gene regulatory networks that bestow therapeutic resistance and limitless proliferative potential are incompletely understood. Here, we use biochemical and bioinformatic approaches to identify effectors and targets of HER2 transcriptional signaling in human breast cancer. Phosphorylation and activity of the Steroid Receptor Coactivator-3 (SRC-3) is reduced upon HER2 inhibition, and recruitment of SRC-3 to regulatory elements of endogenous genes is impaired. Transcripts regulated by HER2 signaling are highly enriched with E2F1 binding sites and define a gene signature associated with proliferative breast tumor subtypes, cell-cycle progression, and DNA replication. We show that HER2 signaling promotes breast cancer cell proliferation through regulation of E2F1-driven DNA metabolism and replication genes together with phosphorylation and activity of the transcriptional coactivator SRC-3. Furthermore, our analyses identified a cyclin-dependent kinase (CDK) signaling node that, when targeted using the CDK4/6 inhibitor palbociclib, defines overlap and divergence of adjuvant pharmacologic targeting. Importantly, lapatinib and palbociclib strictly block de novo synthesis of DNA, mostly through disruption of E2F1 and its target genes. These results have implications for rational discovery of pharmacologic combinations in preclinical models of adjuvant treatment and therapeutic resistance.
Collapse
Affiliation(s)
- Bryan C Nikolai
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Rainer B Lanz
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Brian York
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Subhamoy Dasgupta
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Nicholas Mitsiades
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas. Department of Medicine, Baylor College of Medicine, Houston, Texas. Center for Drug Discovery, Baylor College of Medicine, Houston, Texas
| | - Chad J Creighton
- Department of Medicine, Baylor College of Medicine, Houston, Texas. Dan L. Duncan Cancer Center Division of Biostatistics, Baylor College of Medicine, Houston, Texas
| | - Anna Tsimelzon
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Susan G Hilsenbeck
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - David M Lonard
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Carolyn L Smith
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Bert W O'Malley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
25
|
Fernández Larrosa PN, Ruíz Grecco M, Mengual Gómez D, Alvarado CV, Panelo LC, Rubio MF, Alonso DF, Gómez DE, Costas MA. RAC3 more than a nuclear receptor coactivator: a key inhibitor of senescence that is downregulated in aging. Cell Death Dis 2015; 6:e1902. [PMID: 26469953 PMCID: PMC4632280 DOI: 10.1038/cddis.2015.218] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 06/24/2015] [Accepted: 07/01/2015] [Indexed: 11/10/2022]
Abstract
Receptor-associated coactivator 3 (RAC3) is a nuclear receptor coactivator usually overexpressed in tumors that exerts oncogenic functions in the cytoplasm and the nucleus. Although as part of its oncogenic actions it was previously identified as an inhibitor of apoptosis and autophagy, its expression is required in order to preserve the pluripotency and embryonic stem cell self-renewal. In this work we investigated its role in cellular senescence. We found that RAC3 overexpression in the nontumoral HEK293 cells inhibits the premature senescence induced by hydrogen peroxide or rapamycin. The mechanism involves not only the inhibition of autophagy early induced by these stimuli in the pathway to senescence, but also the increase in levels and nuclear localization of both the cell cycle suppressors p53/p21 and the longevity promoters FOXO1A, FOXO3A and SIRT1. Furthermore, we found that RAC3 overexpression is required in order to maintain the telomerase activity. In tumoral HeLa cells its activity was inhibited by depletion of RAC3 inducing replicative senescence. Moreover, we demonstrated that in vivo, levels of RAC3 are downregulated in the liver from aged as compared with young rats, whereas the levels of p21 are increased, correlating with the expected senescent cell contents in aged tissues. A similar downregulation of RAC3 was observed in the premature and replicative senescence of human fetal WI-38 cells and premature senescence of hepatocyte HepG2 cell line. Taken together, all these results demonstrate that RAC3 is an inhibitor of senescence whose downregulation in aged individuals could be probably a tumor suppressor mechanism, avoiding the clonal expansion of risky old cells having damaged DNA.
Collapse
Affiliation(s)
- P N Fernández Larrosa
- Laboratorio de Biología Molecular y Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150, Buenos Aires C1427ARO, Argentina
| | - M Ruíz Grecco
- Laboratorio de Biología Molecular y Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150, Buenos Aires C1427ARO, Argentina
| | - D Mengual Gómez
- Laboratorio de Oncología Molecular, Universidad Nacional de Quilmes, R. Sáenz Peña 352, Bernal, Buenos Aires B1876BXD Argentina
| | - C V Alvarado
- Laboratorio de Biología Molecular y Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150, Buenos Aires C1427ARO, Argentina
| | - L C Panelo
- Laboratorio de Biología Molecular y Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150, Buenos Aires C1427ARO, Argentina
| | - M F Rubio
- Laboratorio de Biología Molecular y Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150, Buenos Aires C1427ARO, Argentina
| | - D F Alonso
- Laboratorio de Oncología Molecular, Universidad Nacional de Quilmes, R. Sáenz Peña 352, Bernal, Buenos Aires B1876BXD Argentina
| | - D E Gómez
- Laboratorio de Oncología Molecular, Universidad Nacional de Quilmes, R. Sáenz Peña 352, Bernal, Buenos Aires B1876BXD Argentina
| | - M A Costas
- Laboratorio de Biología Molecular y Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150, Buenos Aires C1427ARO, Argentina
| |
Collapse
|
26
|
Steroid Receptor Coactivator-3 (SRC-3/AIB1) as a Novel Therapeutic Target in Triple Negative Breast Cancer and Its Inhibition with a Phospho-Bufalin Prodrug. PLoS One 2015; 10:e0140011. [PMID: 26431029 PMCID: PMC4592245 DOI: 10.1371/journal.pone.0140011] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 09/08/2015] [Indexed: 11/19/2022] Open
Abstract
Triple negative breast cancer (TNBC) has the poorest prognosis of all types of breast cancer and currently lacks efficient targeted therapy. Chemotherapy is the traditional standard-of-care for TNBC, but is frequently accompanied by severe side effects. Despite the fact that high expression of steroid receptor coactivator 3 (SRC-3) is correlated with poor survival in estrogen receptor positive breast cancer patients, its role in TNBC has not been extensively investigated. Here, we show that high expression of SRC-3 correlates with both poor overall survival and post progression survival in TNBC patients, suggesting that SRC-3 can serve as a prognostic marker for TNBC. Furthermore, we demonstrated that bufalin, a SRC-3 small molecule inhibitor, when introduced even at nM concentrations, can significantly reduce TNBC cell viability and motility. However, because bufalin has minimal water solubility, its in vivo application is limited. Therefore, we developed a water soluble prodrug, 3-phospho-bufalin, to facilitate its in vivo administration. In addition, we demonstrated that 3-phospho-bufalin can effectively inhibit tumor growth in an orthotopic TNBC mouse model, suggesting its potential application as a targeted therapy for TNBC treatment.
Collapse
|
27
|
Wang L, Yu Y, Chow DC, Yan F, Hsu CC, Stossi F, Mancini MA, Palzkill T, Liao L, Zhou S, Xu J, Lonard DM, O'Malley BW. Characterization of a Steroid Receptor Coactivator Small Molecule Stimulator that Overstimulates Cancer Cells and Leads to Cell Stress and Death. Cancer Cell 2015; 28:240-52. [PMID: 26267537 PMCID: PMC4536575 DOI: 10.1016/j.ccell.2015.07.005] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 03/12/2015] [Accepted: 07/10/2015] [Indexed: 12/18/2022]
Abstract
By integrating growth pathways on which cancer cells rely, steroid receptor coactivators (SRC-1, SRC-2, and SRC-3) represent emerging targets in cancer therapeutics. High-throughput screening for SRC small molecule inhibitors (SMIs) uncovered MCB-613 as a potent SRC small molecule "stimulator" (SMS). We demonstrate that MCB-613 can super-stimulate SRCs' transcriptional activity. Further investigation revealed that MCB-613 increases SRCs' interactions with other coactivators and markedly induces ER stress coupled to the generation of reactive oxygen species (ROS). Because cancer cells overexpress SRCs and rely on them for growth, we show that we can exploit MCB-613 to selectively induce excessive stress in cancer cells. This suggests that over-stimulating the SRC oncogenic program can be an effective strategy to kill cancer cells.
Collapse
Affiliation(s)
- Lei Wang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yang Yu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Dar-Chone Chow
- Department of Pharmacology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Fei Yan
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Chih-Chao Hsu
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Fabio Stossi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Michael A Mancini
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Timothy Palzkill
- Department of Pharmacology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lan Liao
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Suoling Zhou
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jianming Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - David M Lonard
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Bert W O'Malley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
28
|
Eedunuri VK, Rajapakshe K, Fiskus W, Geng C, Chew SA, Foley C, Shah SS, Shou J, Mohamed JS, Coarfa C, O'Malley BW, Mitsiades N. miR-137 Targets p160 Steroid Receptor Coactivators SRC1, SRC2, and SRC3 and Inhibits Cell Proliferation. Mol Endocrinol 2015; 29:1170-83. [PMID: 26066330 DOI: 10.1210/me.2015-1080] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The p160 family of steroid receptor coactivators (SRCs) are pleiotropic transcription factor coactivators and "master regulators" of gene expression that promote cancer cell proliferation, survival, metabolism, migration, invasion, and metastasis. Cancers with high p160 SRC expression exhibit poor clinical outcomes and resistance to therapy, highlighting the SRCs as critical oncogenic drivers and, thus, therapeutic targets. microRNAs are important epigenetic regulators of protein expression. To examine the regulation of p160 SRCs by microRNAs, we used and combined 4 prediction algorithms to identify microRNAs that could target SRC1, SRC2, and SRC3 expression. For validation of these predictions, we assessed p160 SRC protein expression and cell viability after transfection of corresponding microRNA mimetics in breast cancer, uveal melanoma, and prostate cancer (PC) cell lines. Transfection of selected microRNA mimetics into breast cancer, uveal melanoma, and PC cells depleted SRC protein expression levels and exerted potent antiproliferative activity in these cell types. In particular, microRNA-137 (miR-137) depleted expression of SRC1, SRC2, and very potently, SRC3. The latter effect can be attributed to the presence of 3 miR-137 recognition sequences within the SRC3 3'-untranslated region. Using reverse phase protein array analysis, we identified a network of proteins, in addition to SRC3, that were modulated by miR-137 in PC cells. We also found that miR-137 and its host gene are epigenetically silenced in human cancer specimens and cell lines. These results support the development and testing of microRNA-based therapies (in particular based on restoring miR-137 levels) for targeting the oncogenic family of p160 SRCs in cancer.
Collapse
Affiliation(s)
- Vijay Kumar Eedunuri
- Adrienne Helis Malvin Medical Research Foundation (V.K.E.), New Orleans, Louisiana 70130; and Departments of Molecular and Cellular Biology (K.R., W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., C.C., B.W.O., N.M.) and Department of Medicine (W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., N.M.), Baylor College of Medicine, Houston, Texas 77030
| | - Kimal Rajapakshe
- Adrienne Helis Malvin Medical Research Foundation (V.K.E.), New Orleans, Louisiana 70130; and Departments of Molecular and Cellular Biology (K.R., W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., C.C., B.W.O., N.M.) and Department of Medicine (W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., N.M.), Baylor College of Medicine, Houston, Texas 77030
| | - Warren Fiskus
- Adrienne Helis Malvin Medical Research Foundation (V.K.E.), New Orleans, Louisiana 70130; and Departments of Molecular and Cellular Biology (K.R., W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., C.C., B.W.O., N.M.) and Department of Medicine (W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., N.M.), Baylor College of Medicine, Houston, Texas 77030
| | - Chuandong Geng
- Adrienne Helis Malvin Medical Research Foundation (V.K.E.), New Orleans, Louisiana 70130; and Departments of Molecular and Cellular Biology (K.R., W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., C.C., B.W.O., N.M.) and Department of Medicine (W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., N.M.), Baylor College of Medicine, Houston, Texas 77030
| | - Sue Anne Chew
- Adrienne Helis Malvin Medical Research Foundation (V.K.E.), New Orleans, Louisiana 70130; and Departments of Molecular and Cellular Biology (K.R., W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., C.C., B.W.O., N.M.) and Department of Medicine (W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., N.M.), Baylor College of Medicine, Houston, Texas 77030
| | - Christopher Foley
- Adrienne Helis Malvin Medical Research Foundation (V.K.E.), New Orleans, Louisiana 70130; and Departments of Molecular and Cellular Biology (K.R., W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., C.C., B.W.O., N.M.) and Department of Medicine (W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., N.M.), Baylor College of Medicine, Houston, Texas 77030
| | - Shrijal S Shah
- Adrienne Helis Malvin Medical Research Foundation (V.K.E.), New Orleans, Louisiana 70130; and Departments of Molecular and Cellular Biology (K.R., W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., C.C., B.W.O., N.M.) and Department of Medicine (W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., N.M.), Baylor College of Medicine, Houston, Texas 77030
| | - John Shou
- Adrienne Helis Malvin Medical Research Foundation (V.K.E.), New Orleans, Louisiana 70130; and Departments of Molecular and Cellular Biology (K.R., W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., C.C., B.W.O., N.M.) and Department of Medicine (W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., N.M.), Baylor College of Medicine, Houston, Texas 77030
| | - Junaith S Mohamed
- Adrienne Helis Malvin Medical Research Foundation (V.K.E.), New Orleans, Louisiana 70130; and Departments of Molecular and Cellular Biology (K.R., W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., C.C., B.W.O., N.M.) and Department of Medicine (W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., N.M.), Baylor College of Medicine, Houston, Texas 77030
| | - Cristian Coarfa
- Adrienne Helis Malvin Medical Research Foundation (V.K.E.), New Orleans, Louisiana 70130; and Departments of Molecular and Cellular Biology (K.R., W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., C.C., B.W.O., N.M.) and Department of Medicine (W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., N.M.), Baylor College of Medicine, Houston, Texas 77030
| | - Bert W O'Malley
- Adrienne Helis Malvin Medical Research Foundation (V.K.E.), New Orleans, Louisiana 70130; and Departments of Molecular and Cellular Biology (K.R., W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., C.C., B.W.O., N.M.) and Department of Medicine (W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., N.M.), Baylor College of Medicine, Houston, Texas 77030
| | - Nicholas Mitsiades
- Adrienne Helis Malvin Medical Research Foundation (V.K.E.), New Orleans, Louisiana 70130; and Departments of Molecular and Cellular Biology (K.R., W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., C.C., B.W.O., N.M.) and Department of Medicine (W.F., C.G., S.A.C., C.F., S.S.S., J.S., J.S.M., N.M.), Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
29
|
Abstract
Pancreatic cancer is an insidious type of cancer with its symptoms manifested upon extensive disease. The overall 5-year survival rates between 0.4 and 4%. Surgical resection is an option for only 10% of the patients with pancreatic cancer. Local recurrence and hepatic metastases occur within 2 years after surgery. There are currently several molecular pathways investigated and novel targeted treatments are on the market. However; the nature of pancreatic cancer with its ability to spread locally in the primary site and lymph nodes indicates that further experimentation with local interventional therapies could be a future treatment proposal as palliative care or adjunct to gene therapy and chemotherapy/radiotherapy. In the current review, we will summarize the molecular pathways and present the interventional treatment options for pancreatic cancer.
Collapse
|
30
|
NCOA3-mediated upregulation of mucin expression via transcriptional and post-translational changes during the development of pancreatic cancer. Oncogene 2014; 34:4879-89. [PMID: 25531332 DOI: 10.1038/onc.2014.409] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 10/12/2014] [Accepted: 10/19/2014] [Indexed: 01/28/2023]
Abstract
Pancreatic cancer (PC) is characterized by aberrant overexpression of mucins that contribute to its pathogenesis. Although the inflammatory cytokines contribute to mucin overexpression, the mucin profile of PC is markedly distinct from that of normal or inflamed pancreas. We postulated that de novo expression of various mucins in PC involves chromatin modifications. Analysis of chromatin modifying enzymes by PCR array identified differential expression of NCOA3 in MUC4-expressing PC cell lines. Immunohistochemistry analysis in tumor tissues from patients and spontaneous mouse models, and microarray analysis following the knockdown of NCOA3 were performed to elucidate its role in mucin regulation and overall impact on PC. Silencing of NCOA3 in PC cell lines resulted in significant downregulation of two most differentially expressed mucins in PC, MUC4 and MUC1 (P<0.01). Immunohistochemistry analysis in PC tissues and metastatic lesions established an association between NCOA3 and mucin (MUC1 and MUC4) expression. Spontaneous mouse model of PC (K-ras(G12D); Pdx-1cre) showed early expression of Ncoa3 during pre-neoplastic lesions. Mechanistically, NCOA3 knockdown abrogated retinoic acid-mediated MUC4 upregulation by restricting MUC4 promoter accessibility as demonstrated by micrococcus nuclease digestion (P<0.05) and chromatin immuno-precipitation analysis. NCOA3 also created pro-inflammatory conditions by upregulating chemokines like CXCL1, 2, 5 and CCL20 (P<0.001). AKT, ubiquitin C, ERK1/2 and NF-κB occupied dominant nodes in the networks significantly modulated after NCOA3 silencing. In addition, NCOA3 stabilized mucins post translationally through fucosylation by FUT8, as the knockdown of FUT8 resulted in the downregulation of MUC4 and MUC1 at protein levels.
Collapse
|
31
|
Alvarado CV, Rubio MF, Fernández Larrosa PN, Panelo LC, Azurmendi PJ, Ruiz Grecco M, Martínez-Nöel GA, Costas MA. The levels of RAC3 expression are up regulated by TNF in the inflammatory response. FEBS Open Bio 2014; 4:450-7. [PMID: 24918060 PMCID: PMC4050193 DOI: 10.1016/j.fob.2014.04.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 04/21/2014] [Accepted: 04/21/2014] [Indexed: 12/01/2022] Open
Abstract
The inflammatory response increases the expression of RAC3 in vitro and in vivo. TNF induces the increase of RAC3 at transcriptional level through NF-κB activation. Glucocorticoids also induce the increase of RAC3 expression levels. RAC3 appears to be essential for NF-κB- and GR-mediated transcription.
RAC3 is a coactivator of glucocorticoid receptor and nuclear factor-κB (NF-κB) that is usually over-expressed in tumors and which also has important functions in the immune system. We investigated the role of the inflammatory response in the control of RAC3 expression levels in vivo and in vitro. We found that inflammation regulates RAC3 levels. In mice, sub-lethal doses of lipopolysaccharide induce the increase of RAC3 in spleen and the administration of the synthetic anti-inflammatory glucocorticoid dexamethasone has a similar effect. However, the simultaneous treatment with both stimuli is mutually antagonistic. In vitro stimulation of the HEK293 cell line with tumor necrosis factor (TNF), one of the cytokines induced by lipopolysaccharide, also increases the levels of RAC3 mRNA and protein, which correlates with an enhanced transcription dependent on the RAC3 gene promoter. We found that binding of the transcription factor NF-κB to the RAC3 gene promoter could be responsible for these effects. Our results suggest that increase of RAC3 during the inflammatory response could be a molecular mechanism involved in the control of sensitivity to both pro- and anti-inflammatory stimuli in order to maintain the normal healthy course of the immune response.
Collapse
Affiliation(s)
- Cecilia Viviana Alvarado
- Laboratorio de Biología Molecular y Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150, C1427ARO Buenos Aires, Argentina
| | - María Fernanda Rubio
- Laboratorio de Biología Molecular y Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150, C1427ARO Buenos Aires, Argentina
- Argentine National Research Council (CONICET), Argentina
| | - Pablo Nicolas Fernández Larrosa
- Laboratorio de Biología Molecular y Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150, C1427ARO Buenos Aires, Argentina
- Argentine National Research Council (CONICET), Argentina
| | - Laura Carolina Panelo
- Laboratorio de Biología Molecular y Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150, C1427ARO Buenos Aires, Argentina
| | - Pablo Javier Azurmendi
- Laboratorio de Riñón Experimental, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150, C1427ARO Buenos Aires, Argentina
| | - Marina Ruiz Grecco
- Laboratorio de Biología Molecular y Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150, C1427ARO Buenos Aires, Argentina
| | - Giselle Astrid Martínez-Nöel
- Laboratorio de Biología Molecular y Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150, C1427ARO Buenos Aires, Argentina
- Argentine National Research Council (CONICET), Argentina
| | - Mónica Alejandra Costas
- Laboratorio de Biología Molecular y Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150, C1427ARO Buenos Aires, Argentina
- Argentine National Research Council (CONICET), Argentina
- Corresponding author at: Laboratorio de Biología Molecular y Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150, C1427ARO Buenos Aires, Argentina. Tel.: +54 01145148702; fax: +54 11 4523 8947.
| |
Collapse
|
32
|
Qin L, Wu YL, Toneff MJ, Li D, Liao L, Gao X, Bane FT, Tien JCY, Xu Y, Feng Z, Yang Z, Xu Y, Theissen SM, Li Y, Young L, Xu J. NCOA1 Directly Targets M-CSF1 Expression to Promote Breast Cancer Metastasis. Cancer Res 2014; 74:3477-88. [PMID: 24769444 DOI: 10.1158/0008-5472.can-13-2639] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
In breast cancer, overexpression of the nuclear coactivator NCOA1 (SRC-1) is associated with disease recurrence and resistance to endocrine therapy. To examine the impact of NCOA1 overexpression on morphogenesis and carcinogenesis in the mammary gland (MG), we generated MMTV-hNCOA1 transgenic [Tg(NCOA1)] mice. In the context of two distinct transgenic models of breast cancer, NCOA1 overexpression did not affect the morphology or tumor-forming capability of MG epithelial cells. However, NCOA1 overexpression increased the number of circulating breast cancer cells and the efficiency of lung metastasis. Mechanistic investigations showed that NCOA1 and c-Fos were recruited to a functional AP-1 site in the macrophage attractant CSF1 promoter, directly upregulating colony-simulating factor 1 (CSF1) expression to enhance macrophage recruitment and metastasis. Conversely, silencing NCOA1 reduced CSF1 expression and decreased macrophage recruitment and breast cancer cell metastasis. In a cohort of 453 human breast tumors, NCOA1 and CSF1 levels correlated positively with disease recurrence, higher tumor grade, and poor prognosis. Together, our results define an NCOA1/AP-1/CSF1 regulatory axis that promotes breast cancer metastasis, offering a novel therapeutic target for impeding this process.
Collapse
Affiliation(s)
- Li Qin
- Authors' Affiliations: Department of Molecular and Cellular Biology, Dan L. Duncan Cancer Center, Baylor College of Medicine
| | - Ye-Lin Wu
- Authors' Affiliations: Department of Molecular and Cellular Biology, Dan L. Duncan Cancer Center, Baylor College of Medicine; Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai
| | - Michael J Toneff
- Authors' Affiliations: Department of Molecular and Cellular Biology, Dan L. Duncan Cancer Center, Baylor College of Medicine
| | - Dabing Li
- Authors' Affiliations: Department of Molecular and Cellular Biology, Dan L. Duncan Cancer Center, Baylor College of Medicine; Institute for Cancer Medicine and Department of Pathology, Luzhou Medical College, Luzhou, Sichuan, China; and
| | - Lan Liao
- Authors' Affiliations: Department of Molecular and Cellular Biology, Dan L. Duncan Cancer Center, Baylor College of Medicine
| | - Xiuhua Gao
- Authors' Affiliations: Department of Molecular and Cellular Biology, Dan L. Duncan Cancer Center, Baylor College of Medicine
| | - Fiona T Bane
- Endocrine Oncology Research, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Jean C-Y Tien
- Authors' Affiliations: Department of Molecular and Cellular Biology, Dan L. Duncan Cancer Center, Baylor College of Medicine; Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, Texas
| | - Yixiang Xu
- Authors' Affiliations: Department of Molecular and Cellular Biology, Dan L. Duncan Cancer Center, Baylor College of Medicine; Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, Texas
| | - Zhen Feng
- Authors' Affiliations: Department of Molecular and Cellular Biology, Dan L. Duncan Cancer Center, Baylor College of Medicine; Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai
| | - Zhihui Yang
- Authors' Affiliations: Department of Molecular and Cellular Biology, Dan L. Duncan Cancer Center, Baylor College of Medicine; Institute for Cancer Medicine and Department of Pathology, Luzhou Medical College, Luzhou, Sichuan, China; and
| | - Yan Xu
- Authors' Affiliations: Department of Molecular and Cellular Biology, Dan L. Duncan Cancer Center, Baylor College of Medicine
| | - Sarah M Theissen
- Authors' Affiliations: Department of Molecular and Cellular Biology, Dan L. Duncan Cancer Center, Baylor College of Medicine
| | - Yi Li
- Authors' Affiliations: Department of Molecular and Cellular Biology, Dan L. Duncan Cancer Center, Baylor College of Medicine
| | - Leonie Young
- Endocrine Oncology Research, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Jianming Xu
- Authors' Affiliations: Department of Molecular and Cellular Biology, Dan L. Duncan Cancer Center, Baylor College of Medicine; Institute for Cancer Medicine and Department of Pathology, Luzhou Medical College, Luzhou, Sichuan, China; and
| |
Collapse
|
33
|
Wang Y, Lonard DM, Yu Y, Chow DC, Palzkill TG, Wang J, Qi R, Matzuk AJ, Song X, Madoux F, Hodder P, Chase P, Griffin PR, Zhou S, Liao L, Xu J, O'Malley BW. Bufalin is a potent small-molecule inhibitor of the steroid receptor coactivators SRC-3 and SRC-1. Cancer Res 2014; 74:1506-1517. [PMID: 24390736 DOI: 10.1158/0008-5472.can-13-2939] [Citation(s) in RCA: 138] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Virtually all transcription factors partner with coactivators that recruit chromatin remodeling factors and interact with the basal transcription machinery. Coactivators have been implicated in cancer cell proliferation, invasion, and metastasis, including the p160 steroid receptor coactivator (SRC) family composed of SRC-1 (NCOA1), SRC-2 (TIF2/GRIP1/NCOA2), and SRC-3 (AIB1/ACTR/NCOA3). Given their broad involvement in many cancers, they represent candidate molecular targets for new chemotherapeutics. Here, we report on the results of a high-throughput screening effort that identified the cardiac glycoside bufalin as a potent small-molecule inhibitor for SRC-3 and SRC-1. Bufalin strongly promoted SRC-3 protein degradation and was able to block cancer cell growth at nanomolar concentrations. When incorporated into a nanoparticle delivery system, bufalin was able to reduce tumor growth in a mouse xenograft model of breast cancer. Our work identifies bufalin as a potentially broad-spectrum small-molecule inhibitor for cancer.
Collapse
Affiliation(s)
- Ying Wang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - David M Lonard
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yang Yu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Dar-Chone Chow
- Department of Pharmacology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Timothy G Palzkill
- Department of Pharmacology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jin Wang
- Department of Pharmacology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ruogu Qi
- Department of Pharmacology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alexander J Matzuk
- Department of Pharmacology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xianzhou Song
- Department of Pharmacology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Franck Madoux
- Department of Molecular Therapeutics, The Scripps Research Institute, Scripps Florida, Jupiter, FL 33458, USA
| | - Peter Hodder
- Department of Molecular Therapeutics, The Scripps Research Institute, Scripps Florida, Jupiter, FL 33458, USA
| | - Peter Chase
- Department of Molecular Therapeutics, The Scripps Research Institute, Scripps Florida, Jupiter, FL 33458, USA
| | - Patrick R Griffin
- Department of Molecular Therapeutics, The Scripps Research Institute, Scripps Florida, Jupiter, FL 33458, USA
| | - Suoling Zhou
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lan Liao
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jianming Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Bert W O'Malley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
34
|
Liu MY, Guo HP, Hong CQ, Peng HW, Yang XH, Zhang H. Up-regulation of nuclear receptor coactivator amplified in breast cancer-1 in papillary thyroid carcinoma correlates with lymph node metastasis. Clin Transl Oncol 2013; 15:947-52. [PMID: 23606350 DOI: 10.1007/s12094-013-1029-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 03/14/2013] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Nuclear receptor coactivator amplified in breast cancer-1 (AIB1), a new oncogenic coactivator, is commonly overexpressed and amplified in variety of human cancers. However, the expression of AIB1 in papillary thyroid carcinoma (PTC), the major histologic type of thyroid cancer, and its clinical significance are still unclear. MATERIALS AND METHODS AIB1 expression in PTC was examined by immunohistochemistry using tissue microarrays comprised of 90 primary PTC, 46 matched lymph node, and 20 normal thyroid tissue specimens in this study. RESULTS In the normal thyroid specimens, AIB1 expression was either absent or at low levels. In contrast, AIB1 overexpression was detected in 50 of 83 (60.2 %) primary PTC specimens. Up-regulated AIB1 was evident in 39 of 46 (73.5 %) matched lymph nodes. Overexpression of AIB1 was observed more frequently in PTCs with lymph node metastasis [N1a/N1b, 39/46 (73.5 %)] versus PTCs without lymph node metastasis [N0, 14/34 (41.2 %)]. Furthermore, high-level AIB1 expression was only observed in the lymph node-positive specimens. Moreover, we found no correlation between AIB1 expression and ER expression in PTC tissues. CONCLUSIONS Our findings suggest that overexpression of AIB1 may be a biomarker for tumorigenesis and progression of PTC and may play an important role in its acquisition of a metastatic phenotype.
Collapse
Affiliation(s)
- M-Y Liu
- Department of Head and Neck Surgery, Cancer Hospital, Shantou University Medical College, Shantou, 515000, China
| | | | | | | | | | | |
Collapse
|
35
|
Luo F, Li W, Zhang J, Huang K, Fu J, Xie Z. Overexpression of steroid receptor coactivator-3 in bone cancers: an in vivo immunohistochemical study with tissue microarray. Pathol Res Pract 2013; 209:790-6. [PMID: 24134957 DOI: 10.1016/j.prp.2013.09.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 08/02/2013] [Accepted: 09/16/2013] [Indexed: 11/17/2022]
Abstract
Bone tissue is steroid-responsive and profoundly regulated by steroids and/or their receptors. Bone cancers (either primary or metastatic) belong to the most dangerous tumors. Previous studies have demonstrated overexpression of steroid receptor coactivator-3 (SRC-3) in many cancers, such as breast cancer, prostate cancer, thyroid cancer, functioning in the regulation of cancer cell proliferation, invasion, and metastasis. However, so far, the expression and function of SRC-3 in bone cancers have not yet been clarified. In this study, nickel-intensified immunohistochemistry was conducted using a commercial tissue microarray (with 94 cases of bone cancer tissue and 10 normal bone tissues), and the 4-scoring system was employed to evaluate the expression levels of SRC-3 immunoreactivity. The results showed that in normal bone tissue, levels of SRC-3 are almost negative (score=0), the total positivity (score=1-3) of SRC-3 immunoreactivities in bone cancers was 74.47%. There were no significant differences in gender, status (malignant or benign) or (mean) age (p>0.05). The percentage of positivity was 77.78% in osteogenic tumors, 58.82% in cartilage tumors, 70% in giant cell tumors, 100% in hematopoietic tumors, 77.78% in miscellaneous lesions, and 75% in miscellaneous tumors. Age related differences of SRC-3 immunoreactivities were detected in cartilage tumors and giant cell tumors (p<0.05). The above results clearly demonstrated a high frequency of overexpression of SRC-3 immunoreactivities in different bone cancers, indicating its potential roles in the prognosis and treatment of these cancers.
Collapse
Affiliation(s)
- Fei Luo
- Department of Orthopedic Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | | | | | | | | | | |
Collapse
|
36
|
Hou Y, Zhu P, Chen YQ, Zhu BY, Gu YF. Progress in understanding mechanisms behind anti-tumor effects of gambogic acid. Shijie Huaren Xiaohua Zazhi 2013; 21:2412-2417. [DOI: 10.11569/wcjd.v21.i24.2412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Gambogic acid, a major active component of gamboge resin extracted from Garcinia hanburyi tree, has potent anti-tumor activities. Based on domestic and foreign literature, we speculate that the anti-tumor activities of gambogic acid might be mediated via different mechanisms, including inducing cell apoptosis and cell cycle arrest, inhibiting the activity of telomerase or topoisomerase, reducing tumor cell invasion, adhesion and migration, and reversal of tumor multidrug resistance (MDR). In this paper, we review the mechanisms behind the anti-tumor effects of gambogic acid, with an aim to provide some new clues to the development of anti-tumor drugs.
Collapse
|
37
|
Guo S, Xu J, Xue R, Liu Y, Yu H. Overexpression of AIB1 correlates inversely with E-cadherin expression in pancreatic adenocarcinoma and may promote lymph node metastasis. Int J Clin Oncol 2013; 19:319-24. [PMID: 23542947 DOI: 10.1007/s10147-013-0549-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2012] [Accepted: 03/09/2013] [Indexed: 10/27/2022]
Abstract
BACKGROUND It was reported that the nuclear receptor coactivator amplified in breast cancer1 (AIB1) could regulate cancer cell invasion and migration in a nuclear receptor signaling-independent manner. Meanwhile, the process of epithelial mesenchymal transition (EMT) is critical for tumor invasion and metastasis. The present study aimed to determine the role of AIB1 and EMT markers in human pancreatic adenocarcinoma. METHODS AIB1, ZO-1, E-cadherin, vimentin, and N-cadherin protein expression in 76 pancreatic adenocarcinomas were assessed using immunohistochemistry and analyzed for clinicopathological significance. RESULTS The frequency of AIB1 overexpression in pancreatic adenocarcinomas with lymph node metastasis is 68 % (19/28), which is significantly higher than in pancreatic adenocarcinomas without lymph node metastasis (42 %; 20/48). In addition, the frequency of low expression of E-cadherin in pancreatic carcinomas with lymph node metastasis (68 %; 19/28) was significantly higher than in tumors without lymph node metastasis (44 %; 21/48). Correlation analysis demonstrated that the overexpression of AIB1 was inversely correlated with low expression of E-cadherin in pancreatic adenocarcinomas. CONCLUSION Overexpression of AIB1 might promote invasion and metastasis of cancer cells and is associated with down-regulation of E-cadherin in pancreatic adenocarcinomas.
Collapse
Affiliation(s)
- Si Guo
- Clinical Laboratory, Henan Provincial People's Hospital, 7 Wei Wu Road, Zhengzhou, 450000, China
| | | | | | | | | |
Collapse
|
38
|
PTEN suppresses the oncogenic function of AIB1 through decreasing its protein stability via mechanism involving Fbw7 alpha. Mol Cancer 2013; 12:21. [PMID: 23514585 PMCID: PMC3610140 DOI: 10.1186/1476-4598-12-21] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Accepted: 03/17/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Phosphatase and tensin homologue deleted on chromosome 10 (PTEN) is a phosphatase having both protein and lipid phosphatase activities, and is known to antagonize the phosphoinositide 3-kinase/AKT (PI3K/AKT) signaling pathway, resulting in tumor suppression. PTEN is also known to play a role in the regulation of numerous transcription factors. Amplified in breast cancer 1 (AIB1) is a transcriptional coactivator that mediates the transcriptional activities of nuclear receptors and other transcription factors. The present study investigated how PTEN may regulate AIB1, which is amplified and/or overexpressed in many human carcinomas, including breast cancers. RESULTS PTEN interacted with AIB1 via its phophatase domain and regulated the transcriptional activity of AIB1 by enhancing the ubiquitin-mediated degradation of AIB1. This process did not appear to require the phosphatase activity of PTEN, but instead, involved the interaction between PTEN and F-box and WD repeat domain-containing 7 alpha (Fbw7α), the E3 ubiquitin ligase involved in the ubiquitination of AIB1. PTEN interacted with Fbw7α via its C2 domain, thereby acting as a bridge between AIB1 and Fbw7α, and this led to enhanced degradation of AIB1, which eventually accounted for its decreased transcriptional activity. At the cell level, knockdown of PTEN in MCF-7 cells promoted cell proliferation. However when AIB1 was also knocked down, knockdown of PTEN had no effect on cell proliferation. CONCLUSIONS PTEN might act as a negative regulator of AIB1 whereby the association of PTEN with both AIB1 and Fbw7α could lead to the downregulation of AIB1 transcriptional activity, with the consequence of regulating the oncogenic function of AIB1.
Collapse
|
39
|
Carretero J, Blanco EJ, Carretero M, Carretero-Hernández M, García-Barrado MJ, Iglesias-Osma MC, Burks DJ, Font de Mora J. The expression of AIB1 correlates with cellular proliferation in human prolactinomas. Ann Anat 2013; 195:253-9. [PMID: 23433587 DOI: 10.1016/j.aanat.2013.01.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 01/11/2013] [Accepted: 01/12/2013] [Indexed: 10/27/2022]
Abstract
Estrogens as well as certain growth factors strongly influence the development and growth of prolactinomas. However, the molecular mechanisms by which extracellular factors trigger prolactinomas are not well known. Amplified in breast cancer 1 (AIB1), also known as steroid receptor co-activator 3 (SRC-3), belongs to the p160/SRC family of nuclear receptor co-activators and is a major co-activator of the estrogen receptor. Here, we report that the estrogen receptor coactivator AIB1 is overexpressed in human prolactinomas and correlates with the detection of aromatase and estrogen receptor α (ERα). Of the 87 pituitary tumors evaluated in women, 56%, corresponding to hyperoprolactinemic women, contained an enriched population of prolactin-positive cells and hence were further classified as prolactinomas. All prolactinomas stained positive for both ERα and AIB1. Moreover, AIB1 sub-cellular distribution was indicative of the cell-cycle status of tumors; the nuclear expression of AIB1 was correlated with proliferative markers whereas the cytoplasmic localization of AIB1 coincided with active caspase-3. Thus, our results demonstrate for the first time that AIB1 is expressed in prolactinomas and suggest its participation in the regulation of proliferation and apoptosis of tumoral cells. Because aromatase expression is also enhanced in these prolactinomas and it is involved in the local production of estradiol, both mechanisms, ER-AIB1 and aromatase could be related.
Collapse
Affiliation(s)
- José Carretero
- Department of Human Anatomy and Histology, Faculty of Medicine, University of Salamanca, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Yang LJ, Chen Y. New targets for the antitumor activity of gambogic acid in hematologic malignancies. Acta Pharmacol Sin 2013; 34:191-8. [PMID: 23274413 DOI: 10.1038/aps.2012.163] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Gambogic acid (GA) is the main active ingredient of gamboge, a brownish to orange dry resin secreted from Garcinia hanburyi, a plant that is widely distributed in nature. Recent in vitro and in vivo studies have demonstrated that GA exerts potent antitumor effects against solid tumors of various derivations, and its antitumor mechanisms have been thoroughly investigated. On the other hand, normal cells remain relatively resistant to GA, indicating a therapeutic window. GA is currently in clinical trials in China. Over the last decade, our laboratory demonstrates that GA exhibits potent anticancer activities against hematological malignancies. This review focuses on the new mechanisms through which GA inhibits proliferation and induces apoptosis in malignant hematological cells. These include the regulation of expression and intracellular positioning of nucleoporin and nucleophosmin; downregulation of steroid receptor coactivator-3 (SRC-3) and its downstream proteins; upregulation of death inducer-obliterator (DIO-1); downregulation of HERG potassium channel; as well as induction of reactive oxygen species (ROS) accumulation.
Collapse
|
41
|
Wei J, Cheang T, Tang B, Xia H, Xing Z, Chen Z, Fang Y, Chen W, Xu A, Wang S, Luo J. The inhibition of human bladder cancer growth by calcium carbonate/CaIP6 nanocomposite particles delivering AIB1 siRNA. Biomaterials 2013; 34:1246-54. [DOI: 10.1016/j.biomaterials.2012.09.068] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Accepted: 09/26/2012] [Indexed: 12/31/2022]
|
42
|
Fernandez Larrosa PN, Alvarado CV, Rubio MF, Ruiz Grecco M, Micenmacher S, Martinez-Noel GA, Panelo L, Costas MA. Nuclear receptor coactivator RAC3 inhibits autophagy. Cancer Sci 2012; 103:2064-71. [PMID: 22957814 DOI: 10.1111/cas.12019] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Revised: 08/16/2012] [Accepted: 08/23/2012] [Indexed: 12/16/2022] Open
Abstract
RAC3 is an oncogene naturally overexpressed in several tumors. Besides its role as coactivator, it can exert several protumoral cytoplasmic actions. Autophagy was found to act either as a tumor suppressor during the early stages of tumor development, or as a protector of the tumor cell in later stages under hypoxic conditions. We found that RAC3 overexpression inhibits autophagy when induced by starvation or rapamycin and involves RAC3 nuclear translocation-dependent and -independent mechanisms. Moreover, hypoxia inhibits the RAC3 gene expression leading to the autophagy process, allowing tumor cells to survive until angiogenesis occurs. The interplay between RAC3, hypoxia, and autophagy could be an important mechanism for tumor progression and a good target for a future anticancer therapy.
Collapse
|
43
|
Tien JCY, Xu J. Steroid receptor coactivator-3 as a potential molecular target for cancer therapy. Expert Opin Ther Targets 2012; 16:1085-96. [PMID: 22924430 DOI: 10.1517/14728222.2012.718330] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Steroid receptor coactivator-3 (SRC-3), also called amplified-in-breast cancer-1 (AIB1), is an oncogenic coactivator in endocrine and non-endocrine cancers. Functional studies demonstrate SRC-3 promotes numerous aspects of cancer, through its capacity as a coactivator for nuclear hormone receptors and other transcription factors, and via its ability to control multiple growth pathways simultaneously. Targeting SRC-3 with specific inhibitors therefore holds future promise for clinical cancer therapy. AREAS COVERED We discuss critical advances in understanding SRC-3 as a cancer mediator and prospective drug target. We review SRC-3 structure and function and its role in distinct aspects of cancer. In addition, we discuss SRC-3 regulation and degradation. Finally, we comment on a recently discovered SRC-3 small molecular inhibitor. EXPERT OPINION Most targeted chemotherapeutic drugs block only a single cellular pathway. In response, cancers frequently acquire resistance by upregulating alternative pathways. SRC-3 coordinates multiple signaling networks, suggesting SRC-3 inhibition offers a promising therapeutic strategy. Development of an effective SRC-3 inhibitor faces critical challenges. Better understanding of SRC-3 function and interacting partners, in both the nucleus and cytosol, is required for optimized inhibitor development. Ultimately, blockade of SRC-3 oncogenic function may inhibit multiple cancer-related signaling pathways.
Collapse
Affiliation(s)
- Jean Ching-Yi Tien
- Baylor College of Medicine, Department of Molecular and Cellular Biology, One Baylor Plaza, Houston, TX 77030, USA
| | | |
Collapse
|
44
|
The nuclear cofactor RAC3/AIB1/SRC-3 enhances Nrf2 signaling by interacting with transactivation domains. Oncogene 2012; 32:514-27. [PMID: 22370642 PMCID: PMC3538952 DOI: 10.1038/onc.2012.59] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2, NM 006164, 605 AA) is essential for the antioxidant responsive element (ARE)-mediated expression of a group of detoxifying antioxidant genes that detoxify carcinogens and protect against oxidative stress. Several proteins have been identified as Nrf2-interacting molecules. In this study, we found that the overexpression of receptor-associated coactivator 3 (RAC3)/AIB-1/steroid receptor coactivator-3, a nuclear coregulator and oncogene frequently amplified in human breast cancers, induced heme oxygenase-1 (HO-1) through Nrf2 transactivation in HeLa cells. Next, we determined the interaction between RAC3 and Nrf2 proteins using a co-immunoprecipitation assay and fluorescence resonance energy transfer analysis. The results showed that RAC3 bound directly to the Nrf2 protein in the nucleus. Subsequently, we identified the interacting domains of Nrf2 and RAC3 using a glutathione S-transferase pull-down assay. The results showed that both the N-terminal RAC3-pasB and C-terminal RAC3-R3B3 domains were tightly bound to the Neh4 and Neh5 transactivation domains. Furthermore, chromatin immunoprecipitation showed that RAC3 bound tightly to the ARE enhancer region of the HO-1 promoter via Nrf2 binding. These data suggest that Nrf2 activation is modulated and directly controlled through interactions with the RAC3 protein in HeLa cells.
Collapse
|
45
|
Role of the nuclear receptor coactivator AIB1/SRC-3 in angiogenesis and wound healing. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 180:1474-84. [PMID: 22342158 DOI: 10.1016/j.ajpath.2011.12.032] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Revised: 11/23/2011] [Accepted: 12/22/2011] [Indexed: 12/19/2022]
Abstract
The nuclear receptor coactivator amplified in breast cancer 1 (AIB1/SRC-3) has a well-defined role in steroid and growth factor signaling in cancer and normal epithelial cells. Less is known about its function in stromal cells, although AIB1/SRC-3 is up-regulated in tumor stroma and may, thus, contribute to tumor angiogenesis. Herein, we show that AIB1/SRC-3 depletion from cultured endothelial cells reduces their proliferation and motility in response to growth factors and prevents the formation of intact monolayers with tight junctions and of endothelial tubes. In AIB1/SRC-3(+/-) and (-/-) mice, the angiogenic responses to subcutaneous Matrigel implants was reduced by two-thirds, and exogenously added fibroblast growth factor (FGF) 2 did not overcome this deficiency. Furthermore, AIB1/SRC-3(+/-) and (-/-) mice showed similarly delayed healing of full-thickness excisional skin wounds, indicating that both alleles were required for proper tissue repair. Analysis of this defective wound healing showed reduced recruitment of inflammatory cells and macrophages, cytokine induction, and metalloprotease activity. Skin grafts from animals with different AIB1 genotypes and subsequent wounding of the grafts revealed that the defective healing was attributable to local factors and not to defective bone marrow responses. Indeed, wounds in AIB1(+/-) mice showed reduced expression of FGF10, FGFBP3, FGFR1, FGFR2b, and FGFR3, major local drivers of angiogenesis. We conclude that AIB1/SRC-3 modulates stromal cell responses via cross-talk with the FGF signaling pathway.
Collapse
|
46
|
Song JM, Lu M, Liu FF, Du XJ, Xing BC. AIB1 as an independent prognostic marker in hepatocellular carcinoma after hepatic resection. J Gastrointest Surg 2012; 16:356-60. [PMID: 22052107 DOI: 10.1007/s11605-011-1762-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Accepted: 10/16/2011] [Indexed: 01/31/2023]
Abstract
BACKGROUND Amplified in breast cancer 1 (AIB1) has been shown to promote growth and invasion in several types of human cancers and to have a prognostic role in some of cancers. However, its prognostic significance in hepatocellular carcinoma (HCC) remains unknown. This study aimed to address the issue. METHODS Immunohistochemical staining of AIB1 was performed for HCC and paired paratumorous liver (PTL) tissues from 139 patients. Associations between AIB1 expression with clinicopathological variables and patient survival were evaluated. RESULTS The expression rate of AIB1 was significantly higher in HCC (71/139, 51.1%) than in PTL tissues (1/139, 0.72%, P < 0.001). AIB1 expression in HCC was significantly associated with serum α-fetoprotein levels (P = 0.001) and Edmondson-Steiner grade (P = 0.038). Higher AIB1 expression in HCC was associated with shorter cumulative overall survival of the patients. Multivariate Cox regression analysis revealed that AIB1 was of independent prognostic significance for HCC. CONCLUSIONS AIB1 is independently associated with poor prognosis of HCC.
Collapse
Affiliation(s)
- Jun-Min Song
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Hepatic, Biliary, and Pancreatic Surgery Unit I, Peking University School of Oncology, Beijing Cancer Hospital and Institute, 52 Fu Cheng Road, Beijing, 100142, China
| | | | | | | | | |
Collapse
|
47
|
Johnson AB, O'Malley BW. Steroid receptor coactivators 1, 2, and 3: critical regulators of nuclear receptor activity and steroid receptor modulator (SRM)-based cancer therapy. Mol Cell Endocrinol 2012; 348:430-9. [PMID: 21664237 PMCID: PMC3202666 DOI: 10.1016/j.mce.2011.04.021] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2011] [Revised: 04/04/2011] [Accepted: 04/22/2011] [Indexed: 01/17/2023]
Abstract
Coactivators are a diverse group of non-DNA binding proteins that induce structural changes in agonist-bound nuclear receptors (NRs) that are essential for NR-mediated transcriptional activation. Once bound, coactivators function to bridge enhancer binding proteins to the general transcription machinery, as well as to recruit secondary coactivators that modify promoter and enhancer chromatin in a manner permissive for transcriptional activation. In the following review article, we focus on one of the most in-depth studied families of coactivators, the steroid receptor coactivators (SRC) 1, 2, and 3. SRCs are widely implicated in NR-mediated diseases, especially in cancers, with the majority of studies focused on their roles in breast cancer. We highlight the relevant literature supporting the oncogenic activity of SRCs and their future as diagnostic and prognostic indicators. With much interest in the development of selective receptor modulators (SRMs), we focus on how these coactivators regulate the interactions between SRMs and their respective NRs; and, importantly, the influence that coactivators have on the functional output of SRMs. Furthermore, we speculate that coactivator-specific inhibitors could provide powerful, all-encompassing treatments that target multiple modes of oncogenic regulation in cancers resistant to typical anti-endocrine treatments.
Collapse
Affiliation(s)
- Amber B Johnson
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, United States
| | | |
Collapse
|
48
|
Wang Y, Lonard DM, Yu Y, Chow DC, Palzkill TG, O'Malley BW. Small molecule inhibition of the steroid receptor coactivators, SRC-3 and SRC-1. Mol Endocrinol 2011; 25:2041-53. [PMID: 22053001 DOI: 10.1210/me.2011-1222] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Overexpression of steroid receptor coactivator (SRC)-1 and SRC-3 is associated with cancer initiation, metastasis, advanced disease, and resistance to chemotherapy. In most of these cases, SRC-1 and SRC-3 have been shown to promote tumor cell growth by activating nuclear receptor and multiple growth factor signaling cascades that lead to uncontrolled tumor cell growth. Up until now, most targeted chemotherapeutic drugs have been designed largely to block a single pathway at a time, but cancers frequently acquire resistance by switching to alternative growth factor pathways. We reason that the development of chemotherapeutic agents against SRC coactivators that sit at the nexus of multiple cell growth signaling networks and transcriptional factors should be particularly effective therapeutics. To substantiate this hypothesis, we report the discovery of 2,2'-bis-(Formyl-1,6,7-trihydroxy-5-isopropyl-3-methylnaphthalene (gossypol) as a small molecule inhibitor of coactivator SRC-1 and SRC-3. Our data indicate that gossypol binds directly to SRC-3 in its receptor interacting domain. In MCF-7 breast cancer cells, gossypol selectively reduces the cellular protein concentrations of SRC-1 and SRC-3 without generally altering overall protein expression patterns, SRC-2, or other coactivators, such as p300 and coactivator-associated arginine methyltransferase 1. Gossypol reduces the concentration of SRC-3 in prostate, lung, and liver cancer cell lines. Gossypol inhibits cell viability in the same cancer cell lines where it promotes SRC-3 down-regulation. Additionally, gossypol sensitizes lung and breast cancer cell lines to the inhibitory effects of other chemotherapeutic agents. Importantly, gossypol is selectively cytotoxic to cancer cells, whereas normal cell viability is not affected. This data establish the proof-of-principle that, as a class, SRC-1 and SRC-3 coactivators are accessible chemotherapeutic targets. Given their function as integrators of multiple cell growth signaling systems, SRC-1/SRC-3 small molecule inhibitors comprise a new class of drugs that have potential as novel chemotherapeutics able to defeat aspects of acquired cancer cell resistance mechanisms.
Collapse
Affiliation(s)
- Ying Wang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | |
Collapse
|
49
|
Lee K, Lee A, Song BJ, Kang CS. Expression of AIB1 protein as a prognostic factor in breast cancer. World J Surg Oncol 2011; 9:139. [PMID: 22035181 PMCID: PMC3235064 DOI: 10.1186/1477-7819-9-139] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Accepted: 10/29/2011] [Indexed: 12/01/2022] Open
Abstract
Background AIB1 (amplified in breast cancer I) is a member of the p160 steroid receptor coactivator family. AIB1 is frequently overexpressed in breast cancer and has functions that promote oncogenesis that are independent of estrogen receptor (ER) coactivation. We investigated prognostic significance of AIB1 and relationship between AIB1 and ER, progesterone receptor (PR), androgen receptor (AR), DAX-1, and HER2. Methods RNA in situ hybridization (ISH) and immunohistochemical (IHC) staining for AIB1, IHC staining for ER and the progesterone receptor (PR) and IHC staining and silver in situ hybridization (SISH) for HER2 were performed for 185 breast cancer cases. Results A high level of expression of AIB1 mRNA was observed in 60.0% of tumors. IHC analysis detected AIB1 positivity in 47.3% of tumors, which did not correlate with AIB1 mRNA expression (p = 0.24, r = 0.10). AIB1 protein expression correlated with AR and DAX-1 expression (p = 0.01, r = 0.22 and p = 0.02, r = 0.21, respectively) but not with ER or PR expression (p = 0.14, r = -0.13 and p = 0.16, r = -0.12, respectively). AIB1 protein expression correlated with the amplification of the HER2 gene (p = 0.03, r = 0.19). In contrast to AIB1 protein expression, AIB1 mRNA expression did not correlate with AR, DAX-1, ER, and PR expression, and the amplification of the HER2 gene (p > 0.05 for all). There were trends that strong AIB1 protein expression correlated with poorer disease free survival (p = 0.07). Strong AIB1 protein expression correlated with poorer overall survival (p = 0.04). Among the ER-negative subgroup, strong AIB1 protein expression correlated with poorer disease free survival and overall survival (p = 0.01 and p < 0.01, respectively). Conclusions Strong AIB1 protein expression was poor prognostic factor in breast cancer, especially in ER-negative breast cancers. Further investigation is essential to determine whether AIB1 might be effective therapeutic targets for ER-negative breast cancers.
Collapse
Affiliation(s)
- Kyungji Lee
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Seocho-Gu, Seoul 137-701, Korea.
| | | | | | | |
Collapse
|
50
|
Zakharov MN, Pillai BK, Bhasin S, Ulloor J, Istomin AY, Guo C, Godzik A, Kumar R, Jasuja R. Dynamics of coregulator-induced conformational perturbations in androgen receptor ligand binding domain. Mol Cell Endocrinol 2011; 341:1-8. [PMID: 21605623 DOI: 10.1016/j.mce.2011.03.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Revised: 02/24/2011] [Accepted: 03/02/2011] [Indexed: 11/22/2022]
Abstract
Androgen receptor (AR) coregulators modulate ligand-induced gene expression in a tissue specific manner. The molecular events that follow coactivator binding to AR and the mechanisms that govern the sequence-specific effects of AR coregulators are poorly understood. Using consensus coactivator sequence D11-FxxLF and biophysical techniques, we show that coactivator association is followed by conformational rearrangement in AR ligand binding domain (AR-LBD) that is enthalpically and entropically favorable with activation energy of 29.8±4.2 kJ/mol. Further characterization of ARA70 and SRC3-1 based consensus sequences reveal that each coactivator induces a distinct conformational state in the dihydrotestosterone:AR-LBD:coactivator complex. Complementary computational modeling revealed that coactivator induced specific alterations in the backbone flexibility of AR-LBD distant from the site of coactivator binding and that the intramolecular rearrangements in AR-LBD backbone induced by the two coactivator peptides were different. These data suggest that coactivators may impart specificity in the transcriptional machinery by changing the steady-state conformation of AR-LBD. These data provide direct evidence that even in the presence of same ligand, AR-LBD can occupy distinct conformational states depending on its interactions with specific coactivators in the tissues. We posit that this coactivator-specific conformational gating may then dictate subsequent binding partners and interaction/affinity for the DNA-response elements.
Collapse
Affiliation(s)
- Mikhail N Zakharov
- Section of Endocrinology, Boston University School of Medicine, 670 Albany St., Boston, MA 02118, USA
| | | | | | | | | | | | | | | | | |
Collapse
|