1
|
Hantusch B, Kenner L, Stanulović VS, Hoogenkamp M, Brown G. Targeting Androgen, Thyroid Hormone, and Vitamin A and D Receptors to Treat Prostate Cancer. Int J Mol Sci 2024; 25:9245. [PMID: 39273194 PMCID: PMC11394715 DOI: 10.3390/ijms25179245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/20/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
The nuclear hormone family of receptors regulates gene expression. The androgen receptor (AR), upon ligand binding and homodimerization, shuttles from the cytosol into the nucleus to activate gene expression. Thyroid hormone receptors (TRs), retinoic acid receptors (RARs), and the vitamin D receptor (VDR) are present in the nucleus bound to chromatin as a heterodimer with the retinoid X receptors (RXRs) and repress gene expression. Ligand binding leads to transcription activation. The hormonal ligands for these receptors play crucial roles to ensure the proper conduct of very many tissues and exert effects on prostate cancer (PCa) cells. Androgens support PCa proliferation and androgen deprivation alone or with chemotherapy is the standard therapy for PCa. RARγ activation and 3,5,3'-triiodo-L-thyronine (T3) stimulation of TRβ support the growth of PCa cells. Ligand stimulation of VDR drives growth arrest, differentiation, and apoptosis of PCa cells. Often these receptors are explored as separate avenues to find treatments for PCa and other cancers. However, there is accumulating evidence to support receptor interactions and crosstalk of regulatory events whereby a better understanding might lead to new combinatorial treatments.
Collapse
Affiliation(s)
- Brigitte Hantusch
- Department of Pathology, Department for Experimental and Laboratory Animal Pathology, Medical University of Vienna, 1010 Vienna, Austria;
- Comprehensive Cancer Center, Medical University Vienna, 1090 Vienna, Austria
| | - Lukas Kenner
- Department of Pathology, Department for Experimental and Laboratory Animal Pathology, Medical University of Vienna, 1010 Vienna, Austria;
- Comprehensive Cancer Center, Medical University Vienna, 1090 Vienna, Austria
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
- Department of Molecular Biology, Umeå University, 901 87 Umeå, Sweden
- Christian Doppler Laboratory for Applied Metabolomics, Medical University Vienna, 1090 Vienna, Austria
- Center for Biomarker Research in Medicine (CBmed), 8010 Graz, Austria
| | - Vesna S. Stanulović
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; (V.S.S.); (M.H.)
| | - Maarten Hoogenkamp
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; (V.S.S.); (M.H.)
| | - Geoffrey Brown
- School of Biomedical Sciences, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| |
Collapse
|
2
|
Quan T, Cockburn J, Dhesy-Thind S, Bane A, Leong H, Geleff C, Devion C, Ajel N, Jerzak KJ. The Significance of Thyroid Hormone Receptors in Breast Cancer: A Hypothesis-Generating Narrative Review. Curr Oncol 2024; 31:2364-2375. [PMID: 38785457 PMCID: PMC11119174 DOI: 10.3390/curroncol31050176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 03/04/2024] [Accepted: 03/20/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Breast cancer (BC) is frequently diagnosed among Canadian women. While targeted therapies are available for most BC patients; treatment resistance is common and novel therapeutic targets are of interest. Thyroid hormones (TH) bound to thyroid hormone receptors (THR) influence cell proliferation and differentiation; they are also involved in the growth and development of normal breast tissue. Evidence suggests that THRβ is a tumor suppressor in various solid tumors. PURPOSE This narrative review discusses retrospective studies regarding the clinical relevance of THRβ as a potential prognostic biomarker and therapeutic target in BC. METHODS We consulted with an information specialist to develop a search strategy to find all literature related to THRα expression as a potential prognostic and therapeutic biomarker in breast cancer. The primary search was developed for Medline and translated to Embase. The searches were conducted on the Ovid platform on 18 August 2023. RESULTS Across seven retrospective studies identified, several have shown an association between higher THRβ1 expression with a lower risk of BC recurrence and with longer overall survival. CONCLUSIONS Some evidence suggests that THRβ expression is associated with a lower risk of BC recurrence and death. Validation of THRβ as an independent prognostic biomarker and possible predictive biomarker of response to endocrine therapy and/or chemotherapy is of interest. Given that THRβ is upstream of the AKT/PI3K pathway, its potential as a predictive biomarker of response to AKT inhibitors and/or PI3K inhibitors may also be of value. Finally, the potential re-purposing of THRβ agonists as anti-cancer agents warrants investigation.
Collapse
Affiliation(s)
- Trinity Quan
- Division of Medical Oncology, Sunnybrook Odette Cancer Centre, University of Toronto, Toronto, ON M4N 3M5, Canada; (T.Q.); (N.A.)
| | - Jessica Cockburn
- University Health Network, Toronto, ON M5G 2C4, Canada; (J.C.); (A.B.)
| | - Sukhbinder Dhesy-Thind
- Department of Oncology, Juravinski Cancer Centre, 699 Concession St, Hamilton, ON L8V 5C2, Canada;
| | - Anita Bane
- University Health Network, Toronto, ON M5G 2C4, Canada; (J.C.); (A.B.)
| | - Hon Leong
- Department of Medical Biophysics Temerty, Faculty of Medicine, University of Toronto, Toronto, ON M4N 3M5, Canada; (H.L.); (C.G.)
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
| | - Christopher Geleff
- Department of Medical Biophysics Temerty, Faculty of Medicine, University of Toronto, Toronto, ON M4N 3M5, Canada; (H.L.); (C.G.)
| | - Catherine Devion
- Library Services, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada;
| | - Noor Ajel
- Division of Medical Oncology, Sunnybrook Odette Cancer Centre, University of Toronto, Toronto, ON M4N 3M5, Canada; (T.Q.); (N.A.)
| | - Katarzyna J. Jerzak
- Division of Medical Oncology, Sunnybrook Odette Cancer Centre, University of Toronto, Toronto, ON M4N 3M5, Canada; (T.Q.); (N.A.)
- Library Services, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada;
| |
Collapse
|
3
|
Redfern A, Agarwal V, Alahari S. Editorial: Reviews and advances in the molecular mechanisms of breast cancer. Front Cell Dev Biol 2024; 12:1380475. [PMID: 38516129 PMCID: PMC10955372 DOI: 10.3389/fcell.2024.1380475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 02/06/2024] [Indexed: 03/23/2024] Open
Affiliation(s)
- A. Redfern
- School of Medicine, University of Western Australia, Perth, WA, Australia
- Harry Perkins Institute of Medical Research, Perth, WA, Australia
- Department of Medical Oncology, Fiona Stanley Hospital, Perth, WA, Australia
| | - V. Agarwal
- Department of Medical Oncology, Fiona Stanley Hospital, Perth, WA, Australia
| | - S. Alahari
- Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University, New Orleans, LA, United States
| |
Collapse
|
4
|
Zhang A, Wang R, Liu Q, Yang Z, Lin X, Pang J, Li X, Wang D, He J, Li J, Zhang M, Yu Y, Cao XC, Chen X, Tang NJ. Breast adipose metabolites mediates the association of tetrabromobisphenol a with breast cancer: A case-control study in Chinese population. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 316:120701. [PMID: 36423888 DOI: 10.1016/j.envpol.2022.120701] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 11/13/2022] [Accepted: 11/17/2022] [Indexed: 06/16/2023]
Abstract
Studies exploring the association of tetrabromobisphenol A (TBBPA) with breast cancer and related mechanisms are limited. To investigate the relationship between TBBPA levels in breast adipose and breast cancer, we carried out case-control research. As well as further examine the mediating role of adipose metabolites between TBBPA and breast cancer using the metabolomics approach. In this study, the concentration of TBBPA was determined utilizing ultra-performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS) after a solid phase extraction (SPE) pretreatment. High-performance liquid chromatography tandem mass spectrometry (HPLC-MS/MS) was employed to analyze adipose metabolomics. Evaluation of metabolites linked to TBBPA exposure and breast cancer was performed utilizing mediation analysis. With an estimated OR (95%CI) of 1.153 (1.023, 1.299), TBBPA was firmly linked with breast cancer. We also used propensity score matching analysis and sensitivity analysis to reduce the effect of confounding factors on the results. Metabolomics of adipose suggested significant perturbation in the linoleic acid metabolism pathway. In addition, for PC (16:0/16:0) as phospholipids, a mediation effect on the associations of TBBPA exposure with breast cancer risks was observed (estimated mediation percentage: 56.58%). Understanding the relationship between TBBPA exposure and the risk of breast cancer may be facilitated by the findings, which point to potential mediation metabolites.
Collapse
Affiliation(s)
- Ai Zhang
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| | - Rui Wang
- Health Inspection and Testing Institute Integrated Operations Section, Tianjin Centers for Disease Control and Prevention, Tianjin, 300011, China
| | - Qianfeng Liu
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| | - Zhengjun Yang
- Tianjin Medical University Cancer Institute and Hospital: Tianjin Tumor Hospital, Tianjin, 300060, China
| | - Xiaohui Lin
- Health Inspection and Testing Institute Physical and Chemical Section, Tianjin Centers for Disease Control and Prevention, Tianjin, 300011, China
| | - Jing Pang
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| | - Xiaoyu Li
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| | - Dan Wang
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| | - Jiayu He
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| | - Jianping Li
- Health Inspection and Testing Institute Physical and Chemical Section, Tianjin Centers for Disease Control and Prevention, Tianjin, 300011, China
| | - Mingyue Zhang
- Tianjin Centers for Disease Control and Prevention, Tianjin, 300011, China
| | - Yue Yu
- Tianjin Medical University Cancer Institute and Hospital: Tianjin Tumor Hospital, Tianjin, 300060, China
| | - Xu-Chen Cao
- Tianjin Medical University Cancer Institute and Hospital: Tianjin Tumor Hospital, Tianjin, 300060, China
| | - Xi Chen
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| | - Nai-Jun Tang
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
5
|
Dhingra M, Mahalanobis S, Das A. Thyroid receptor β might be responsible for breast cancer associated with Hashimoto's thyroiditis: a new insight into pathogenesis. Immunol Res 2022; 70:441-448. [PMID: 35562625 DOI: 10.1007/s12026-022-09288-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 05/04/2022] [Indexed: 11/30/2022]
Abstract
Breast cancer is the most common cancer affecting females worldwide. Often it is observed that women suffering from Hashimoto's thyroiditis exhibit a greater propensity towards development of breast cancer. The exact mechanism for the same is unknown. However, multiple experimental evidences suggest a significant role of thyroid receptor β (TR-β) in regulating cell growth and proliferation and thus play a potent role as a tumor suppressor in several cancers, including breast cancer. Thyroid receptor β shows anti-proliferative action through mediators like β-catenin, RUNX2, PI3K/AKT, and cyclin regulation. The present review explores the link between these pathways and how they may be dysregulated due to Hashimoto's thyroiditis. Further, we propose a new mechanism for cancer prognosis associated with Hashimoto's thyroiditis, which may lead to the development of TR-β targeting as a novel therapeutic approach.
Collapse
Affiliation(s)
- Muskaan Dhingra
- Department of Biotechnology, Delhi Technological University, Delhi, 110042, India
| | - Shayon Mahalanobis
- Department of Biotechnology, Delhi Technological University, Delhi, 110042, India
| | - Asmita Das
- Department of Biotechnology, Delhi Technological University, Delhi, 110042, India.
| |
Collapse
|
6
|
Voutsadakis IA. The TSH/Thyroid Hormones Axis and Breast Cancer. J Clin Med 2022; 11:687. [PMID: 35160139 PMCID: PMC8836919 DOI: 10.3390/jcm11030687] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/22/2022] [Accepted: 01/27/2022] [Indexed: 12/20/2022] Open
Abstract
Breast cancer, the most prevalent female carcinoma, is characterized by the expression of steroid nuclear receptors in a subset of cases. The most important nuclear receptor with prognostic and therapeutic implications is the Estrogen Receptor (ER), which is expressed in about three out of four breast cancers. The Progesterone Receptor (PR) and the Androgen Receptor (AR) are also commonly expressed. Moreover, non-steroid nuclear receptors, including the vitamin D receptor (VDR) and the thyroid receptors (TRs), are also present in breast cancers and have pathophysiologic implications. Circulating thyroid hormones may influence breast cancer risk and breast cancer cell survival, through ligating their canonical receptors TRα and TRβ but also through additional membrane receptors that are expressed in breast cancer. The expression of TR subtypes and their respective isotypes have diverse effects in breast cancers through co-operation with ER and influence on other cancer-associated pathways. Other components of the TSH/thyroid hormone axis, such as TSH and selenoiodinase enzymes, have putative effects in breast cancer pathophysiology. This paper reviews the pathophysiologic and prognostic implications of the thyroid axis in breast cancer and provides a brief therapeutic perspective.
Collapse
Affiliation(s)
- Ioannis A. Voutsadakis
- Algoma District Cancer Program, Sault Area Hospital, Sault Ste. Marie, ON P6B 0A8, Canada; or
- Section of Internal Medicine, Division of Clinical Sciences, Northern Ontario School of Medicine, Sudbury, ON P6B 0A8, Canada
| |
Collapse
|
7
|
Ezh2 promotes TRβ lysine methylation-mediated degradation in hepatocellular carcinoma. Genes Genomics 2021; 44:369-377. [PMID: 34851506 DOI: 10.1007/s13258-021-01196-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 11/23/2021] [Indexed: 01/01/2023]
Abstract
BACKGROUND Post-translational modification (PTM) of proteins controls various cellular functions of transcriptional regulators and participates in diverse signal transduction pathways in cancer. The thyroid hormone (triiodothyronine, T3) plays a critical role in metabolic homeostasis via its direct interaction with the thyroid hormone receptor beta (TRβ). TRβ is involved in physiological processes, such as cell growth, differentiation, apoptosis, and maintenance of metabolic homeostasis through transcriptional regulation of target genes. OBJECTIVE This study was performed to characterize the specific PTM of TRβ is an active control mechanism for the proteasomal degradation of TRβ in transcriptional signaling pathways in hepatocellular carcinoma cells. METHODS Based on a previous study, we predicted that the lysine methyltransferase and methylation sites of TRβ by comparing the amino acid sequences of histone H3 and TRβ. Methyl-acceptor site of TRβ was confirmed by point mutation. TRβ protein stability was evaluated by ubiquitination assay with MG132. For glucose starvation, HepG2 cells were incubated in media without D-glucose. Proliferation-related proteins were detected by western blotting. MicroRNA level and autophagy marker were measured by real-time qPCR. RESULTS The presence of enhancer of zeste homolog 2 (Ezh2), a methyltransferase of H3 lysine 27, as a methyltransferase of TRβ also revealed that direct lysine methylation and consequent stimulated protein degradation of TRβ underlies the negative correlation between Ezh2 and TRβ. Notably, glucose starvation significantly increased lysine methylation, and methylated TRβ showed further protein instability leading to an increase in the proliferation and growth of hepatocellular carcinoma cells. CONCLUSIONS TRβ functions as a tumor suppressor in various cancers; therefore, we evaluated the effect of TRβ degradation on oncogenesis during glucose starvation. These data clearly define a functional model and provide a link between metabolism and cancer by regulating methyl-dependent protein levels of tumor suppressors. Taken together, maintaining TRβ against methyl-dependent degradation is considered a possible therapeutic target for cancer progression.
Collapse
|
8
|
Bolf EL, Gillis NE, Davidson CD, Cozzens LM, Kogut S, Tomczak JA, Frietze S, Carr FE. Common tumor-suppressive signaling of thyroid hormone receptor beta in breast and thyroid cancer cells. Mol Carcinog 2021; 60:874-885. [PMID: 34534367 DOI: 10.1002/mc.23352] [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] [Received: 06/08/2021] [Revised: 09/01/2021] [Accepted: 09/02/2021] [Indexed: 01/16/2023]
Abstract
The thyroid hormone receptor beta (TRβ) is a tumor suppressor in multiple types of solid tumors, most prominently in breast and thyroid cancer. An increased understanding of the molecular mechanisms by which TRβ abrogates tumorigenesis will aid in understanding the core tumor-suppressive functions of TRβ. Here, we restored TRβ expression in the MDA-MB-468 basal-like breast cancer cell line and perform RNA-sequencing to determine the TRβ-mediated changes in gene expression and associated signaling pathways. The TRβ expressing MDA-MB-468 cells exhibit a more epithelial character as determined by principle component analysis-based iterative PAM50 subtyping score and through reduced expression of mesenchymal cytokeratins. The epithelial to mesenchymal transition pathway is also significantly reduced. The MDA-MB-468 data set was further compared with RNA sequencing results from TRβ expressing thyroid cancer cell line SW1736 to determine which genes are TRβ correspondingly regulated across both cell types. Several pathways including lipid metabolism and chromatin remodeling processes were observed to be altered in the shared gene set. These data provide novel insights into the molecular mechanisms by which TRβ suppresses breast tumorigenesis.
Collapse
Affiliation(s)
- Eric L Bolf
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA.,University of Vermont Cancer Center, University of Vermont, Burlington, Vermont, USA
| | - Noelle E Gillis
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA.,University of Vermont Cancer Center, University of Vermont, Burlington, Vermont, USA
| | - Cole D Davidson
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA.,University of Vermont Cancer Center, University of Vermont, Burlington, Vermont, USA
| | - Lauren M Cozzens
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Sophie Kogut
- Department of Biomedical and Health Sciences, College of Nursing and Health Sciences, University of Vermont, Burlington, Vermont, USA
| | - Jennifer A Tomczak
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Seth Frietze
- University of Vermont Cancer Center, University of Vermont, Burlington, Vermont, USA.,Department of Biomedical and Health Sciences, College of Nursing and Health Sciences, University of Vermont, Burlington, Vermont, USA
| | - Frances E Carr
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA.,University of Vermont Cancer Center, University of Vermont, Burlington, Vermont, USA
| |
Collapse
|
9
|
Zheng Y, Karnoub AE. Endocrine regulation of cancer stem cell compartments in breast tumors. Mol Cell Endocrinol 2021; 535:111374. [PMID: 34242715 DOI: 10.1016/j.mce.2021.111374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 05/28/2021] [Accepted: 06/01/2021] [Indexed: 10/20/2022]
Abstract
Cancer cells within breast tumors exist within a hierarchy in which only a small and rare subset of cells is able to regenerate growths with the heterogeneity of the original tumor. These highly malignant cancer cells, which behave like stem cells for new cancers and are called "cancer stem cells" or CSCs, have also been shown to possess increased resistance to therapeutics, and represent the root cause underlying therapy failures, persistence of residual disease, and relapse. As >90% of cancer deaths are due to refractory tumors, identification of critical molecular drivers of the CSC-state would reveal vulnerabilities that can be leveraged in designing therapeutics that eradicate advanced disease and improve patient survival outcomes. An expanding and complex body of work has now described the exquisite susceptibility of CSC pools to the regulatory influences of local and systemic hormones. Indeed, breast CSCs express a plethora of hormonal receptors, which funnel hormonal influences over every aspect of breast neoplasia - be it tumor onset, growth, survival, invasion, metastasis, or therapy resistance - via directly impacting CSC behavior. This article is intended to shed light on this active area of investigation by attempting to provide a systematic and comprehensive overview of the available evidence directly linking hormones to breast CSC biology.
Collapse
Affiliation(s)
- Yurong Zheng
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Antoine E Karnoub
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA; Harvard Stem Cell Institute, Cambridge, MA, 02138, USA; Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
| |
Collapse
|
10
|
Hwang SH, Yang Y, Woo SH, Jung JH, Seo D, Kim DY, Kim Y. Thyroglobulin as a negative marker for malignancy in canine and human breast tumors. Mol Carcinog 2021; 60:455-468. [PMID: 34003526 DOI: 10.1002/mc.23304] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/14/2021] [Accepted: 04/16/2021] [Indexed: 11/09/2022]
Abstract
Canine mammary gland tumors (CMTs) are the most common tumor type in female dogs. This study evaluated the expression pattern and role of thyroglobulin (Tg) in CMT and in human breast cancer (HBC). CMT samples were subjected to fine-needle aspiration, primary cell culture, and histopathology. The expression level of Tg was higher in benign CMT than in malignant CMT (mCMT) primary cells, particularly in the epithelial lineage. Moreover, treatment with Tg enhanced the sensitivity of doxorubicin in mCMT epithelial cells and mitigated proinflammatory response by increasing nuclear factor erythroid 2-related factor 2 (Nrf2). The proximal region of the Tg promoter was hypermethylated in mCMT epithelial cells, silencing Tg expression with concurrent downregulation of Nrf2-mediated antioxidant signaling. An identical pattern of Tg expression was observed in cytological and tissue samples. Tissue microarray analysis showed that Tg was highly expressed in normal and benign tissues when compared with their malignant counterparts, which was diminished along with higher histological grades. The survival rate was significantly higher in HBC patients with high Tg expression than those with low Tg expression. This study also showed that the progression of HBC is accompanied by the reduction of Tg expression along with augmentation of proinflammatory signaling. Our data suggested that Tg could be a negative indicator of malignancy in canine and human breast neoplasia.
Collapse
Affiliation(s)
- Sung-Hyun Hwang
- Laboratory of Clinical Pathology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.,BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Yeseul Yang
- Laboratory of Clinical Pathology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.,BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Sang-Ho Woo
- BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.,Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Jae-Ha Jung
- Laboratory of Clinical Pathology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.,BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Dansong Seo
- Laboratory of Clinical Pathology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.,BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Dae-Yong Kim
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.,Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Yongbaek Kim
- Laboratory of Clinical Pathology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.,Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
11
|
Identification of Potential Prognostic Biomarkers for Breast Cancer Based on lncRNA-TF-Associated ceRNA Network and Functional Module. BIOMED RESEARCH INTERNATIONAL 2021; 2020:5257896. [PMID: 32802855 PMCID: PMC7411464 DOI: 10.1155/2020/5257896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/23/2020] [Accepted: 06/29/2020] [Indexed: 11/17/2022]
Abstract
Breast cancer leads to most of cancer deaths among women worldwide. Systematically analyzing the competing endogenous RNA (ceRNA) network and their functional modules may provide valuable insight into the pathogenesis of breast cancer. In this study, we constructed a lncRNA-TF-associated ceRNA network via combining all the significant lncRNA-TF ceRNA pairs and TF-TF PPI pairs. We computed important topological features of the network, such as degree and average path length. Hub nodes in the lncRNA-TF-associated ceRNA network were extracted to detect differential expression in different subtypes and tumor stages of breast cancer. MCODE was used for identifying the closely connected modules from the ceRNA network. Survival analysis was further used for evaluating whether the modules had prognosis effects on breast cancer. TF motif searching analysis was performed for investigating the binding potentials between lncRNAs and TFs. As a result, a lncRNA-TF-associated ceRNA network in breast cancer was constructed, which had a scale-free property. Hub nodes such as MDM4, ZNF410, AC0842-19, and CTB-89H12 were differentially expressed between cancer and normal sample in different subtypes and tumor stages. Two closely connected modules were identified to significantly classify patients into a low-risk group and high-risk group with different clinical outcomes. TF motif searching analysis suggested that TFs, such as NFAT5, might bind to the promoter and enhancer regions of hub lncRNAs and function in breast cancer biology. The results demonstrated that the synergistic, competitive lncRNA-TF ceRNA network and their functional modules played important roles in the biological processes and molecular functions of breast cancer.
Collapse
|
12
|
Zhang W, Li A, Pan Y, Wang F, Li M, Liang Y, Yao X, Song J, Song M, Jiang G. Tetrabromobisphenol A induces THR β-mediated inflammation and uterine injury in mice at environmentally relevant exposure concentrations. JOURNAL OF HAZARDOUS MATERIALS 2021; 407:124859. [PMID: 33360189 DOI: 10.1016/j.jhazmat.2020.124859] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 11/19/2020] [Accepted: 12/12/2020] [Indexed: 06/12/2023]
Abstract
Tetrabromobisphenol A (TBBPA) is a widely used flame retardant, but the adverse outcomes induced by TBBPA has not been fully elucidated. In this study, TBBPA was detected in 54.9% of 102 female Chinese volunteers with an average serum concentration of 0.34 ng/mL. To investigate whether TBBPA induces adverse outcomes at environmentally relevant exposure doses, the mice were exposed to TBBPA for 14 and 28 days. The internal doses of TBBPA in mice serum were nearly the internal doses in volunteers. TBBPA significantly increased the secretion of some pro-inflammatory cytokines and suppressed immune responses in mice under such serum concentrations after 14- and 28-days exposure. Interestingly, uterine edema was observed in TBBPA-treated mice. In primary uterine cells model, the results showed TBBPA exposure suppressed THRβ expression, leading to the activation of the inflammatory PI3K/NF-κB signaling pathway. Our findings indicated that the uterus is the susceptible target organ of TBBPA and TBBPA exposure might increase risk of uterine cancer through deregulating inflammation pathways.
Collapse
Affiliation(s)
- Wenjuan Zhang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China; Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, Institute of Environment and Health, Jianghan University, Wuhan 430056, Hubei, PR China
| | - Aijing Li
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China
| | - Yu Pan
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, Institute of Environment and Health, Jianghan University, Wuhan 430056, Hubei, PR China
| | - Fengbang Wang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China
| | - Ming Li
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, Institute of Environment and Health, Jianghan University, Wuhan 430056, Hubei, PR China
| | - Yong Liang
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, Institute of Environment and Health, Jianghan University, Wuhan 430056, Hubei, PR China
| | - Xinglei Yao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China
| | - Jinghai Song
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Beijing 1000730, PR China
| | - Maoyong Song
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China.
| | - Guibin Jiang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China
| |
Collapse
|
13
|
Malavia N, Kuche K, Ghadi R, Jain S. A bird's eye view of the advanced approaches and strategies for overshadowing triple negative breast cancer. J Control Release 2020; 330:72-100. [PMID: 33321156 DOI: 10.1016/j.jconrel.2020.12.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/07/2020] [Accepted: 12/09/2020] [Indexed: 12/12/2022]
Abstract
Triple negative breast cancer (TNBC) is one of the most aggressive form of breast cancer. It is characterized by the absence of estrogen, progesterone and human epidermal growth factor receptors. The main issue with TNBC is that it exhibits poor prognosis, high risk of relapse, short progression-free survival and low overall survival in patients. This is because the conventional therapy used for managing TNBC has issues pertaining to poor bioavailability, lower cellular uptake, increased off-target effects and development of resistance. To overcome such pitfalls, several other approaches are explored. In this context, the present manuscript showcases three of the most widely used approaches which are (i) nanotechnology-based approach; (ii) gene therapy approach and (iii) Phytochemical-based approach. The ultimate focus is to present and explain the insightful reports based on these approaches. Further, the review also expounds on the identified molecular targets and novel targeting ligands which are explored for managing TNBC effectively. Thus, in a nutshell, the review tries to highlight these existing treatment approaches which might inspire for future development of novel therapies with a potential of overshadowing TNBC.
Collapse
Affiliation(s)
- Nilesh Malavia
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Mohali, Punjab, India
| | - Kaushik Kuche
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Mohali, Punjab, India
| | - Rohan Ghadi
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Mohali, Punjab, India
| | - Sanyog Jain
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Mohali, Punjab, India.
| |
Collapse
|
14
|
Cytoplasmic and Nuclear Forms of Thyroid Hormone Receptor β1 Are Inversely Associated with Survival in Primary Breast Cancer. Int J Mol Sci 2020; 21:ijms21010330. [PMID: 31947762 PMCID: PMC6981495 DOI: 10.3390/ijms21010330] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/24/2019] [Accepted: 12/31/2019] [Indexed: 12/24/2022] Open
Abstract
The aim of this study was to investigate the expression of thyroid hormone receptor β1 (THRβ1) by immunohistochemistry in breast cancer (BC) tissues and to correlate the results with clinico-biological parameters. In a well-characterized cohort of 274 primary BC patients, THRβ1 was widely expressed with a predominant nuclear location, although cytoplasmic staining was also frequently observed. Both nuclear and cytoplasmic THRβ1 were correlated with high-risk BC markers such as human epidermal growth factor receptor 2 (HER2), Ki67 (also known as MKI67), prominin-1 (CD133), and N-cadherin. Overall survival analysis demonstrated that cytoplasmic THRβ1 was correlated with favourable survival (p = 0.015), whereas nuclear THRβ1 had a statistically significant correlation with poor outcome (p = 0.038). Interestingly, in our cohort, nuclear and cytoplasmic THRβ1 appeared to be independent markers either for poor (p = 0.0004) or for good (p = 0.048) prognosis, respectively. Altogether, these data indicate that the subcellular expression of THRβ1 may play an important role in oncogenesis. Moreover, the expression of nuclear THRβ1 is a negative outcome marker, which may help to identify high-risk BC subgroups.
Collapse
|
15
|
López-Mateo I, Alonso-Merino E, Suarez-Cabrera C, Park JW, Cheng SY, Alemany S, Paramio JM, Aranda A. Thyroid Hormone Receptor β Inhibits Self-Renewal Capacity of Breast Cancer Stem Cells. Thyroid 2020; 30:116-132. [PMID: 31760908 PMCID: PMC6998057 DOI: 10.1089/thy.2019.0175] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background: A subpopulation of cancer stem cells (CSCs) with capacity for self-renewal is believed to drive initiation, progression, and relapse of breast tumors. Methods: Since the thyroid hormone receptor β (TRβ) appears to suppress breast tumor growth and metastasis, we have analyzed the possibility that TRβ could affect the CSC population using MCF-7 cells grown under adherent conditions or as mammospheres, as well as inoculation into immunodeficient mice. Results: Treatment of TRβ-expressing MCF-7 cells (MCF7-TRβ cells) with the thyroid hormone triiodothyronine (T3) decreased significantly CD44+/CD24- and ALDH+ cell subpopulations, the efficiency of mammosphere formation, the self-renewal capacity of CSCs in limiting dilution assays, the expression of the pluripotency factors in the mammospheres, and tumor initiating capacity in immunodeficient mice, indicating that the hormone reduces the CSC population present within the bulk MCF7-TRβ cultures. T3 also decreased migration and invasion, a hallmark of CSCs. Transcriptome analysis showed downregulation of the estrogen receptor alpha (ERα) and ER-responsive genes by T3. Furthermore, among the T3-repressed genes, there was an enrichment in genes containing binding sites for transcription factors that are key determinants of luminal-type breast cancers and are required for ER binding to chromatin. Conclusion: We demonstrate a novel role of TRβ in the biology of CSCs that may be related to its action as a tumor suppressor in breast cancer.
Collapse
Affiliation(s)
- Irene López-Mateo
- Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Instituto de Investigaciones Biomédicas “Alberto Sols,” Madrid, Spain
| | - Elvira Alonso-Merino
- Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Instituto de Investigaciones Biomédicas “Alberto Sols,” Madrid, Spain
| | | | - Jeong Won Park
- Laboratory of Molecular Biology, National Cancer Institute, Bethesda, Maryland
| | - Sheue-yann Cheng
- Laboratory of Molecular Biology, National Cancer Institute, Bethesda, Maryland
| | - Susana Alemany
- Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Instituto de Investigaciones Biomédicas “Alberto Sols,” Madrid, Spain
| | - Jesús M. Paramio
- Molecular Oncology Unit, CIEMAT, Madrid, Spain
- Institute of Biomedical Research, Hosp Univ. “12 de Octubre,” Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Ana Aranda
- Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Instituto de Investigaciones Biomédicas “Alberto Sols,” Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Address correspondence to: Ana Aranda, PhD, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Instituto de Investigaciones Biomédicas “Alberto Sols”, Arturo Duperier 4, Madrid 28029, Spain
| |
Collapse
|
16
|
Bolf EL, Gillis NE, Barnum MS, Beaudet CM, Yu GY, Tomczak JA, Stein JL, Lian JB, Stein GS, Carr FE. The Thyroid Hormone Receptor-RUNX2 Axis: A Novel Tumor Suppressive Pathway in Breast Cancer. Discov Oncol 2019; 11:34-41. [PMID: 31865591 DOI: 10.1007/s12672-019-00373-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 11/25/2019] [Indexed: 01/07/2023] Open
Abstract
Metastatic breast cancer is refractory to conventional therapies and is an end-stage disease. RUNX2 is a transcription factor that becomes oncogenic when aberrantly expressed in multiple tumor types, including breast cancer, supporting tumor progression and metastases. Our previous work demonstrated that the thyroid hormone receptor beta (TRβ) inhibits RUNX2 expression and tumorigenic characteristics in thyroid cells. As TRβ is a tumor suppressor, we investigated the compelling question whether TRβ also regulates RUNX2 in breast cancer. The Cancer Genome Atlas indicates that TRβ expression is decreased in the most aggressive basal-like subtype of breast cancer. We established that modulated levels of TRβ results in corresponding changes in the high levels of RUNX2 expression in metastatic, basal-like breast cells. The MDA-MB-231 triple-negative breast cancer cell line exhibits low expression of TRβ and high levels of RUNX2. Increased expression of TRβ decreased RUNX2 levels. The thyroid hormone-mediated suppression of RUNX2 is TRβ specific as TRα overexpression failed to alter RUNX2 expression. Consistent with these findings, knockdown of TRβ in non-tumor MCF10A mammary epithelial-like cells results in an increase in RUNX2 and RUNX2 target genes. Mechanistically, TRβ directly interacts with the proximal promoter of RUNX2 through a thyroid hormone response element to reduce promoter activity. The TRβ suppression of the oncogene RUNX2 is a signaling pathway shared by thyroid and breast cancers. Our findings provide a novel mechanism for TRβ-mediated tumor suppression in breast cancers. This pathway may be common to many solid tumors and impact treatment for metastatic cancers.
Collapse
Affiliation(s)
- Eric L Bolf
- Department of Pharmacology, University of Vermont, 89 Beaumont Avenue, Burlington, VT, 05405, USA.,University of Vermont Cancer Center, Larner College of Medicine, University of Vermont, 89 Beaumont Avenue, Burlington, VT, 05405, USA
| | - Noelle E Gillis
- Department of Pharmacology, University of Vermont, 89 Beaumont Avenue, Burlington, VT, 05405, USA.,University of Vermont Cancer Center, Larner College of Medicine, University of Vermont, 89 Beaumont Avenue, Burlington, VT, 05405, USA
| | - Michael S Barnum
- Department of Pharmacology, University of Vermont, 89 Beaumont Avenue, Burlington, VT, 05405, USA
| | - Caitlin M Beaudet
- Department of Pharmacology, University of Vermont, 89 Beaumont Avenue, Burlington, VT, 05405, USA
| | - Grace Y Yu
- Department of Pharmacology, University of Vermont, 89 Beaumont Avenue, Burlington, VT, 05405, USA
| | - Jennifer A Tomczak
- Department of Pharmacology, University of Vermont, 89 Beaumont Avenue, Burlington, VT, 05405, USA
| | - Janet L Stein
- University of Vermont Cancer Center, Larner College of Medicine, University of Vermont, 89 Beaumont Avenue, Burlington, VT, 05405, USA.,Department of Biochemistry, University of Vermont, Burlington, VT, 05405, USA
| | - Jane B Lian
- University of Vermont Cancer Center, Larner College of Medicine, University of Vermont, 89 Beaumont Avenue, Burlington, VT, 05405, USA.,Department of Biochemistry, University of Vermont, Burlington, VT, 05405, USA
| | - Gary S Stein
- University of Vermont Cancer Center, Larner College of Medicine, University of Vermont, 89 Beaumont Avenue, Burlington, VT, 05405, USA.,Department of Biochemistry, University of Vermont, Burlington, VT, 05405, USA
| | - Frances E Carr
- Department of Pharmacology, University of Vermont, 89 Beaumont Avenue, Burlington, VT, 05405, USA. .,University of Vermont Cancer Center, Larner College of Medicine, University of Vermont, 89 Beaumont Avenue, Burlington, VT, 05405, USA.
| |
Collapse
|
17
|
Fritz AJ, Gillis NE, Gerrard DL, Rodriguez PD, Hong D, Rose JT, Ghule PN, Bolf EL, Gordon JA, Tye CE, Boyd JR, Tracy KM, Nickerson JA, van Wijnen AJ, Imbalzano AN, Heath JL, Frietze SE, Zaidi SK, Carr FE, Lian JB, Stein JL, Stein GS. Higher order genomic organization and epigenetic control maintain cellular identity and prevent breast cancer. Genes Chromosomes Cancer 2019; 58:484-499. [PMID: 30873710 DOI: 10.1002/gcc.22731] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/07/2019] [Accepted: 01/07/2019] [Indexed: 12/24/2022] Open
Abstract
Cells establish and sustain structural and functional integrity of the genome to support cellular identity and prevent malignant transformation. In this review, we present a strategic overview of epigenetic regulatory mechanisms including histone modifications and higher order chromatin organization (HCO) that are perturbed in breast cancer onset and progression. Implications for dysfunctions that occur in hormone regulation, cell cycle control, and mitotic bookmarking in breast cancer are considered, with an emphasis on epithelial-to-mesenchymal transition and cancer stem cell activities. The architectural organization of regulatory machinery is addressed within the contexts of translating cancer-compromised genomic organization to advances in breast cancer risk assessment, diagnosis, prognosis, and identification of novel therapeutic targets with high specificity and minimal off target effects.
Collapse
Affiliation(s)
- A J Fritz
- Department of Biochemistry, Larner College of Medicine, University of Vermont, Burlington, Vermont.,University of Vermont Cancer Center, Burlington, Vermont
| | - N E Gillis
- University of Vermont Cancer Center, Burlington, Vermont.,Department of Pharmacology, Larner college of Medicine, University of Vermont, Burlington, Vermont
| | - D L Gerrard
- Cellular Molecular Biomedical Sciences Program, University of Vermont, Burlington, Vermont.,Department of Biomedical and Health Sciences, University of Vermont, Burlington, Vermont
| | - P D Rodriguez
- Cellular Molecular Biomedical Sciences Program, University of Vermont, Burlington, Vermont.,Department of Biomedical and Health Sciences, University of Vermont, Burlington, Vermont
| | - D Hong
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts
| | - J T Rose
- Department of Biochemistry, Larner College of Medicine, University of Vermont, Burlington, Vermont.,University of Vermont Cancer Center, Burlington, Vermont
| | - P N Ghule
- Department of Biochemistry, Larner College of Medicine, University of Vermont, Burlington, Vermont.,University of Vermont Cancer Center, Burlington, Vermont
| | - E L Bolf
- University of Vermont Cancer Center, Burlington, Vermont.,Department of Pharmacology, Larner college of Medicine, University of Vermont, Burlington, Vermont
| | - J A Gordon
- Department of Biochemistry, Larner College of Medicine, University of Vermont, Burlington, Vermont.,University of Vermont Cancer Center, Burlington, Vermont
| | - C E Tye
- Department of Biochemistry, Larner College of Medicine, University of Vermont, Burlington, Vermont.,University of Vermont Cancer Center, Burlington, Vermont
| | - J R Boyd
- Department of Biochemistry, Larner College of Medicine, University of Vermont, Burlington, Vermont.,University of Vermont Cancer Center, Burlington, Vermont
| | - K M Tracy
- Department of Biochemistry, Larner College of Medicine, University of Vermont, Burlington, Vermont.,University of Vermont Cancer Center, Burlington, Vermont
| | - J A Nickerson
- Division of Genes and Development of the Department of Pediatrics, University of Massachusetts Medical School, Worcester, Massachusetts
| | - A J van Wijnen
- Orthopedic Surgery and Biochemistry and Molecular Biology, Mayo Clinic Minnesota, Rochester, Minnesota
| | - A N Imbalzano
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts
| | - J L Heath
- Department of Biochemistry, Larner College of Medicine, University of Vermont, Burlington, Vermont.,University of Vermont Cancer Center, Burlington, Vermont.,Department of Pediatrics, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - S E Frietze
- Cellular Molecular Biomedical Sciences Program, University of Vermont, Burlington, Vermont.,Department of Biomedical and Health Sciences, University of Vermont, Burlington, Vermont
| | - S K Zaidi
- Department of Biochemistry, Larner College of Medicine, University of Vermont, Burlington, Vermont.,University of Vermont Cancer Center, Burlington, Vermont
| | - F E Carr
- Department of Biochemistry, Larner College of Medicine, University of Vermont, Burlington, Vermont.,University of Vermont Cancer Center, Burlington, Vermont.,Department of Pharmacology, Larner college of Medicine, University of Vermont, Burlington, Vermont
| | - J B Lian
- Department of Biochemistry, Larner College of Medicine, University of Vermont, Burlington, Vermont.,University of Vermont Cancer Center, Burlington, Vermont
| | - J L Stein
- Department of Biochemistry, Larner College of Medicine, University of Vermont, Burlington, Vermont.,University of Vermont Cancer Center, Burlington, Vermont
| | - G S Stein
- Department of Biochemistry, Larner College of Medicine, University of Vermont, Burlington, Vermont.,University of Vermont Cancer Center, Burlington, Vermont
| |
Collapse
|
18
|
Rodríguez-Molinero A, Hercbergs A, Sarrias M, Yuste A. Plasma 3,3',5-Triiodo-L-thyronine [T3] level mirrors changes in tumor markers in two cases of metastatic cancer of the breast and pancreas treated with exogenous L-T3. Cancer Biomark 2018; 21:433-438. [PMID: 29125479 PMCID: PMC5859424 DOI: 10.3233/cbm-170668] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Preclinical studies have attributed 3,3’,5-triiodo-L-thyronine (T3) a direct negative effect on tumor progression, as well as chemosensitizing, differentiating and immunomodulatory properties. On the other hand, L-thyroxine (T4), via a thyroid hormone receptor on plasma membrane integrin αvβ3, promotes solid tumor growth and neoangiogenesis, therefore lowering endogenous T4 reduces tumor growth rate. We present the case of two patients with metastatic triple negative breast cancer and metastatic pancreatic cancer respectively, who benefit of the sole treatment with antithyroid drugs and exogenous administration of T3 (liothyronine). In these cases tumor growth was accompanied by T3 depletion in plasma, which may represent a novel marker for progression.
Collapse
Affiliation(s)
| | - Aleck Hercbergs
- Departments of Radiation Oncology and Neuroradiology, Cleveland Clinic, Cleveland, OH, USA
| | - Manuel Sarrias
- Department of Radiology, ConsorciSanitari del Garraf, Vilanova i la Geltrú, Spain
| | - Antonio Yuste
- Clinical Research Unit, Consorci Sanitari del Garraf, Vilanova i la Geltrú, Spain
| |
Collapse
|
19
|
Thyroid hormone receptor beta-1 expression in early breast cancer: a validation study. Breast Cancer Res Treat 2018; 171:709-717. [DOI: 10.1007/s10549-018-4844-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 05/29/2018] [Indexed: 12/19/2022]
|
20
|
Peng X, Zhang Y, Sun Y, Wang L, Song W, Li Q, Zhao R. Overexpressing modified human TRβ1 suppresses the proliferation of breast cancer MDA-MB-468 cells. Oncol Lett 2018; 16:785-792. [PMID: 29963146 PMCID: PMC6019938 DOI: 10.3892/ol.2018.8764] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 04/04/2018] [Indexed: 12/31/2022] Open
Abstract
A number of studies have indicated that thyroid hormone receptor β1 (TRβ1) functions as a tumor suppressor. TRs mediate transcriptional responses through a highly conserved DNA-binding domain (DBD). A novel rat TRβ isoform (rTRβΔ) was previously identified, in which a novel exon, N (108 bp), is located between exons 3 and 4 within the DBD; this exon represents the only difference between rTRβΔ and rTRβ1. In vitro, rTRβΔ exhibits a stronger tumor-suppressive capacity than rTRβ1, and further analysis revealed a high level of conservation between the rat and human DBD sequences. In the present study, an artificially modified human TRβ1 (m-hTRβ1) was constructed via the introduction of the 108-bp sequence into the corresponding position of the wild-type human TRβ1 (wt-hTRβ1) DBD. An electrophoretic mobility shift assay and transfection experiments confirmed that m-hTRβ1 is functional. Overexpression of m-hTRβ1 inhibits the proliferation of MDA-MB-468 cells in the presence of triiodothyronine by promoting apoptosis, which may be associated with the upregulation of Caspase-3 and Bak gene expression and the activation of the Caspase-3 protein. In addition, the pro-apoptotic effect of m-hTRβ1 was stronger, compared with wt-hTRβ1. These results indicated that m-hTRβ1 may act as a tumor suppressor in MDA-MB-468 cells. These data provided a novel insight into gene therapy for breast cancer.
Collapse
Affiliation(s)
- Xiaoxiang Peng
- Department of Laboratory Medicine, Weifang Medical University, Weifang, Shandong 261053, P.R. China.,Department of Clinical Laboratory, Key Discipline of Clinical Laboratory Medicine of Shandong, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Yangyang Zhang
- Department of Laboratory Medicine, Weifang Medical University, Weifang, Shandong 261053, P.R. China.,Department of Clinical Laboratory, Key Discipline of Clinical Laboratory Medicine of Shandong, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Yanli Sun
- Department of Laboratory Medicine, Weifang Medical University, Weifang, Shandong 261053, P.R. China.,Department of Clinical Laboratory, Key Discipline of Clinical Laboratory Medicine of Shandong, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Lujuan Wang
- Department of Laboratory Medicine, Weifang Medical University, Weifang, Shandong 261053, P.R. China.,Department of Clinical Laboratory, Key Discipline of Clinical Laboratory Medicine of Shandong, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Wei Song
- Department of Laboratory Medicine, Weifang Medical University, Weifang, Shandong 261053, P.R. China.,Department of Clinical Laboratory, Key Discipline of Clinical Laboratory Medicine of Shandong, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Qian Li
- Department of Laboratory Medicine, Weifang Medical University, Weifang, Shandong 261053, P.R. China.,Department of Clinical Laboratory, Key Discipline of Clinical Laboratory Medicine of Shandong, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Ronglan Zhao
- Department of Laboratory Medicine, Weifang Medical University, Weifang, Shandong 261053, P.R. China.,Department of Clinical Laboratory, Key Discipline of Clinical Laboratory Medicine of Shandong, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261053, P.R. China
| |
Collapse
|
21
|
Gates LA, Gu G, Chen Y, Rohira AD, Lei JT, Hamilton RA, Yu Y, Lonard DM, Wang J, Wang SP, Edwards DG, Lavere PF, Shao J, Yi P, Jain A, Jung SY, Malovannaya A, Li S, Shao J, Roeder RG, Ellis MJ, Qin J, Fuqua SAW, O'Malley BW, Foulds CE. Proteomic profiling identifies key coactivators utilized by mutant ERα proteins as potential new therapeutic targets. Oncogene 2018; 37:4581-4598. [PMID: 29748621 PMCID: PMC6095836 DOI: 10.1038/s41388-018-0284-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 04/02/2018] [Accepted: 04/05/2018] [Indexed: 12/05/2022]
Abstract
Approximately 75% of breast cancers are estrogen receptor alpha (ERα)-positive and are treatable with endocrine therapies, but often patients develop lethal resistant disease. Frequent mutations (10–40%) in the ligand-binding domain (LBD) codons in the gene encoding ERα (ESR1) have been identified, resulting in ligand-independent, constitutively active receptors. In addition, ESR1 chromosomal translocations can occur, resulting in fusion proteins that lack the LBD and are entirely unresponsive to all endocrine treatments. Thus, identifying coactivators that bind to these mutant ERα proteins may offer new therapeutic targets for endocrine-resistant cancer. To define coactivator candidate targets, a proteomics approach was performed profiling proteins recruited to the two most common ERα LBD mutants, Y537S and D538G, and an ESR1-YAP1 fusion protein. These mutants displayed enhanced coactivator interactions as compared to unliganded wild-type ERα. Inhibition of these coactivators decreased the ability of ESR1 mutants to activate transcription and promote breast cancer growth in vitro and in vivo. Thus, we have identified specific coactivators that may be useful as targets for endocrine-resistant breast cancers.
Collapse
Affiliation(s)
- Leah A Gates
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.,Laboratory of Chromatin Biology and Epigenetics, The Rockefeller University, New York, NY, 10065, USA
| | - Guowei Gu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.,Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yue Chen
- Employee of Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA, 70130, USA
| | - Aarti D Rohira
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jonathan T Lei
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA.,Interdepartmental Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ross A Hamilton
- 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
| | - David M Lonard
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jin Wang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.,Department of Pharmacology, Baylor College of Medicine, Houston, TX, 77030, USA.,Center for Drug Discovery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Shu-Ping Wang
- Laboratory of Biochemistry and Molecular Biology, The Rockefeller University, New York, NY, 10065, USA
| | - David G Edwards
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Philip F Lavere
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jiangyong Shao
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ping Yi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Antrix Jain
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Sung Yun Jung
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Anna Malovannaya
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.,Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Shunqiang Li
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Jieya Shao
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Robert G Roeder
- Laboratory of Biochemistry and Molecular Biology, The Rockefeller University, New York, NY, 10065, USA
| | - Matthew J Ellis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA.,Interdepartmental Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, 77030, USA.,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jun Qin
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.,Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Suzanne A W Fuqua
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA.,Dan L. Duncan Cancer Center, 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.,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Charles E Foulds
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA. .,Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
22
|
Peng X, Zhou Y, Sun Y, Song W, Meng X, Zhao C, Zhao R. Overexpression of modified human TRβ1 suppresses the growth of hepatocarcinoma SK-hep1 cells in vitro and in xenograft models. Mol Cell Biochem 2018; 449:207-218. [PMID: 29679278 PMCID: PMC6223806 DOI: 10.1007/s11010-018-3357-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 04/16/2018] [Indexed: 11/25/2022]
Abstract
Association studies suggest that TRβ1 functions as a tumor suppressor. Thyroid hormone receptors (TRs) mediate transcriptional responses through a highly conserved DNA-binding domain (DBD). We previously constructed an artificially modified human TRβ1 (m-TRβ1) via the introduction of a 108-bp exon sequence into the corresponding position of the wild-type human TRβ1 (TRβ1) DBD. Studies confirmed that m-TRβ1 was functional and could inhibit the proliferation of breast cancer MDA-MB-468 cells in vitro. To understand the role of m-TRβ1 in liver tumor development, we adopted a gain-of-function approach by stably expressing TRβ (m-TRβ1 and TRβ1) genes in a human hepatocarcinoma cell line, SK-hep1 (without endogenous TRβ), and then evaluated the effects of the expressed TRβ on cancer cell proliferation, migration, and tumor growth in cell-based studies and xenograft models. In the presence of 3,5,3-l-triiodothyronine (T3), the expression of TRβ in SK-hep1 cells inhibited cancer cell proliferation and impeded tumor cell migration through the up-regulation of 4-1BB, Caspase-3, and Bak gene expression; down-regulation of Bcl-2 gene expression; and activation of the Caspase-3 protein. TRβ expression in SK-hep1 led to less tumor growth in xenograft models. Additionally, the anti-tumor effect of m-TRβ1 was stronger than that of TRβ1. These data indicate that m-TRβ1 can act as a tumor suppressor in hepatocarcinoma and its role was significantly better than that of TRβ1.
Collapse
Affiliation(s)
- Xiaoxiang Peng
- Department of Laboratory Medicine, Weifang Medical University, Weifang, 261053, Shandong, China
- Key Discipline of Clinical Laboratory Medicine of Shandong Province, Affiliated Hospital of Weifang Medical University, Weifang, 261053, Shandong, China
| | - Yuntao Zhou
- Central Hospital of Zibo, Zibo, 255020, Shandong, China
| | - Yanli Sun
- Department of Laboratory Medicine, Weifang Medical University, Weifang, 261053, Shandong, China
- Key Discipline of Clinical Laboratory Medicine of Shandong Province, Affiliated Hospital of Weifang Medical University, Weifang, 261053, Shandong, China
| | - Wei Song
- Department of Laboratory Medicine, Weifang Medical University, Weifang, 261053, Shandong, China
- Key Discipline of Clinical Laboratory Medicine of Shandong Province, Affiliated Hospital of Weifang Medical University, Weifang, 261053, Shandong, China
| | - Xiangying Meng
- Department of Laboratory Medicine, Weifang Medical University, Weifang, 261053, Shandong, China
- Key Discipline of Clinical Laboratory Medicine of Shandong Province, Affiliated Hospital of Weifang Medical University, Weifang, 261053, Shandong, China
| | - Chunling Zhao
- Key Laboratory of Biological Medicine in Universities of Shandong Province, Weifang Medical University, Weifang, 261053, Shandong, China
| | - Ronglan Zhao
- Department of Laboratory Medicine, Weifang Medical University, Weifang, 261053, Shandong, China.
- Key Discipline of Clinical Laboratory Medicine of Shandong Province, Affiliated Hospital of Weifang Medical University, Weifang, 261053, Shandong, China.
| |
Collapse
|
23
|
GPCRs profiling and identification of GPR110 as a potential new target in HER2+ breast cancer. Breast Cancer Res Treat 2018; 170:279-292. [PMID: 29574636 DOI: 10.1007/s10549-018-4751-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 03/13/2018] [Indexed: 12/20/2022]
Abstract
PURPOSE G protein-coupled receptors (GPCRs) represent the largest family of druggable targets in human genome. Although several GPCRs can cross-talk with the human epidermal growth factor receptors (HERs), the expression and function of most GPCRs remain unknown in HER2+ breast cancer (BC). In this study, we aimed to evaluate gene expression of GPCRs in tumorigenic or anti-HER2 drug-resistant cells and to understand the potential role of candidate GPCRs in HER2+ BC. METHODS Gene expression of 352 GPCRs was profiled in Aldeflur+ tumorigenic versus Aldeflur- population and anti-HER2 therapy-resistant derivatives versus parental cells of HER2+ BT474 cells. The GPCR candidates were confirmed in 7 additional HER2+ BC cell line models and publicly available patient dataset. Anchorage-dependent and anchorage-independent cell growth, mammosphere formation, and migration/invasion were evaluated upon GPR110 knockdown by siRNA in BT474 and SKBR3 parental and lapatinib+ trastuzumab-resistant (LTR) cells. RESULTS Adhesion and class A GPCRs were overexpressed in Aldeflur+ and anti-HER2 therapy-resistant population of BT474 cells, respectively. GPR110 was the only GPCR overexpressed in Aldeflur+ and anti-HER2 therapy-resistant population in BT474, SKBR3, HCC1569, MDA-MB-361, AU565, and/or HCC202 cells and in HER2+ BC subtype in patient tumors. Using BT474 and SKBR3 parental and LTR cells, we found that GPR110 knockdown significantly reduced anchorage-dependent/independent cell growth as well as migration/invasion of parental and LTR cells and mammosphere formation in LTR derivatives and not in parental cells. CONCLUSION Our data suggest a potential role of GPR110 in tumorigenicity and in tumor cell dissemination in HER2+ BC.
Collapse
|
24
|
Little AG. Local Regulation of Thyroid Hormone Signaling. VITAMINS AND HORMONES 2018; 106:1-17. [DOI: 10.1016/bs.vh.2017.06.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
25
|
Miao Y, Zhang X, Xing R, Zhou W, Liu C. Relationship between the expression of TRβ1 and the molecular typing and clinicopathological features of breast cancer. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2017; 10:8875-8883. [PMID: 31966755 PMCID: PMC6965471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 07/13/2017] [Indexed: 06/10/2023]
Abstract
OBJECTIVE To study the relationship between the expression of TRβ1 and the molecular typing and clinicopathological features of breast cancer. METHODS The expression of TRβ1, ER, PR and HER-2 proteins in 208 cases of invasive breast cancer, 52 intraductal carcinoma and 22 normal breast tissue was detected by immunohistochemistry in order to analyze the relationship between the expression of TRβ1 protein and clinicopathological parameters of breast cancer. Western blot was performed to detect the effect of TRβ1 silencing on the expression of Notch signaling pathway proteins and Epithelial-mesenchymal transition (EMT)-related proteins in MCF-7 cells. RESULTS In the 208 cases of invasive breast cancer tissues, over expression of TRβ1 protein was found in 88 cases while low expression in 120 cases, and the immunohistochemical score was (3.9±3.1). TRβ1 protein was found over expressed in all the 52 cases of intraductal carcinoma and 22 cases of normal breast tissue, with the immunohistochemical score of (9.7±2.1) and 12.0, respectively, and there was no significant difference between the two groups (P>0.05) while both of them were significantly lower than the invasive breast cancer group (P<0.05). The expression of TRβ1 protein in the invasive breast cancer tissues was significantly correlated with lymph node metastasis (P=0.041), molecular typing (P=0.037) and histological grade (P<0.001) while it was negatively correlated with HER-2 expression (r=0.926; P<0.001) and irrelevant with age (P=1.024), ER expression (P=0.834), PR expression (P=0.351) or TNM staging (P=1.032). Compared to normal MCF-7 cells, the expression of Notch1, Dell 1, Jagged-1 and vimentin proteins increased by 1.44 times, 1.53 times, 1.50 times and 1.45 times respectively in the TRβ1 expression silenced MCF-7 cell. CONCLUSIONS The expression of TRβ1 protein in breast cancer tissues decreased with the increase of HER-2 expression and histological grade. The depletion of TRβ1 protein may activate the Notch signaling pathway and enhance the EMT ability of breast cancer cells thus promoting the cancer cell migration.
Collapse
Affiliation(s)
- Yu Miao
- Department of Pathology, Cangzhou Central Hospital of Hebei Province Cangzhou, China
| | - Xin Zhang
- Department of Pathology, Cangzhou Central Hospital of Hebei Province Cangzhou, China
| | - Rongge Xing
- Department of Pathology, Cangzhou Central Hospital of Hebei Province Cangzhou, China
| | - Weiwei Zhou
- Department of Pathology, Cangzhou Central Hospital of Hebei Province Cangzhou, China
| | - Chunrong Liu
- Department of Pathology, Cangzhou Central Hospital of Hebei Province Cangzhou, China
| |
Collapse
|
26
|
Charalampoudis P, Agrogiannis G, Kontzoglou K, Kouraklis G, Sotiropoulos GC. Thyroid hormone receptor alpha (TRa) tissue expression in ductal invasive breast cancer: A study combining quantitative immunohistochemistry with digital slide image analysis. Eur J Surg Oncol 2017; 43:1428-1432. [PMID: 28583788 DOI: 10.1016/j.ejso.2017.05.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 05/02/2017] [Accepted: 05/11/2017] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND In breast cancer, hormonal receptors hold promise for developing novel targeted therapies. The thyroid exerts its actions via the thyroid hormone receptors alpha and beta. The clinical significance of the expression of thyroid hormone receptors in breast cancer is unclear. MATERIAL AND METHODS We studied thyroid hormone receptor alpha (TRa) expression in 82 samples from 41 women with ductal invasive breast cancer and no thyroid disease. We performed quantitative immunohistochemistry with digital image analysis and correlated TRa expression with clinicopathological parameters. RESULTS TRa was expressed in both normal breast epithelium and breast cancer, but expression in breast cancer was significantly lower. TRa was expressed significantly less in larger and grade III tumors. Conversely, breast cancers with lymphovascular invasion showed increased TRa expression compared to cancers without lymphovascular invasion. TRa expression was not significantly different between node-positive and node-negative breast cancers, or among different hormonal profiles and intrinsic subtypes. DISCUSSION This is the first-in-human study to combine quantitative immunohistochemistry with image analysis to study TRa expression in women with ductal invasive breast cancer and no clinical or biochemical evidence of thyroid dysfunction. We confirm that TRa is expressed in both normal and malignant breast epithelium and suggest that TRa expression is downregulated during breast carcinogenesis. Larger and higher grade breast cancers demonstrate partial loss in TRa expression. Alterations in TRa expression take place even in the absence of clinical or biochemical thyroid disease. The underlying mechanism of these findings and their potential significance in survival and relapse mandate further research.
Collapse
Affiliation(s)
- P Charalampoudis
- Breast Unit, Second Propedeutic Department of Surgery, Laiko Hospital, Athens University School of Medicine, Athens, Greece; Breast Unit, Guy's and Saint Thomas' NHS Foundation Trust, London, United Kingdom; Division of Cancer Studies, King's College London, United Kingdom. petros.charalampoudis.@gmail.com
| | - G Agrogiannis
- First Department of Pathology, Athens University School of Medicine, Athens, Greece
| | - K Kontzoglou
- Breast Unit, Second Propedeutic Department of Surgery, Laiko Hospital, Athens University School of Medicine, Athens, Greece
| | - G Kouraklis
- Breast Unit, Second Propedeutic Department of Surgery, Laiko Hospital, Athens University School of Medicine, Athens, Greece
| | - G C Sotiropoulos
- Breast Unit, Second Propedeutic Department of Surgery, Laiko Hospital, Athens University School of Medicine, Athens, Greece
| |
Collapse
|
27
|
Gu G, Dustin D, Fuqua SA. Targeted therapy for breast cancer and molecular mechanisms of resistance to treatment. Curr Opin Pharmacol 2016; 31:97-103. [PMID: 27883943 DOI: 10.1016/j.coph.2016.11.005] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 09/23/2016] [Accepted: 11/07/2016] [Indexed: 01/05/2023]
Abstract
In recent years, clinical trials investigating new drugs and therapeutic combinations have led to promising advances in breast cancer therapy. Subtyping breast cancers into hormone receptor (HR) positive, epidermal growth factor receptor (HER2) positive, and triple negative breast cancer (TNBC) is currently the basis of diagnosing and treating this disease. In addition to endocrine and HER2-targeted therapies in their respective subtypes, evidence from recent preclinical studies have shown several targetable pathways that overcome resistance in the clinical setting. The mTOR inhibitor everolimus and the CDK4/6 inhibitor palbociclib have been approved in HR-positive metastatic breast cancer (MBC) due to improved disease-free survival (DFS). Adding pertuzumab to trastuzumab in combination with taxanes further improves DFS in HER2-positive breast cancer. Targeted therapy to the heterogeneous group of TNBC is needed in combination with chemotherapy. However, patient selection and predictive biomarker development remains a big challenge for targeted therapy development in TNBC.
Collapse
Affiliation(s)
- Guowei Gu
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Derek Dustin
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Suzanne Aw Fuqua
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L Duncan Cancer Center, One Baylor Plaza, Houston, TX 77030, USA.
| |
Collapse
|
28
|
Zhang JF, Liu J, Wang Y, Zhang B. Novel therapeutic strategies for patients with triple-negative breast cancer. Onco Targets Ther 2016; 9:6519-6528. [PMID: 27799799 PMCID: PMC5085278 DOI: 10.2147/ott.s105716] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Triple-negative breast cancer (TNBC) represents a very heterogeneous group of breast diseases. Currently, the backbone of therapy for TNBC is mainly chemotherapy as there are no effective specific targeted agents approved to treat TNBC. Despite initial responses to chemotherapy, resistance frequently and rapidly develops and metastatic TNBC has a poor prognosis. Therefore, new targeted strategies are, accordingly, urgently needed. This article discusses the recent developments in targeted agents explored for TNBC, aiming to offer novel therapeutic strategies that can potentially assist in designing personalized therapeutics in the future as well as provide the basis for further research in an attempt to target TNBC.
Collapse
Affiliation(s)
- Jun-Fei Zhang
- Affiliated Hospital of Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, People's Republic of China
| | - Jia Liu
- Affiliated Hospital of Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, People's Republic of China; Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, People's Republic of China
| | - Yu Wang
- Affiliated Hospital of Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, People's Republic of China; Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, People's Republic of China
| | - Bin Zhang
- Affiliated Hospital of Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, People's Republic of China; Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, People's Republic of China
| |
Collapse
|
29
|
Perra A, Plateroti M, Columbano A. T3/TRs axis in hepatocellular carcinoma: new concepts for an old pair. Endocr Relat Cancer 2016; 23:R353-69. [PMID: 27353037 DOI: 10.1530/erc-16-0152] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 06/27/2016] [Indexed: 02/06/2023]
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer-related death worldwide, and its burden is expected to further increase in the next years. Chronic inflammation, induced by multiple viruses or metabolic alterations, and epigenetic and genetic modifications, cooperate in cancer development via a combination of common and distinct aetiology-specific pathways. In spite of the advances of classical therapies, the prognosis of this neoplasm has not considerably improved over the past few years. The advent of targeted therapies and the approval of the systemic treatment of advanced HCC with the kinase inhibitor sorafenib have provided some hope for the future. However, the benefits obtained from this treatment are still disappointing, as it extends the median life expectancy of patients by only few months. It is thus mandatory to find alternative effective treatments. Although the role played by thyroid hormones (THs) and their nuclear receptors (TRs) in human cancer is still unclear, mounting evidence indicates that they behave as oncosuppressors in HCC. However, the molecular mechanisms by which they exert this effect and the consequence of their activation following ligand binding on HCC progression remain elusive. In this review, we re-evaluate the existing evidence of the role of TH/TRs in HCC development; we will also discuss how TR alterations could affect fundamental biological processes, such as hepatocyte proliferation and differentiation, and consequently HCC progression. Finally, we will discuss if and how TRs can be foreseen as therapeutic targets in HCC and whether selective TR modulation by TH analogues may hold promise for HCC treatment.
Collapse
Affiliation(s)
- Andrea Perra
- Department of Biomedical SciencesUniversity of Cagliari, Cagliari, Italy
| | - Michelina Plateroti
- Cancer Research Center of Lyon INSERM U1052CNRS UMR5286, Université de Lyon, Université Lyon 1, Centre Léon Bérard, Département de la Recherche, Lyon, France
| | - Amedeo Columbano
- Department of Biomedical SciencesUniversity of Cagliari, Cagliari, Italy
| |
Collapse
|
30
|
Novel targets for paclitaxel nano formulations: Hopes and hypes in triple negative breast cancer. Pharmacol Res 2016; 111:577-591. [PMID: 27461138 DOI: 10.1016/j.phrs.2016.07.023] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 07/18/2016] [Accepted: 07/19/2016] [Indexed: 12/30/2022]
Abstract
Triple negative breast cancer is defined as one of the utmost prevailing breast cancers worldwide, possessing an inadequate prognosis and treatment option limited to chemotherapy and radiotherapy, creating a challenge for researchers as far as developing a specific targeted therapy is concerned. The past research era has shown several promising outcomes for TNBC such as nano-formulations of the chemotherapeutic agents already used for the management of the malignant tumor. Taking a glance at paclitaxel nano formulations, it has been proven beneficial in several researches in the past decade; nevertheless its solubility is often a challenge to scientists in achieving success. We have henceforth discussed the basic heterogeneity of triple negative breast cancer along with the current management options as well as a brief outlook on pros and cons of paclitaxel, known as the most widely used chemotherapeutic agent for the treatment of the disease. We further analyzed the need of nanotechnology pertaining to the problems encountered with the current paclitaxel formulations available discussing the strategic progress in various nano-formulations till date taking into account the basic research strategies required in terms of solubility, permeability, physicochemical properties, active and passive targeting. A thorough review in recent advances in active targeting for TNBC was carried out whereby the various ligands which are at present finding its way into TNBC research such as hyaluronic acid, folic acid, transferrin, etc. were discussed. These ligands have specific receptor affinity to TNBC tumor cells hence can be beneficial for novel drug targeting approaches. Conversely, there are currently several novel strategies in the research pipeline whose targeting ligands have not yet been studied. Therefore, we reviewed upon the numerous novel receptor targets along with the respective nano-formulation aspects which have not yet been fully researched upon and could be exemplified as outstanding target strategies for TNBC which is currently an urgent requirement.
Collapse
|
31
|
Gelsomino L, Gu G, Rechoum Y, Beyer AR, Pejerrey SM, Tsimelzon A, Wang T, Huffman K, Ludlow A, Andò S, Fuqua SAW. ESR1 mutations affect anti-proliferative responses to tamoxifen through enhanced cross-talk with IGF signaling. Breast Cancer Res Treat 2016; 157:253-265. [PMID: 27178332 DOI: 10.1007/s10549-016-3829-5] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 05/05/2016] [Indexed: 01/06/2023]
Abstract
The purpose of this study was to address the role of ESR1 hormone-binding mutations in breast cancer. Soft agar anchorage-independent growth assay, Western blot, ERE reporter transactivation assay, proximity ligation assay (PLA), coimmunoprecipitation assay, silencing assay, digital droplet PCR (ddPCR), Kaplan-Meier analysis, and statistical analysis. It is now generally accepted that estrogen receptor (ESR1) mutations occur frequently in metastatic breast cancers; however, we do not yet know how to best treat these patients. We have modeled the three most frequent hormone-binding ESR1 (HBD-ESR1) mutations (Y537N, Y537S, and D538G) using stable lentiviral transduction in human breast cancer cell lines. Effects on growth were examined in response to hormonal and targeted agents, and mutation-specific changes were studied using microarray and Western blot analysis. We determined that the HBD-ESR1 mutations alter anti-proliferative effects to tamoxifen (Tam), due to cell-intrinsic changes in activation of the insulin-like growth factor receptor (IGF1R) signaling pathway and levels of PIK3R1/PIK3R3. The selective estrogen receptor degrader, fulvestrant, significantly reduced the anchorage-independent growth of ESR1 mutant-expressing cells, while combination treatments with the mTOR inhibitor everolimus, or an inhibitor blocking IGF1R, and the insulin receptor significantly enhanced anti-proliferative responses. Using digital drop (dd) PCR, we identified mutations at high frequencies ranging from 12 % for Y537N, 5 % for Y537S, and 2 % for D538G in archived primary breast tumors from women treated with adjuvant mono-tamoxifen therapy. The HBD-ESR1 mutations were not associated with recurrence-free or overall survival in response in this patient cohort and suggest that knowledge of other cell-intrinsic factors in combination with ESR1 mutation status will be needed determine anti-proliferative responses to Tam.
Collapse
Affiliation(s)
- Luca Gelsomino
- Department of Pharmacy, Health, and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | - Guowei Gu
- Lester & Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Yassine Rechoum
- Lester & Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Amanda R Beyer
- Lester & Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Sasha M Pejerrey
- Lester & Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Anna Tsimelzon
- Lester & Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Tao Wang
- Lester & Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Kenneth Huffman
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Andrew Ludlow
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Sebastiano Andò
- Department of Pharmacy, Health, and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | - Suzanne A W Fuqua
- Lester & Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
- Dan L Duncan Cancer Center, Houston, TX, USA.
| |
Collapse
|