1
|
de Klein B, Eickhoff N, Zwart W. The emerging regulatory interface between DNA repair and steroid hormone receptors in cancer. Trends Mol Med 2025:S1471-4914(25)00006-1. [PMID: 39934021 DOI: 10.1016/j.molmed.2025.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 02/13/2025]
Abstract
Human cells potentiate highly diverse functions through tight transcriptional regulation and maintenance of genome integrity. While the DNA damage response (DDR) safeguards the genome, ligand-activated transcription factors, such as steroid hormone receptors (SHRs), provide complex transcriptional outputs. Interestingly, an increasing body of evidence reveals a direct biological and functional interplay between DDR factors and SHR cascades in cancer. SHRs can directly affect DDR gene expression, but DDR factors in turn act as transcriptional coregulators, enabling oncogenic SHR-mediated signaling, which has the potential for novel therapeutic interventions. With a focus on breast and prostate cancer, we describe in this review recent developments in, and insights into, the complex interplay between SHR signaling and the DDR, highlighting opportunities for future clinical interventions.
Collapse
Affiliation(s)
- Bim de Klein
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Nils Eickhoff
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Wilbert Zwart
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands; Department of Biomedical Engineering, Eindhoven University of Technology, 5600, MB, Eindhoven, The Netherlands.
| |
Collapse
|
2
|
Zhou Z, Sicairos B, Zhou J, Du Y. Proteomic Analysis Reveals Major Proteins and Pathways That Mediate the Effect of 17-β-Estradiol in Cell Division and Apoptosis in Breast Cancer MCF7 Cells. J Proteome Res 2024; 23:4835-4848. [PMID: 39392593 PMCID: PMC11536429 DOI: 10.1021/acs.jproteome.4c00102] [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: 02/13/2024] [Revised: 09/28/2024] [Accepted: 10/01/2024] [Indexed: 10/12/2024]
Abstract
Despite extensive research, the genes/proteins and pathways responsible for the physiological effects of estrogen remain elusive. In this study, we determined the effect of estrogen on global protein expression in breast cancer MCF7 cells using a proteomic method. The expression of 77 cytosolic, 74 nuclear, and 81 membrane/organelle proteins was significantly altered by 17-β-estradiol (E2). Protein enrichment analyses suggest that E2 may stimulate cell division primarily by promoting the G1 to S phase transition and advancing the G2/M checkpoint. The effect of E2 on cell survival was complex, as it could simultaneously enhance and inhibit apoptosis. Bioinformatics analysis suggests that E2 may enhance apoptosis by promoting the accumulation of the pore-forming protein Bax in the mitochondria and inhibit apoptosis by activating the PI3K/AKT/mTOR signaling pathway. We verified the activation of the PI3K signaling and the accumulation of Bax in the membrane/organelle fraction in E2-treated cells using immunoblotting. Treatment of MCF7 cells with E2 and the PI3K inhibitor Ly294002 significantly enhanced apoptosis compared to those treated with E2 alone, suggesting that combining estrogen with a PI3K inhibitor could be a promising strategy for treating ERα-positive breast cancer. Interestingly, many of the E2-upregulated proteins contained the HEAT, KH, and RRM domains.
Collapse
Affiliation(s)
| | | | | | - Yuchun Du
- Department of Biological
Sciences, University of Arkansas, Fayetteville, Arkansas 72701, United States
| |
Collapse
|
3
|
Barjesteh F, Heidari-Kalvani N, Alipourfard I, Najafi M, Bahreini E. Testosterone, β-estradiol, and hepatocellular carcinoma: stimulation or inhibition? A comparative effect analysis on cell cycle, apoptosis, and Wnt signaling of HepG2 cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:6121-6133. [PMID: 38421409 DOI: 10.1007/s00210-024-03019-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 02/20/2024] [Indexed: 03/02/2024]
Abstract
Unlike breast and prostate cancers, which are specifically affected by estrogens or androgens, hepatocellular carcinoma has been reported to be influenced by both sex hormones. Given the coincidental differences of hepatocellular carcinoma in men and women, we investigated the effects of β-estradiol and testosterone on the cell cycle, apoptosis, and Wnt signaling in a model of hepatocellular carcinoma to understand the sex hormone-related etiology. To determine the effective concentration of both hormones, an MTT assay was performed. The effects of β-estradiol and testosterone on cell proliferation and death were evaluated by specific staining and flow cytometry. In addition, gene expression levels of estimated factors involved in GPC3-Wnt survival signaling were analyzed using quantitative real-time polymerase chain reaction. Both hormones inhibited hepatic cell proliferation through arresting the cell cycle at S/G2 and increased the apoptosis rate in HepG2 cells. Both hormones dose-dependently decreased GPC3, Wnt, and DVL expression levels as activators of the Wnt-signaling pathway. In the case of Wnt-signaling inhibitors, the effects of both hormones on WIF were negligible, but they increased DKK1 levels in a dose-dependent manner. In each of the effects mentioned above, β-estradiol was notably more potent than testosterone. In contrast to the primary hypothesis of the project, in which testosterone was considered a stimulating carcinogenic factor in HCC pathogenesis, testosterone inhibited the occurrence of HCC similarly to β-estradiol. However, this inhibitory effect was weaker than that of β-estradiol and requires further study.
Collapse
Affiliation(s)
- Fereshteh Barjesteh
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, 1449614525, Iran
| | - Nafiseh Heidari-Kalvani
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, 1449614525, Iran
| | - Iraj Alipourfard
- Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | - Mohammad Najafi
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, 1449614525, Iran
| | - Elham Bahreini
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, 1449614525, Iran.
| |
Collapse
|
4
|
Pacheco JHL, Elizondo G. Interplay between Estrogen, Kynurenine, and AHR Pathways: An immunosuppressive axis with therapeutic potential for breast cancer treatment. Biochem Pharmacol 2023; 217:115804. [PMID: 37716620 DOI: 10.1016/j.bcp.2023.115804] [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: 06/09/2023] [Revised: 09/11/2023] [Accepted: 09/11/2023] [Indexed: 09/18/2023]
Abstract
Breast cancer is one of the most common malignancies among women worldwide. Estrogen exposure via endogenous and exogenous sources during a lifetime, together with environmental exposure to estrogenic compounds, represent the most significant risk factor for breast cancer development. As breast tumors establish, multiple pathways are deregulated. Among them is the aryl hydrocarbon receptor (AHR) signaling pathway. AHR, a ligand-activated transcription factor associated with the metabolism of polycyclic aromatic hydrocarbons and estrogens, is overexpressed in breast cancer. Furthermore, AHR and estrogen receptor (ER) cross-talk pathways have been observed. Additionally, the Tryptophan (Trp) catabolizing enzymes indolamine-2,3-dioxygenase (IDO) and tryptophan-2,3-dioxygenase (TDO) are overexpressed in breast cancer. IDO/TDO catalyzes the formation of Kynurenine (KYN) and other tryptophan-derived metabolites, which are ligands of AHR. Once KYN activates AHR, it stimulates the expression of the IDO enzyme, increases the level of KYN, and activates non-canonical pathways to control inflammation and immunosuppression in breast tumors. The interplay between E2, AHR, and IDO/TDO/KYN pathways and their impact on the immune system represents an immunosuppressive axis on breast cancer. The potential modulation of the immunosuppressive E2-AHR-IDO/TDO/KYN axis has aroused great expectations in oncotherapy. The present article will review the mechanisms implicated in generating the immunosuppressive axis E2-AHR-IDO/TDO/KYN in breast cancer and the current state of knowledge as a potential therapeutic target.
Collapse
Affiliation(s)
| | - Guillermo Elizondo
- Departamento de Biología Celular, CINVESTAV-IPN, Av. IPN 2508, C.P. 07360 Ciudad de México, México.
| |
Collapse
|
5
|
Nahmias-Blank D, Maimon O, Meirovitz A, Sheva K, Peretz-Yablonski T, Elkin M. Excess body weight and postmenopausal breast cancer: Emerging molecular mechanisms and perspectives. Semin Cancer Biol 2023; 96:26-35. [PMID: 37739109 DOI: 10.1016/j.semcancer.2023.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/24/2023]
Abstract
Postmenopausal, obese women have a significantly higher risk of developing estrogen receptor-positive (ER+) breast tumors, that are resistant to therapies and are associated with higher recurrence and death rates. The global prevalence of overweight/obese women has reached alarming proportions and with postmenopausal ER+ breast carcinoma (BC) having the highest incidence among the three obesity-related cancers in females (i.e., breast, endometrial and ovarian), this is of significant concern. Elucidation of the precise molecular mechanisms underlying the pro-cancerous action of obesity in ER+BC is therefore critical for disease prevention and novel treatment initiatives. Interestingly, accumulating data has shown opposing relationships between obesity and cancer in either pre- or post-menopausal women. Excess body weight is associated with an increased risk of breast cancer in postmenopausal women and a decreased risk in pre-menopausal women. Moreover, excess adiposity during early life appears to be protective against postmenopausal breast cancer, including both ER+ and ER negative BC subtypes. Overall, estrogen-dependent mechanisms have been implicated as the main driving force in obesity-related breast tumorigenesis. In the present review we discuss the epidemiologic and mechanistic aspects of association between obesity and breast tumors after menopause, mainly in the context of hormone dependency. Molecular and cellular events underlying this association present as potential avenues for both therapeutic intervention as well as the prevention of BC-promoting processes linked to excess adiposity, which is proving to be vital in an increasingly obese global population.
Collapse
Affiliation(s)
- Daniela Nahmias-Blank
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Ofra Maimon
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Amichay Meirovitz
- Legacy Heritage Oncology Center and Dr. Larry Norton Institute, Soroka University Medical Center, Be'er Sheva 84101, Israel
| | - Kim Sheva
- Legacy Heritage Oncology Center and Dr. Larry Norton Institute, Soroka University Medical Center, Be'er Sheva 84101, Israel
| | - Tamar Peretz-Yablonski
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel; Hebrew University Medical School, Jerusalem 91120, Israel
| | - Michael Elkin
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel; Hebrew University Medical School, Jerusalem 91120, Israel.
| |
Collapse
|
6
|
Cha J, Aguayo-Mazzucato C, Thompson PJ. Pancreatic β-cell senescence in diabetes: mechanisms, markers and therapies. Front Endocrinol (Lausanne) 2023; 14:1212716. [PMID: 37720527 PMCID: PMC10501801 DOI: 10.3389/fendo.2023.1212716] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 08/15/2023] [Indexed: 09/19/2023] Open
Abstract
Cellular senescence is a response to a wide variety of stressors, including DNA damage, oncogene activation and physiologic aging, and pathologically accelerated senescence contributes to human disease, including diabetes mellitus. Indeed, recent work in this field has demonstrated a role for pancreatic β-cell senescence in the pathogenesis of Type 1 Diabetes, Type 2 Diabetes and monogenic diabetes. Small molecule or genetic targeting of senescent β-cells has shown promise as a novel therapeutic approach for preventing and treating diabetes. Despite these advances, major questions remain around the molecular mechanisms driving senescence in the β-cell, identification of molecular markers that distinguish senescent from non-senescent β-cell subpopulations, and translation of proof-of-concept therapies into novel treatments for diabetes in humans. Here, we summarize the current state of the field of β-cell senescence, highlighting insights from mouse models as well as studies on human islets and β-cells. We identify markers that have been used to detect β-cell senescence to unify future research efforts in this field. We discuss emerging concepts of the natural history of senescence in β-cells, heterogeneity of senescent β-cells subpopulations, role of sex differences in senescent responses, and the consequences of senescence on integrated islet function and microenvironment. As a young and developing field, there remain many open research questions which need to be addressed to move senescence-targeted approaches towards clinical investigation.
Collapse
Affiliation(s)
- Jeeyeon Cha
- Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN, United States
| | | | - Peter J. Thompson
- Diabetes Research Envisioned and Accomplished in Manitoba Theme, Children’s Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
- Department of Physiology & Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
7
|
Maenosono R. Sex difference and immunosenescence affect transplantation outcomes. FRONTIERS IN TRANSPLANTATION 2023; 2:1235740. [PMID: 38993850 PMCID: PMC11235384 DOI: 10.3389/frtra.2023.1235740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 08/09/2023] [Indexed: 07/13/2024]
Abstract
Kidney transplantation is a well-established alternative to renal replacement therapy. Although the number of patients with end-stage renal disease (ESRD) is increasing, the availability of kidney for transplantation is still insufficient to meet the needs. As age increases, the prevalence of ESRD increases; thus, the population of aged donors and recipients occupies large proportion. Accumulated senescent cells secrete pro-inflammatory factors and induce senescence. Additionally, it is gradually becoming clear that biological sex differences can influence aging and cause differences in senescence. Here, we review whether age-related sex differences affect organ transplant outcomes and what should be done in the future.
Collapse
Affiliation(s)
- Ryoichi Maenosono
- Department of Urology, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| |
Collapse
|
8
|
Ansari A, Walton SL, Denton KM. Sex- and age-related differences in renal and cardiac injury and senescence in stroke-prone spontaneously hypertensive rats. Biol Sex Differ 2023; 14:33. [PMID: 37217968 DOI: 10.1186/s13293-023-00519-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/05/2023] [Indexed: 05/24/2023] Open
Abstract
BACKGROUND Sex differences play a critical role in the incidence and severity of cardiovascular diseases, whereby men are at a higher risk of developing cardiovascular disease compared to age-matched premenopausal women. Marked sex differences at the cellular and tissue level may contribute to susceptibility to cardiovascular disease and end-organ damage. In this study, we have performed an in-depth histological analysis of sex differences in hypertensive cardiac and renal injury in middle-aged stroke-prone spontaneously hypertensive rats (SHRSPs) to determine the interaction between age, sex and cell senescence. METHODS Kidneys, hearts and urine samples were collected from 6.5- and 8-month-old (Mo) male and female SHRSPs. Urine samples were assayed for albumin and creatinine content. Kidneys and hearts were screened for a suite of cellular senescence markers (senescence-associated β-galactosidase, p16INK4a, p21, γH2AX). Renal and cardiac fibrosis was quantified using Masson's trichrome staining, and glomerular hypertrophy and sclerosis were quantified using Periodic acid-Schiff staining. RESULTS Marked renal and cardiac fibrosis, concomitant with albuminuria, were evident in all SHRSPs. These sequelae were differentially affected by age, sex and organ. That is, the level of fibrosis was greater in the kidney than the heart, males had greater levels of fibrosis than females in both the heart and kidney, and even a 6-week increase in age resulted in greater levels of kidney fibrosis in males. The differences in kidney fibrosis were reflected by elevated levels of cellular senescence in the kidney in males but not females. Senescent cell burden was significantly less in cardiac tissue compared to renal tissue and was not affected by age or sex. CONCLUSIONS Our study demonstrates a clear sex pattern in age-related progression of renal and cardiac fibrosis and cellular senescence in SHRSP rats. A 6-week time frame was associated with increased indices of cardiac and renal fibrosis and cellular senescence in male SHRSPs. Female SHRSP rats were protected from renal and cardiac damage compared to age-matched males. Thus, the SHRSP is an ideal model to investigate the effects of sex and aging on organ injury over a short timeframe.
Collapse
Affiliation(s)
- Aneesa Ansari
- Department of Physiology, Monash University, Melbourne, VIC, Australia
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Sarah L Walton
- Department of Physiology, Monash University, Melbourne, VIC, Australia
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Kate M Denton
- Department of Physiology, Monash University, Melbourne, VIC, Australia.
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
9
|
Rangsrikitphoti P, Marquez-Garban DC, Pietras RJ, McGowan E, Boonyaratanakornkit V. Sex steroid hormones and DNA repair regulation: Implications on cancer treatment responses. J Steroid Biochem Mol Biol 2023; 227:106230. [PMID: 36450315 DOI: 10.1016/j.jsbmb.2022.106230] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022]
Abstract
The role of sex steroid hormones (SSHs) has been shown to modulate cancer cytotoxic treatment sensitivity. Dysregulation of DNA repair associated with genomic instability, abnormal cell survival and not only promotes cancer progression but also resistance to cancer treatment. The three major SSHs, androgen, estrogen, and progesterone, have been shown to interact with several essential DNA repair components. The presence of androgens directly regulates key molecules in DNA double-strand break (DSB) repair. Estrogen can promote cell proliferation and DNA repair, allowing cancer cells to tolerate chemotherapy and radiotherapy. Information on the role of progesterone in DNA repair is limited: progesterone interaction with some DNA repair components has been identified, but the biological significance is still unknown. Here, we review the roles of how each SSH affects DNA repair regulation and modulates response to genotoxic therapies and discuss future research that can be beneficial when combining SSHs with cancer therapy. We also provide preliminary analysis from publicly available databases defining the link between progesterone/PR and DDRs & DNA repair regulation that plausibly contribute to chemotherapy response and breast cancer patient survival.
Collapse
Affiliation(s)
- Pattarasiri Rangsrikitphoti
- Graduate Program in Clinical Biochemistry and Molecular Medicine and Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Diana C Marquez-Garban
- UCLA Jonsson Comprehensive Cancer and Department of Medicine, Division of Hematology-Oncology, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Richard J Pietras
- UCLA Jonsson Comprehensive Cancer and Department of Medicine, Division of Hematology-Oncology, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Eileen McGowan
- School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
| | - Viroj Boonyaratanakornkit
- Graduate Program in Clinical Biochemistry and Molecular Medicine and Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand; Age-related Inflammation and Degeneration Research Unit, Chulalongkorn University, Bangkok 10330, Thailand.
| |
Collapse
|
10
|
The Interplay between the Cellular Response to DNA Double-Strand Breaks and Estrogen. Cells 2022; 11:cells11193097. [PMID: 36231059 PMCID: PMC9563627 DOI: 10.3390/cells11193097] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 09/28/2022] [Accepted: 09/29/2022] [Indexed: 11/17/2022] Open
Abstract
Cancer development is often connected to impaired DNA repair and DNA damage signaling pathways. The presence of DNA damage in cells activates DNA damage response, which is a complex cellular signaling network that includes DNA repair, activation of the cell cycle checkpoints, cellular senescence, and apoptosis. DNA double-strand breaks (DSBs) are toxic lesions that are mainly repaired by the non-homologous end joining and homologous recombination repair (HRR) pathways. Estrogen-dependent cancers, like breast and ovarian cancers, are frequently associated with mutations in genes that play a role in HRR. The female sex hormone estrogen binds and activates the estrogen receptors (ERs), ERα, ERβ and G-protein-coupled ER 1 (GPER1). ERα drives proliferation, while ERβ inhibits cell growth. Estrogen regulates the transcription, stability and activity of numerus DDR factors and DDR factors in turn modulate ERα expression, stability and transcriptional activity. Additionally, estrogen stimulates DSB formation in cells as part of its metabolism and proliferative effect. In this review, we will present an overview on the crosstalk between estrogen and the cellular response to DSBs. We will discuss how estrogen regulates DSB signaling and repair, and how DDR factors modulate the expression, stability and activity of estrogen. We will also discuss how the regulation of HRR genes by estrogen promotes the development of estrogen-dependent cancers.
Collapse
|
11
|
Evidence of Sex Differences in Cellular Senescence. Neurobiol Aging 2022; 120:88-104. [PMID: 36166919 DOI: 10.1016/j.neurobiolaging.2022.08.014] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/30/2022] [Accepted: 08/30/2022] [Indexed: 11/20/2022]
|
12
|
Zhao Y, Wang X, Liu Y, Wang HY, Xiang J. The effects of estrogen on targeted cancer therapy drugs. Pharmacol Res 2022; 177:106131. [DOI: 10.1016/j.phrs.2022.106131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/29/2022] [Accepted: 02/10/2022] [Indexed: 10/19/2022]
|
13
|
Thiebaut C, Vlaeminck-Guillem V, Trédan O, Poulard C, Le Romancer M. Non-genomic signaling of steroid receptors in cancer. Mol Cell Endocrinol 2021; 538:111453. [PMID: 34520815 DOI: 10.1016/j.mce.2021.111453] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 09/03/2021] [Accepted: 09/05/2021] [Indexed: 12/21/2022]
Abstract
Steroid receptors (SRs) are members of the nuclear receptor family, which are ligand-activated transcription factors. SRs regulate many physiological functions including development and reproduction, though they can also be involved in several pathologies, especially cancer. Highly controlled cellular responses to steroids involve transcriptional regulation (genomic activity) combined with direct activation of signaling cascades (non-genomic activity). Non-genomic signaling has been extensively studied in cancer, mainly in breast cancer for ER and PR, and prostate cancer for AR. Even though most of the studies have been conducted in cells, some of them have been confirmed in vivo, highlighting the relevance of this pathway in cancer. This review provides an overview of the current and emerging knowledge on non-genomic signaling with a focus on breast and prostate cancers and its clinical relevance. A thorough understanding of ER, PR, AR and GR non-genomic pathways may open new perspectives for the development of therapeutic strategies.
Collapse
Affiliation(s)
- Charlène Thiebaut
- Université de Lyon, F-69000, Lyon, France; Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000, Lyon, France; CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000, Lyon, France
| | - Virginie Vlaeminck-Guillem
- Université de Lyon, F-69000, Lyon, France; Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000, Lyon, France; CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000, Lyon, France; Service de Biochimie Biologie Moléculaire Sud, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, F-69495, Pierre-Bénite, France
| | - Olivier Trédan
- Université de Lyon, F-69000, Lyon, France; Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000, Lyon, France; CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000, Lyon, France; Medical Oncology Department, Centre Léon Bérard, F-69000, Lyon, France
| | - Coralie Poulard
- Université de Lyon, F-69000, Lyon, France; Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000, Lyon, France; CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000, Lyon, France
| | - Muriel Le Romancer
- Université de Lyon, F-69000, Lyon, France; Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000, Lyon, France; CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000, Lyon, France.
| |
Collapse
|
14
|
Abstract
Significant sex differences exist across cellular, tissue organization, and body system scales to serve the distinct sex-specific functions required for reproduction. They are present in all animals that reproduce sexually and have widespread impacts on normal development, aging, and disease. Observed from the moment of fertilization, sex differences are patterned by sexual differentiation, a lifelong process that involves mechanisms related to sex chromosome complement and the epigenetic and acute activational effects of sex hormones. In this mini-review, we examine evidence for sex differences in cellular responses to DNA damage, their underlying mechanisms, and how they might relate to sex differences in cancer incidence and response to DNA-damaging treatments.
Collapse
Affiliation(s)
- Lauren Broestl
- Department of Pediatrics, Washington University School of Medicine, St Louis, MO, USA
| | - Joshua B Rubin
- Department of Pediatrics, Washington University School of Medicine, St Louis, MO, USA
- Department of Neuroscience, Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
15
|
Bhardwaj P, Brown KA. Obese Adipose Tissue as a Driver of Breast Cancer Growth and Development: Update and Emerging Evidence. Front Oncol 2021; 11:638918. [PMID: 33859943 PMCID: PMC8042134 DOI: 10.3389/fonc.2021.638918] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 03/12/2021] [Indexed: 12/24/2022] Open
Abstract
Obesity is an established risk factor for breast cancer growth and progression. A number of advances have been made in recent years revealing new insights into this link. Early events in breast cancer development involve the neoplastic transformation of breast epithelial cells to cancer cells. In obesity, breast adipose tissue undergoes significant hormonal and inflammatory changes that create a mitogenic microenvironment. Many factors that are produced in obesity have also been shown to promote tumorigenesis. Given that breast epithelial cells are surrounded by adipose tissue, the crosstalk between the adipose compartment and breast epithelial cells is hypothesized to be a significant player in the initiation and progression of breast cancer in individuals with excess adiposity. The present review examines this crosstalk with a focus on obese breast adipose-derived estrogen, inflammatory mediators and adipokines, and how they are mechanistically linked to breast cancer risk and growth through stimulation of oxidative stress, DNA damage, and pro-oncogenic transcriptional programs. Pharmacological and lifestyle strategies targeting these factors and their downstream effects are evaluated for feasibility and efficacy in decreasing the risk of obesity-induced breast epithelial cell transformation and consequently, breast cancer development.
Collapse
Affiliation(s)
- Priya Bhardwaj
- Department of Medicine, Weill Cornell Medicine, New York, NY, United States
- Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, United States
| | - Kristy A. Brown
- Department of Medicine, Weill Cornell Medicine, New York, NY, United States
- Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, United States
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
16
|
Pescatori S, Berardinelli F, Albanesi J, Ascenzi P, Marino M, Antoccia A, di Masi A, Acconcia F. A Tale of Ice and Fire: The Dual Role for 17β-Estradiol in Balancing DNA Damage and Genome Integrity. Cancers (Basel) 2021; 13:1583. [PMID: 33808099 PMCID: PMC8036963 DOI: 10.3390/cancers13071583] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/24/2021] [Accepted: 03/26/2021] [Indexed: 12/21/2022] Open
Abstract
17β-estradiol (E2) regulates human physiology both in females and in males. At the same time, E2 acts as a genotoxic substance as it could induce DNA damages, causing the initiation of cellular transformation. Indeed, increased E2 plasma levels are a risk factor for the development of several types of cancers including breast cancer. This paradoxical identity of E2 undermines the foundations of the physiological definition of "hormone" as E2 works both as a homeostatic regulator of body functions and as a genotoxic compound. Here, (i) the molecular circuitries underlying this double face of E2 are reviewed, and (ii) a possible framework to reconcile the intrinsic discrepancies of the E2 function is reported. Indeed, E2 is a regulator of the DNA damage response, which this hormone exploits to calibrate its genotoxicity with its physiological effects. Accordingly, the genes required to maintain genome integrity belong to the E2-controlled cellular signaling network and are essential for the appearance of the E2-induced cellular effects. This concept requires an "upgrade" to the vision of E2 as a "genotoxic hormone", which balances physiological and detrimental pathways to guarantee human body homeostasis. Deregulation of this equilibrium between cellular pathways would determine the E2 pathological effects.
Collapse
Affiliation(s)
- Sara Pescatori
- Department of Sciences, Section Biomedical Sciences, and Technology, University Roma Tre, Viale Guglielmo Marconi, 446, I-00146 Rome, Italy; (S.P.); (F.B.); (J.A.); (P.A.); (M.M.)
| | - Francesco Berardinelli
- Department of Sciences, Section Biomedical Sciences, and Technology, University Roma Tre, Viale Guglielmo Marconi, 446, I-00146 Rome, Italy; (S.P.); (F.B.); (J.A.); (P.A.); (M.M.)
- Neurodevelopment, Neurogenetics and Molecular Neurobiology Unit, IRCCS Santa Lucia Foundation, 00143 Rome, Italy
| | - Jacopo Albanesi
- Department of Sciences, Section Biomedical Sciences, and Technology, University Roma Tre, Viale Guglielmo Marconi, 446, I-00146 Rome, Italy; (S.P.); (F.B.); (J.A.); (P.A.); (M.M.)
| | - Paolo Ascenzi
- Department of Sciences, Section Biomedical Sciences, and Technology, University Roma Tre, Viale Guglielmo Marconi, 446, I-00146 Rome, Italy; (S.P.); (F.B.); (J.A.); (P.A.); (M.M.)
- Neuroendocrinology, Metabolism and Neuropharmacology Unit, IRCCS Santa Lucia Foundation, 00143 Rome, Italy
| | - Maria Marino
- Department of Sciences, Section Biomedical Sciences, and Technology, University Roma Tre, Viale Guglielmo Marconi, 446, I-00146 Rome, Italy; (S.P.); (F.B.); (J.A.); (P.A.); (M.M.)
- Neuroendocrinology, Metabolism and Neuropharmacology Unit, IRCCS Santa Lucia Foundation, 00143 Rome, Italy
| | - Antonio Antoccia
- Department of Sciences, Section Biomedical Sciences, and Technology, University Roma Tre, Viale Guglielmo Marconi, 446, I-00146 Rome, Italy; (S.P.); (F.B.); (J.A.); (P.A.); (M.M.)
| | - Alessandra di Masi
- Department of Sciences, Section Biomedical Sciences, and Technology, University Roma Tre, Viale Guglielmo Marconi, 446, I-00146 Rome, Italy; (S.P.); (F.B.); (J.A.); (P.A.); (M.M.)
| | - Filippo Acconcia
- Department of Sciences, Section Biomedical Sciences, and Technology, University Roma Tre, Viale Guglielmo Marconi, 446, I-00146 Rome, Italy; (S.P.); (F.B.); (J.A.); (P.A.); (M.M.)
| |
Collapse
|
17
|
Jiménez-Salazar JE, Damian-Ferrara R, Arteaga M, Batina N, Damián-Matsumura P. Non-Genomic Actions of Estrogens on the DNA Repair Pathways Are Associated With Chemotherapy Resistance in Breast Cancer. Front Oncol 2021; 11:631007. [PMID: 33869016 PMCID: PMC8044931 DOI: 10.3389/fonc.2021.631007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/27/2021] [Indexed: 12/12/2022] Open
Abstract
Estrogens have been implicated in the etiology of breast cancer for a long time. It has been stated that long-term exposure to estrogens is associated with a higher incidence of breast cancer, since estradiol (E2) stimulates breast cell growth; however, its effect on DNA damage/repair is only starting to be investigated. Recent studies have documented that estrogens are able to modify the DNA damage response (DDR) and DNA repair mechanisms. On the other hand, it has been proposed that DDR machinery can be altered by estrogen signaling pathways, that can be related to cancer progression and chemoresistance. We have demonstrated that E2 promotes c-Src activation and breast cancer cell motility, through a non-genomic pathway. This review discusses scientific evidence supporting this non-genomic mechanism where estrogen modifies the DNA repair pathways, and its relationship to potential causes of chemoresistance.
Collapse
Affiliation(s)
- Javier E Jiménez-Salazar
- Department of Biology of Reproduction, Division of Biological Sciences and Health (DCBS), Autonomous Metropolitan University (UAM), Mexico City, Mexico.,School of Medicine, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| | - Rebeca Damian-Ferrara
- Monterrey Institute of Technology and Higher Education (ITESM), School of Engineering and Sciences, Monterrey, Mexico
| | - Marcela Arteaga
- Department of Biology of Reproduction, Division of Biological Sciences and Health (DCBS), Autonomous Metropolitan University (UAM), Mexico City, Mexico
| | - Nikola Batina
- Nanotechnology and Molecular Engineering Laboratory, Department of Chemistry, Division of Basic Science and Engineering (DCBI), Autonomous Metropolitan University (UAM), Mexico City, Mexico
| | - Pablo Damián-Matsumura
- Department of Biology of Reproduction, Division of Biological Sciences and Health (DCBS), Autonomous Metropolitan University (UAM), Mexico City, Mexico
| |
Collapse
|
18
|
Ganguly S, Naik D, Muskara A, Mian OY. The Nexus of Endocrine Signaling and Cancer: How Steroid Hormones Influence Genomic Stability. Endocrinology 2021; 162:bqaa177. [PMID: 33260197 PMCID: PMC7707372 DOI: 10.1210/endocr/bqaa177] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Indexed: 12/13/2022]
Abstract
Endocrine-driven malignancies, including breast and prostate cancer, are among the most common human cancers. The relationship between sex steroid hormones (eg, androgen, estrogen, and progesterone), their cognate receptors, and genomic stability lie at the center of endocrine-driven cancer development, progression, and therapeutic resistance. A variety of direct and indirect mechanisms have been described that link steroid hormone signaling to the loss of genomic integrity that drives early carcinogenesis. These effects are often enriched within endocrine receptor cistromes, accounting for the high proportion of mutations and rearrangements in the region of hormone response elements. In other cases, the effects are generalized and rely on a complex array of genetic, epigenetic, and metabolic interactions. Both androgen and estrogen receptors directly modulate the DNA damage response by trans-activating DNA damage response genes and redirecting the cellular repair machinery in the wake of genotoxic stress. Here we review the key mechanistic underpinnings of the relationship between sex steroid hormone receptors and genomic stability. In addition, we summarize emerging research in this area and discuss important implications for cancer prevention and treatment.
Collapse
Affiliation(s)
| | - Divya Naik
- Cleveland Clinic Lerner Research Institute, Cleveland, OH
| | - Andrew Muskara
- Cleveland Clinic Lerner Research Institute, Cleveland, OH
| | - Omar Y Mian
- Cleveland Clinic Lerner Research Institute, Cleveland, OH
- Cleveland Clinic Taussig Cancer Center, Cleveland, OH
| |
Collapse
|
19
|
Deregulated estrogen receptor signaling and DNA damage response in breast tumorigenesis. Biochim Biophys Acta Rev Cancer 2020; 1875:188482. [PMID: 33260050 DOI: 10.1016/j.bbcan.2020.188482] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 11/05/2020] [Accepted: 11/12/2020] [Indexed: 02/07/2023]
Abstract
Carriers of BRCA1 mutations have a higher chance of developing cancers in hormone-responsive tissues like the breast, ovary and prostate, compared to other tissues. These tumors generally exhibit basal-like characters and do not express estrogen receptor (ER) or progesterone receptor (PR). Intriguingly, BRCA1 mutated breast cancers have a less favorable clinical outcome, as they will not respond to hormone therapy. BRCA1 has been reported to exhibit ligand dependent and independent transcriptional inhibition of ER-α; however, there exists a controversy on whether BRCA1 induces or inhibits ER-α expression. The mechanisms associated with resistance of BRCA1 mutated cancers to hormone therapy, as well as the tissue restriction exhibited by BRCA1 mutated tumors are still largely unknown. BRCA1 mutated tumors possess increased DNA damages and decreased genomic integrity, as BRCA1 plays a cardinal role in high fidelity DNA damage repair pathways, like homologous recombination (HR). The existence of cross regulatory signaling networks between ER-α and BRCA1 speculates a role of ER on BRCA1 dependent DDR pathways. Thus, the loss or haploinsufficiency of BRCA1 and the consequential deregulation of ER-α signaling may result in persistence of unrepaired DNA damages, eventually leading to tumorigenesis. Therefore, understanding of this cross-talk between ER-α and BRCA1, with regard to DDR, will provide critical insights to steer drug development and therapy for breast/ovarian cancers. This review discusses the mechanisms by which estrogen and ER signaling influence BRCA1 mediated DNA damage response and repair pathways in the mammalian system.
Collapse
|
20
|
Sjöström M, Veenstra C, Holmberg E, Karlsson P, Killander F, Malmström P, Niméus E, Fernö M, Stål O. Expression of HGF, pMet, and pAkt is related to benefit of radiotherapy after breast-conserving surgery: a long-term follow-up of the SweBCG91-RT randomised trial. Mol Oncol 2020; 14:2713-2726. [PMID: 32946618 PMCID: PMC7607179 DOI: 10.1002/1878-0261.12803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 08/19/2020] [Accepted: 09/11/2020] [Indexed: 01/15/2023] Open
Abstract
Experimental studies suggest that hepatocyte growth factor (HGF) and its transmembrane tyrosine kinase receptor, Met, in part also relying on Akt kinase activity, mediate radioresistance. We investigated the importance of these biomarkers for the risk of ipsilateral breast tumour recurrence (IBTR) after adjuvant radiotherapy (RT) in primary breast cancer. HGF, phosphorylated Met (pMet) and phosphorylated Akt (pAkt) were evaluated immunohistochemically on tissue microarrays from 1004 patients in the SweBCG91‐RT trial, which randomly assigned patients to breast‐conserving therapy, with or without adjuvant RT. HGF was evaluated in the stroma (HGFstr); pMet in the membrane (pMetmem); HGF, pMet and pAkt in the cytoplasm (HGFcyt, pMetcyt, pAktcyt); and pAkt in the nucleus (pAktnuc). The prognostic and treatment predictive effects were evaluated to primary endpoint IBTR as first event during the first 5 years. Patients with tumours expressing low levels of HGFcyt and pMetcyt and high levels of pAktnuc derived a larger benefit from RT [hazard ratio (HR): 0.11 (0.037–0.30), 0.066 (0.016–0.28) and 0.094 (0.028–0.31), respectively] compared to patients with high expression of HGFcyt and pMetcyt, and low pAktnuc [HR: 0.36 (0.19–0.67), 0.35 (0.20–0.64) and 0.47 (0.32–0.71), respectively; interaction analyses: P = 0.052, 0.035 and 0.013, respectively]. These differences remained in multivariable analysis when adjusting for patient age, tumour size, histological grade, St Gallen subtype and systemic treatment (interaction analysis, P‐values: 0.085, 0.027, and 0.023, respectively). This study suggests that patients with immunohistochemically low HGFcyt, low pMetcyt and high pAktnuc may derive an increased benefit from RT after breast‐conserving surgery concerning the risk of developing IBTR.
Collapse
Affiliation(s)
- Martin Sjöström
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden
| | - Cynthia Veenstra
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.,Department of Oncology, Linköping University, Linköping, Sweden
| | - Erik Holmberg
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden
| | - Per Karlsson
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden
| | - Fredrika Killander
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden.,Department of Haematology, Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Per Malmström
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden.,Department of Haematology, Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Emma Niméus
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden.,Division of Surgery, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden.,Department of Surgery, Skåne University Hospital, Lund, Sweden
| | - Mårten Fernö
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden
| | - Olle Stål
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.,Department of Oncology, Linköping University, Linköping, Sweden
| |
Collapse
|
21
|
Chen K, Jiao X, Ashton A, Di Rocco A, Pestell TG, Sun Y, Zhao J, Casimiro MC, Li Z, Lisanti MP, McCue PA, Shen D, Achilefu S, Rui H, Pestell RG. The membrane-associated form of cyclin D1 enhances cellular invasion. Oncogenesis 2020; 9:83. [PMID: 32948740 PMCID: PMC7501870 DOI: 10.1038/s41389-020-00266-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 08/22/2020] [Accepted: 09/02/2020] [Indexed: 02/07/2023] Open
Abstract
The essential G1-cyclin, CCND1, is a collaborative nuclear oncogene that is frequently overexpressed in cancer. D-type cyclins bind and activate CDK4 and CDK6 thereby contributing to G1–S cell-cycle progression. In addition to the nucleus, herein cyclin D1 was also located in the cytoplasmic membrane. In contrast with the nuclear-localized form of cyclin D1 (cyclin D1NL), the cytoplasmic membrane-localized form of cyclin D1 (cyclin D1MEM) induced transwell migration and the velocity of cellular migration. The cyclin D1MEM was sufficient to induce G1–S cell-cycle progression, cellular proliferation, and colony formation. The cyclin D1MEM was sufficient to induce phosphorylation of the serine threonine kinase Akt (Ser473) and augmented extranuclear localized 17β-estradiol dendrimer conjugate (EDC)-mediated phosphorylation of Akt (Ser473). These studies suggest distinct subcellular compartments of cell cycle proteins may convey distinct functions.
Collapse
Affiliation(s)
- Ke Chen
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Xuanmao Jiao
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, PA, 19096, USA
| | - Anthony Ashton
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, PA, 19096, USA
| | - Agnese Di Rocco
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, PA, 19096, USA
| | - Timothy G Pestell
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Yunguang Sun
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Jun Zhao
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, PA, 19096, USA
| | - Mathew C Casimiro
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, PA, 19096, USA.,Dept of Science and Math, Abraham Baldwin Agricultural college, Tifton, GA, 31794, Georgia
| | - Zhiping Li
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, PA, 19096, USA
| | - Michael P Lisanti
- Biomedical Research Centre (BRC), Translational Medicine, School of Environment and Life Sciences, University of Salford, Manchester, United Kingdom
| | - Peter A McCue
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Duanwen Shen
- Departments of Biomedical Engineering, Washington University, St. Louis, MO, 63110, USA
| | - Samuel Achilefu
- Departments of Biomedical Engineering, Washington University, St. Louis, MO, 63110, USA.,Departments of Radiology, Washington University, St. Louis, MO, 63110, USA.,Departments of Biochemistry & Molecular Biophysics, Washington University, St. Louis, MO, 63110, USA
| | - Hallgeir Rui
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Richard G Pestell
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, PA, 19096, USA. .,The Wistar Cancer Center, Wistar Institute, Philadelphia, PA, 19104, USA.
| |
Collapse
|
22
|
Lakshmanan MD, Shaheer K. Endocrine disrupting chemicals may deregulate DNA repair through estrogen receptor mediated seizing of CBP/p300 acetylase. J Endocrinol Invest 2020; 43:1189-1196. [PMID: 32253726 DOI: 10.1007/s40618-020-01241-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 03/27/2020] [Indexed: 12/20/2022]
Abstract
PURPOSE Environmental pollutants are known to induce DNA breaks, leading to genomic instability. Here, we propose a novel mechanism for the genotoxic effects exerted by environmentally exposed endocrine-disrupting chemicals (EDCs). METHODS Bibliographic research and presentation of the analysis. DISCUSSION In mammals, nucleotide excision repair, base excision repair, homologous recombination and non-homologous end-joining pathways are some of the major DNA repair pathways. p300 along with CREB-binding protein (CBP) contributes to chromatin remodeling, DNA damage response and repair of both single- and double-stranded DNA breaks. In addition to its role in DNA repair, CBP/p300 also acts as a coactivator to interact with the estrogen receptor and androgen receptor during its estrogen- and androgen-dependent transactivation, respectively. Since activated estrogen receptors (ERs) seize p300 from the repressed genes and redistribute it to the enhancer genes to activate transcription, the cellular functioning may be based on a balance between these pathways and any disturbance in one may alter the other, leading to undesirable physiological effects. CONCLUSION In conclusion, CBP/p300 is important for DNA repair and nuclear hormone receptor transactivation. Activated hormone receptors can sequester p300 to regulate the hormonal effects. Hence, we believe that activation of ERs by EDCs results in sequestration of CBP/p300 for ER transactivation and transcription initiation of its target genes, leading to a competition for CBP/P300, resulting in the deregulation of all other pathways involving p300/CBP.
Collapse
Affiliation(s)
- M D Lakshmanan
- Molecular Biology Division, Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore, Karnataka, 575018, India.
| | - K Shaheer
- Molecular Biology Division, Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore, Karnataka, 575018, India
| |
Collapse
|
23
|
Helm JS, Rudel RA. Adverse outcome pathways for ionizing radiation and breast cancer involve direct and indirect DNA damage, oxidative stress, inflammation, genomic instability, and interaction with hormonal regulation of the breast. Arch Toxicol 2020. [PMID: 32399610 DOI: 10.1007/s00204-020-02752-z)] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
Abstract
Knowledge about established breast carcinogens can support improved and modernized toxicological testing methods by identifying key mechanistic events. Ionizing radiation (IR) increases the risk of breast cancer, especially for women and for exposure at younger ages, and evidence overall supports a linear dose-response relationship. We used the Adverse Outcome Pathway (AOP) framework to outline and evaluate the evidence linking ionizing radiation with breast cancer from molecular initiating events to the adverse outcome through intermediate key events, creating a qualitative AOP. We identified key events based on review articles, searched PubMed for recent literature on key events and IR, and identified additional papers using references. We manually curated publications and evaluated data quality. Ionizing radiation directly and indirectly causes DNA damage and increases production of reactive oxygen and nitrogen species (RONS). RONS lead to DNA damage and epigenetic changes leading to mutations and genomic instability (GI). Proliferation amplifies the effects of DNA damage and mutations leading to the AO of breast cancer. Separately, RONS and DNA damage also increase inflammation. Inflammation contributes to direct and indirect effects (effects in cells not directly reached by IR) via positive feedback to RONS and DNA damage, and separately increases proliferation and breast cancer through pro-carcinogenic effects on cells and tissue. For example, gene expression changes alter inflammatory mediators, resulting in improved survival and growth of cancer cells and a more hospitable tissue environment. All of these events overlap at multiple points with events characteristic of "background" induction of breast carcinogenesis, including hormone-responsive proliferation, oxidative activity, and DNA damage. These overlaps make the breast particularly susceptible to ionizing radiation and reinforce that these biological activities are important characteristics of carcinogens. Agents that increase these biological processes should be considered potential breast carcinogens, and predictive methods are needed to identify chemicals that increase these processes. Techniques are available to measure RONS, DNA damage and mutation, cell proliferation, and some inflammatory proteins or processes. Improved assays are needed to measure GI and chronic inflammation, as well as the interaction with hormonally driven development and proliferation. Several methods measure diverse epigenetic changes, but it is not clear which changes are relevant to breast cancer. In addition, most toxicological assays are not conducted in mammary tissue, and so it is a priority to evaluate if results from other tissues are generalizable to breast, or to conduct assays in breast tissue. Developing and applying these assays to identify exposures of concern will facilitate efforts to reduce subsequent breast cancer risk.
Collapse
Affiliation(s)
- Jessica S Helm
- Silent Spring Institute, 320 Nevada Street, Suite 302, Newton, MA, 02460, USA
| | - Ruthann A Rudel
- Silent Spring Institute, 320 Nevada Street, Suite 302, Newton, MA, 02460, USA.
| |
Collapse
|
24
|
Helm JS, Rudel RA. Adverse outcome pathways for ionizing radiation and breast cancer involve direct and indirect DNA damage, oxidative stress, inflammation, genomic instability, and interaction with hormonal regulation of the breast. Arch Toxicol 2020; 94:1511-1549. [PMID: 32399610 PMCID: PMC7261741 DOI: 10.1007/s00204-020-02752-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 04/16/2020] [Indexed: 12/15/2022]
Abstract
Knowledge about established breast carcinogens can support improved and modernized toxicological testing methods by identifying key mechanistic events. Ionizing radiation (IR) increases the risk of breast cancer, especially for women and for exposure at younger ages, and evidence overall supports a linear dose-response relationship. We used the Adverse Outcome Pathway (AOP) framework to outline and evaluate the evidence linking ionizing radiation with breast cancer from molecular initiating events to the adverse outcome through intermediate key events, creating a qualitative AOP. We identified key events based on review articles, searched PubMed for recent literature on key events and IR, and identified additional papers using references. We manually curated publications and evaluated data quality. Ionizing radiation directly and indirectly causes DNA damage and increases production of reactive oxygen and nitrogen species (RONS). RONS lead to DNA damage and epigenetic changes leading to mutations and genomic instability (GI). Proliferation amplifies the effects of DNA damage and mutations leading to the AO of breast cancer. Separately, RONS and DNA damage also increase inflammation. Inflammation contributes to direct and indirect effects (effects in cells not directly reached by IR) via positive feedback to RONS and DNA damage, and separately increases proliferation and breast cancer through pro-carcinogenic effects on cells and tissue. For example, gene expression changes alter inflammatory mediators, resulting in improved survival and growth of cancer cells and a more hospitable tissue environment. All of these events overlap at multiple points with events characteristic of "background" induction of breast carcinogenesis, including hormone-responsive proliferation, oxidative activity, and DNA damage. These overlaps make the breast particularly susceptible to ionizing radiation and reinforce that these biological activities are important characteristics of carcinogens. Agents that increase these biological processes should be considered potential breast carcinogens, and predictive methods are needed to identify chemicals that increase these processes. Techniques are available to measure RONS, DNA damage and mutation, cell proliferation, and some inflammatory proteins or processes. Improved assays are needed to measure GI and chronic inflammation, as well as the interaction with hormonally driven development and proliferation. Several methods measure diverse epigenetic changes, but it is not clear which changes are relevant to breast cancer. In addition, most toxicological assays are not conducted in mammary tissue, and so it is a priority to evaluate if results from other tissues are generalizable to breast, or to conduct assays in breast tissue. Developing and applying these assays to identify exposures of concern will facilitate efforts to reduce subsequent breast cancer risk.
Collapse
Affiliation(s)
- Jessica S Helm
- Silent Spring Institute, 320 Nevada Street, Suite 302, Newton, MA, 02460, USA
| | - Ruthann A Rudel
- Silent Spring Institute, 320 Nevada Street, Suite 302, Newton, MA, 02460, USA.
| |
Collapse
|
25
|
Maidarti M, Anderson RA, Telfer EE. Crosstalk between PTEN/PI3K/Akt Signalling and DNA Damage in the Oocyte: Implications for Primordial Follicle Activation, Oocyte Quality and Ageing. Cells 2020; 9:200. [PMID: 31947601 PMCID: PMC7016612 DOI: 10.3390/cells9010200] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/06/2020] [Accepted: 01/13/2020] [Indexed: 12/18/2022] Open
Abstract
The preservation of genome integrity in the mammalian female germline from primordial follicle arrest to activation of growth to oocyte maturation is fundamental to ensure reproductive success. As oocytes are formed before birth and may remain dormant for many years, it is essential that defence mechanisms are monitored and well maintained. The phosphatase and tensin homolog of chromosome 10 (PTEN)/phosphatidylinositol 3-kinase (PI3K)/protein kinase B (PKB, Akt) is a major signalling pathway governing primordial follicle recruitment and growth. This pathway also contributes to cell growth, survival and metabolism, and to the maintenance of genomic integrity. Accelerated primordial follicle activation through this pathway may result in a compromised DNA damage response (DDR). Additionally, the distinct DDR mechanisms in oocytes may become less efficient with ageing. This review considers DNA damage surveillance mechanisms and their links to the PTEN/PI3K/Akt signalling pathway, impacting on the DDR during growth activation of primordial follicles, and in ovarian ageing. Targeting DDR mechanisms within oocytes may be of value in developing techniques to protect ovaries against chemotherapy and in advancing clinical approaches to regulate primordial follicle activation.
Collapse
Affiliation(s)
- Mila Maidarti
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK; (M.M.); (R.A.A.)
- Institute of Cell Biology, University of Edinburgh, Edinburgh EH9 3FF, UK
- Obstetrics and Gynaecology Department, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
| | - Richard A. Anderson
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK; (M.M.); (R.A.A.)
| | - Evelyn E. Telfer
- Institute of Cell Biology, University of Edinburgh, Edinburgh EH9 3FF, UK
| |
Collapse
|
26
|
DNA damage repair functions and targeted treatment in breast cancer. Breast Cancer 2020; 27:355-362. [PMID: 31898156 DOI: 10.1007/s12282-019-01038-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 12/19/2019] [Indexed: 12/14/2022]
Abstract
Cell DNA is continuously attacked by endogenous and exogenous agents, which causes DNA damage. During long-term evolution, complex defense systems for DNA damage repair are formed by cells to maintain genome stability. Defects in the DNA damage repair process may lead to various diseases, including tumors. Therefore, DNA damage repair systems have become a new anti-tumor drug target. To date, a number of inhibitors related to DNA damage repair systems have been developed, particularly for tumors with BRCA1 and BRCA2 mutations. Poly (ADP-ribose) polymerase inhibitors developed by synthetic lethality are widely used in individualized tumor therapy. In this review, we briefly introduce the mechanisms underlying DNA damage repair, particularly in breast cancer, and mainly focus on new treatments targeting the DNA damage repair pathway in breast cancer.
Collapse
|
27
|
Tu Z, Mu X, Chen X, Geng Y, Zhang Y, Li Q, Gao R, Liu T, Wang Y, He J. Dibutyl phthalate exposure disrupts the progression of meiotic prophase I by interfering with homologous recombination in fetal mouse oocytes. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 252:388-398. [PMID: 31158667 DOI: 10.1016/j.envpol.2019.05.107] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/05/2019] [Accepted: 05/21/2019] [Indexed: 06/09/2023]
Abstract
Dibutyl phthalate (DBP), one of the most widely used plasticizers, is a known environmental endocrine disruptor that impairs male and female fertility. In this study, oral administration of DBP was given to pregnant mice on 14.5 days post coitus (dpc) for 3 days; and additionally, DBP was added into the culture of 14.5 dpc fetal ovaries for 3 days. DBP exposure during gestation disturbed the progression of meiotic prophase I of mouse oocytes, specifically from the zygotene to pachytene stages. Meanwhile, the DBP-exposed pachytene oocytes showed increased homologous recombination sites and unrepaired DNA damage. Furthermore, DBP caused DNA damage by increasing oxidative stress, decreased the expression of multiple critical meiotic regulators, and consequently induced oocyte apoptosis. Moreover, the effect of DBP on meiosis I prophase involved estrogen receptors α and β. Collectively, these results demonstrated a set of meiotic defects in DBP-exposed fetal oocytes. As aberrations in homologous recombination can result in aneuploid gametes and embryos, this study provides new support for the deleterious effects of phthalates.
Collapse
Affiliation(s)
- Zhihan Tu
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing, 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China
| | - Xinyi Mu
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China; College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, PR China
| | - Xuemei Chen
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing, 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China
| | - Yanqing Geng
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China; College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, PR China
| | - Yan Zhang
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing, 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China
| | - Qingying Li
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China; College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, PR China
| | - Rufei Gao
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing, 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China
| | - Taihang Liu
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China; College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, PR China
| | - Yingxiong Wang
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China; College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, PR China
| | - Junlin He
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing, 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China.
| |
Collapse
|
28
|
Bhardwaj P, Au CC, Benito-Martin A, Ladumor H, Oshchepkova S, Moges R, Brown KA. Estrogens and breast cancer: Mechanisms involved in obesity-related development, growth and progression. J Steroid Biochem Mol Biol 2019; 189:161-170. [PMID: 30851382 PMCID: PMC6502693 DOI: 10.1016/j.jsbmb.2019.03.002] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 02/27/2019] [Accepted: 03/01/2019] [Indexed: 12/21/2022]
Abstract
Obesity is a risk factor for estrogen receptor-positive (ER+) breast cancer after menopause. The pro-proliferative effects of estrogens are well characterized and there is a growing body of evidence to also suggest an important role in tumorigenesis. Importantly, obesity not only increases the risk of breast cancer, but it also increases the risk of recurrence and cancer-associated death. Aromatase is the rate-limiting enzyme in estrogen biosynthesis and its expression in breast adipose stromal cells is hypothesized to drive the growth of breast tumors and confer resistance to endocrine therapy in obese postmenopausal women. The molecular regulation of aromatase has been characterized in response to many obesity-related molecules, including inflammatory mediators and adipokines. This review is aimed at providing an overview of our current knowledge in relation to the regulation of estrogens in adipose tissue and their role in driving breast tumor development, growth and progression.
Collapse
Affiliation(s)
- Priya Bhardwaj
- Department of Medicine, Weill Cornell Medicine, New York, USA; Graduate School of Medical Sciences, Weill Cornell Medicine, New York, USA
| | - CheukMan C Au
- Department of Medicine, Weill Cornell Medicine, New York, USA
| | | | - Heta Ladumor
- Department of Medicine, Weill Cornell Medicine, New York, USA; Weill Cornell Medicine - Qatar, Doha, Qatar
| | | | - Ruth Moges
- Department of Medicine, Weill Cornell Medicine, New York, USA
| | - Kristy A Brown
- Department of Medicine, Weill Cornell Medicine, New York, USA; Graduate School of Medical Sciences, Weill Cornell Medicine, New York, USA; Department of Physiology, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
29
|
Elucidation of possible molecular mechanisms underlying the estrogen-induced disruption of cartilage development in zebrafish larvae. Toxicol Lett 2018; 289:22-27. [DOI: 10.1016/j.toxlet.2018.02.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 02/09/2018] [Accepted: 02/13/2018] [Indexed: 11/17/2022]
|
30
|
Parl FF, Crooke PS, Plummer WD, Dupont WD. Genomic-Epidemiologic Evidence That Estrogens Promote Breast Cancer Development. Cancer Epidemiol Biomarkers Prev 2018; 27:899-907. [PMID: 29789325 DOI: 10.1158/1055-9965.epi-17-1174] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 02/23/2018] [Accepted: 05/14/2018] [Indexed: 12/21/2022] Open
Abstract
Background: Estrogens are a prime risk factor for breast cancer, yet their causal relation to tumor formation remains uncertain. A recent study of 560 breast cancers identified 82 genes with 916 point mutations as drivers in the genesis of this malignancy. Because estrogens play a major role in breast cancer development and are also known to regulate the expression of numerous genes, we hypothesize that the 82 driver genes are likely to be influenced by estrogens, such as 17ß-estradiol (E2), and the estrogen receptor ESR1 (ERα). Because different types of tumors are characterized by unique sets of cancer driver genes, we also argue that the fraction of driver genes regulated by E2-ESR1 is lower in malignancies not associated with estrogens, e.g., acute myeloid leukemia (AML).Methods: We performed a literature search of each driver gene to determine its E2-ESR1 regulation.Results: Fifty-three of the 82 driver genes (64.6%) identified in breast cancers showed evidence of E2-ESR1 regulation. In contrast, only 19 of 54 mutated driver genes (35.2%) identified in AML were linked to E2-ESR1. Among the 916 driver mutations found in breast cancers, 813 (88.8%) were linked to E2-ESR1 compared with 2,046 of 3,833 in AML (53.4%).Conclusions: Risk assessment revealed that mutations in estrogen-regulated genes are much more likely to be associated with elevated breast cancer risk, while mutations in unregulated genes are more likely to be associated with AML.Impact: These results increase the plausibility that estrogens promote breast cancer development. Cancer Epidemiol Biomarkers Prev; 27(8); 899-907. ©2018 AACR.
Collapse
Affiliation(s)
- Fritz F Parl
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, Tennessee.
| | - Philip S Crooke
- Department of Mathematics, Vanderbilt University, Nashville, Tennessee
| | - W Dale Plummer
- Department of Health Policy, Vanderbilt University, Nashville, Tennessee
| | - William D Dupont
- Department of Health Policy, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
31
|
Eric JG, John BC, Gerald BS, Hidetaka E, Richard GS, Shizue I, Young-Min K, Amy Berrington de G, Waka O, Kei N. Bioavailable serum estradiol may alter radiation risk of postmenopausal breast cancer: a nested case-control study. Int J Radiat Biol 2018; 94:97-105. [PMID: 29307255 PMCID: PMC6135639 DOI: 10.1080/09553002.2018.1419303] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 11/21/2017] [Accepted: 12/11/2017] [Indexed: 01/05/2023]
Abstract
PURPOSE Ionizing radiation and high levels of circulating estradiol are known breast cancer carcinogens. We investigated the risk of first primary postmenopausal breast cancer in relation to the combined effects of whole-body ionizing radiation exposure and prediagnostic levels of postmenopausal sex hormones, particularly bioavailable estradiol (bE2). MATERIALS AND METHODS A nested case-control study of 57 incident breast cancer cases matched with 110 controls among atomic bomb survivors. Joint effects of breast radiation dose and circulating levels of sex hormones were assessed using binary regression and path analysis. RESULTS AND CONCLUSION Radiation exposure, higher levels of bE2, testosterone and progesterone, and established reproductive risk factors were positively associated with postmenopausal breast cancer risk. A test for mediation of the effect of radiation via bE2 level suggested a small (14%) but significant mediation (p = 0.004). The estimated interaction between radiation and bE2 was large but not significant (interaction = 3.86; p = 0.32). There is accumulating evidence that ionizing radiation not only damages DNA but also alters other organ systems. While caution is needed, some portion of the radiation risk of postmenopausal breast cancer appeared to be mediated through bE2 levels, which may be evidence for cancer risks due to both direct and indirect effects of radiation.
Collapse
Affiliation(s)
- J. Grant Eric
- Radiation Effects Research Foundation, Hiroshima, Japan
| | - B. Cologne John
- Department of Statistics, Radiation Effects Research Foundation, Hiroshima, Japan
| | - B. Sharp Gerald
- Epidemiology Branch, Basic Sciences Program, Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Eguchi Hidetaka
- Translational Research Division, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| | - G. Stevens Richard
- Department of Community Medicine, University of Connecticut Health Center, Connecticut, USA
| | - Izumi Shizue
- Center for Data Science Education and Research, Shiga University, Hikone, Japan
| | - Kim Young-Min
- Department of Statistics, College of Natural Science, Kyungpook National University, Daegu, South Korea
| | | | - Ohishi Waka
- Department of Clinical Studies, Radiation Effects Research Foundation, Hiroshima, Japan
| | - Nakachi Kei
- Department of Molecular Biosciences, Radiation Effects Research Foundation, Hiroshima, Japan
| |
Collapse
|
32
|
Why Human Papillomaviruses Activate the DNA Damage Response (DDR) and How Cellular and Viral Replication Persists in the Presence of DDR Signaling. Viruses 2017; 9:v9100268. [PMID: 28934154 PMCID: PMC5691620 DOI: 10.3390/v9100268] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 09/12/2017] [Accepted: 09/15/2017] [Indexed: 12/15/2022] Open
Abstract
Human papillomaviruses (HPV) require the activation of the DNA damage response (DDR) in order to undergo a successful life cycle. This activation presents a challenge for the virus and the infected cell: how does viral and host replication proceed in the presence of a DDR that ordinarily arrests replication; and how do HPV16 infected cells retain the ability to proliferate in the presence of a DDR that ordinarily arrests the cell cycle? This raises a further question: why do HPV activate the DDR? The answers to these questions are only partially understood; a full understanding could identify novel therapeutic strategies to target HPV cancers. Here, we propose that the rapid replication of an 8 kb double stranded circular genome during infection creates aberrant DNA structures that attract and activate DDR proteins. Therefore, HPV replication in the presence of an active DDR is a necessity for a successful viral life cycle in order to resolve these DNA structures on viral genomes; without an active DDR, successful replication of the viral genome would not proceed. We discuss the essential role of TopBP1 in this process and also how viral and cellular replication proceeds in HPV infected cells in the presence of DDR signals.
Collapse
|
33
|
Di Sante G, Di Rocco A, Pupo C, Casimiro MC, Pestell RG. Hormone-induced DNA damage response and repair mediated by cyclin D1 in breast and prostate cancer. Oncotarget 2017; 8:81803-81812. [PMID: 29137223 PMCID: PMC5669849 DOI: 10.18632/oncotarget.19413] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 06/26/2017] [Indexed: 11/25/2022] Open
Abstract
Cell cycle control proteins govern events that leads to the production of two identical daughter cells. Distinct sequential temporal phases, Gap 1 (G1), Gap 0 (G0), Synthesis (S), Gap 2 (G2) and Mitosis (M) are negotiated through a series of check points during which the favorability of the local cellular environment is assessed, prior to replicating DNA [1]. Cyclin D1 has been characterized as a key regulatory subunit of the holoenzyme that promotes the G1/S-phase transition through phosphorylating the pRB protein. Cyclin D1 overexpression is considered a driving force in several types of cancers and cdk inhibitors are being used effectively in the clinic for treatment of ERα+ breast cancer [1, 2]. Genomic DNA is assaulted by damaging ionizing radiation, chemical carcinogens, and reactive oxygen species (ROS) which are generated by cellular metabolism. Furthermore, specific hormones including estrogens [3, 4] and androgens [5] govern pathways that damage DNA. Defects in the DNA Damage Response (DDR) pathway can lead to genomic instability and cancer. Evidence is emerging that cyclin D1 bind proteins involved in DNA repair including BRCA1 [6], RAD51 [7], BRCA2 [8] and is involved in the DNA damage and DNA repair processes [7, 8]. Because the repair of damaged DNA appears to be an important and unexpected role for cyclin D1, and inhibitors of cyclin D1-dependent kinase activity are being used in the clinic, the latest findings on the role of cyclin D1 in mediating the DDR including the DDR induced by the hormones estrogen [9] and androgen [10, 11] is reviewed.
Collapse
Affiliation(s)
- Gabriele Di Sante
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, PA, USA
| | - Agnese Di Rocco
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, PA, USA
| | - Claudia Pupo
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, PA, USA
| | - Mathew C Casimiro
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, PA, USA
| | - Richard G Pestell
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, PA, USA.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| |
Collapse
|
34
|
Chen X, Ma N, Zhou Z, Wang Z, Hu Q, Luo J, Mei X, Yang Z, Zhang L, Wang X, Feng Y, Yu X, Ma J, Guo X. Estrogen Receptor Mediates the Radiosensitivity of Triple-Negative Breast Cancer Cells. Med Sci Monit 2017; 23:2674-2683. [PMID: 28570501 PMCID: PMC5466376 DOI: 10.12659/msm.904810] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background This study aimed to evaluate differences in the radiosensitivities of triple-negative breast cancer (TNBC) and luminal-type breast cancer cells and to investigate the effects of estrogen receptor (ER) expression on the biological behaviors of the cells. Material/Methods Colony-forming assays were performed to detect differences in radiosensitivities in breast cancer cell lines. Gene transfection technology was used to introduce the expression of ERα in TNBC cells to compare the difference in radiosensitivity between the TNBC cells and ERα transfected cells. CCK-8 assays were used to observe changes in the proliferation of TNBC cells after ERα transfection. Immunofluorescence was used to detect the number of γH2AX foci in nuclei. Flow cytometry was used to detect changes in cell cycle distribution and apoptosis. Western blotting was used to detect changes in autophagy-associated proteins. Results The radioresistance of the TNBC cell line MDA-MB-231 (231 cells) was greater than that of ERα-positive luminal-type breast cancer cell line MCF-7. Moreover, 231 cell proliferation and radioresistance decreased after ERα transfection. Interestingly, ERα-transfected 231 cells showed increased double-stranded breaks and delayed repair compared with 231 cells, and ERα-transfected 231 cells showed increased G2/M phase arrest and apoptosis after irradiation compared with those in 231 cells. ERα transfection in 231 cells reduced autophagy-related protein expression, suggesting that autophagy activity decreased in 231 ER-positive cells after irradiation. Conclusions TNBC cells were more resistant to radiation than luminal-type breast cancer cells. ERα expression may have major roles in modulating breast cancer cell radiosensitivity.
Collapse
Affiliation(s)
- Xingxing Chen
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China (mainland).,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China (mainland)
| | - Ningyi Ma
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China (mainland).,Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China (mainland)
| | - Zhirui Zhou
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China (mainland).,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China (mainland)
| | - Ziliang Wang
- Cancer Institutions, Fudan University Shanghai Cancer Center, Shanghai, China (mainland)
| | - Qunchao Hu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China (mainland).,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China (mainland)
| | - Jurui Luo
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China (mainland).,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China (mainland)
| | - Xin Mei
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China (mainland).,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China (mainland)
| | - Zhaozhi Yang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China (mainland).,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China (mainland)
| | - Li Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China (mainland).,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China (mainland)
| | - Xiaofang Wang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China (mainland).,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China (mainland)
| | - Yan Feng
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China (mainland).,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China (mainland)
| | - Xiaoli Yu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China (mainland).,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China (mainland)
| | - Jinli Ma
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China (mainland).,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China (mainland)
| | - Xiaomao Guo
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China (mainland).,Department of Oncology, Shanghai Medical College, Fudan University,, Shanghai, China (mainland)
| |
Collapse
|
35
|
Majidinia M, Yousefi B. DNA repair and damage pathways in breast cancer development and therapy. DNA Repair (Amst) 2017; 54:22-29. [DOI: 10.1016/j.dnarep.2017.03.009] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 03/31/2017] [Accepted: 03/31/2017] [Indexed: 12/22/2022]
|
36
|
DNA damage repair in breast cancer and its therapeutic implications. Pathology 2016; 49:156-165. [PMID: 28034453 DOI: 10.1016/j.pathol.2016.11.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 10/06/2016] [Accepted: 11/02/2016] [Indexed: 11/23/2022]
Abstract
The DNA damage response (DDR) involves the activation of numerous cellular activities that repair DNA lesions and maintain genomic integrity, and is critical in preventing tumorigenesis. Inherited or acquired mutations in specific genes involved in the DNA damage response, for example the breast cancer susceptibility genes 1/2 (BRCA1/2), phosphatase and tensin homolog (PTEN) and P53 are associated with various subtypes of breast cancer. Such changes can render breast cancer cells particularly sensitive to specific DNA damage response inhibitors, for example BRCA1/2 germline mutated cells are sensitive to poly (ADP-ribose) polymerase (PARP) inhibitors. The aims of this review are to discuss specific DNA damage response defects in breast cancer and to present the current stage of development of various DDR inhibitors (namely PARP, ATM/ATR, DNA-PK, PARG, RECQL5, FEN1 and APE1) for breast cancer mono- and combination therapy.
Collapse
|
37
|
Alshareeda AT, Negm OH, Aleskandarany MA, Green AR, Nolan C, TigHhe PJ, Madhusudan S, Ellis IO, Rakha EA. Clinical and biological significance of RAD51 expression in breast cancer: a key DNA damage response protein. Breast Cancer Res Treat 2016; 159:41-53. [DOI: 10.1007/s10549-016-3915-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Accepted: 07/15/2016] [Indexed: 01/15/2023]
|
38
|
Li L, Chen X, Liu CC, Lee LS, Man C, Cheng SH. Phytoestrogen Bakuchiol Exhibits In Vitro and In Vivo Anti-breast Cancer Effects by Inducing S Phase Arrest and Apoptosis. Front Pharmacol 2016; 7:128. [PMID: 27252650 PMCID: PMC4877368 DOI: 10.3389/fphar.2016.00128] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 05/05/2016] [Indexed: 11/25/2022] Open
Abstract
Phytoestrogen has been proposed as an alternative to hormone replacement therapy, which has been demonstrated to promote a high risk of breast cancer. However, the effect of phytoestrogen on breast cancer development has not been fully understood. Bakuchiol is an active ingredient of a traditional Chinese herbal medicine Fructus Psoraleae, the dried ripe fruit of Psoralea corylifolia L. (Fabaceae). The in vitro and in vivo estrogenic activities and anti-breast cancer effects of bakuchiol have not been well-studied. We found that bakuchiol induced the GFP expression in transgenic medaka (Oryzias melastigma, Tg, Chg:GFP) dose-dependently (0–1 μg/ml), demonstrating its in vivo estrogenic activity. Low dose of bakuchiol (1 μg/ml) induced the cell proliferation and ERα expression in MCF-7 cells, which could be blocked by the anti-estrogen ICI 182780, suggesting the in vitro estrogenic activity of bakuchiol. Our data indicated that high doses of bakuchiol (>2 μg/ml) inhibited breast cancer cell growth, with a stronger anti-proliferative effect than resveratrol, a widely studied analog of bakuchiol. High doses of bakuchiol (4, 7, and 10 μg/ml) were used for the further in vitro anti-breast cancer studies. Bakuchiol induced ERβ expression and suppressed ERα expression in MCF-7 cells. It also induced S phase arrest in both MCF-7 and MDA-MB-231 cells, which could be rescued by caffeine. Knock-down of p21 also marginally rescued S phase arrest in MCF-7 cells. The S phase arrest was accompanied by the upregulation of ATM, P-Cdc2 (Tyr15), Myt1, P-Wee1 (Ser642), p21 and Cyclin B1, suggesting that blocking of Cdc2 activation may play an important role in bakuchiol-induced S phase arrest. Furthermore, bakuchiol induced cell apoptosis and disturbed mitochondrial membrane potential in MCF-7 cells. The bakuchiol-induced apoptosis was associated with increased expression of Caspase family and Bcl-2 family proteins, suggesting that bakuchiol may induce apoptosis via intrinsic apoptotic pathway. The in vivo anti-breast cancer effect of bakuchiol was further proved in zebrafish (Danio rerio, wild-type AB) xenografts. 0.5 μg/ml of bakuchiol significantly reduced the MCF-7 cell mass in zebrafish xenografts. Overall, these results suggested the potential of using bakuchiol in HRT and breast cancer treatment.
Collapse
Affiliation(s)
- Li Li
- Department of Biomedical Sciences, City University of Hong Kong Hong Kong, China
| | - Xueping Chen
- Vitargent (International) Biotechnology Limited Hong Kong, China
| | - Chi C Liu
- Department of Biomedical Sciences, City University of Hong Kong Hong Kong, China
| | - Lai S Lee
- Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University Hong Kong, China
| | - Cornelia Man
- Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University Hong Kong, China
| | - Shuk H Cheng
- Department of Biomedical Sciences, City University of Hong Kong Hong Kong, China
| |
Collapse
|
39
|
Matta J, Morales L, Ortiz C, Adams D, Vargas W, Casbas P, Dutil J, Echenique M, Suárez E. Estrogen Receptor Expression Is Associated with DNA Repair Capacity in Breast Cancer. PLoS One 2016; 11:e0152422. [PMID: 27032101 PMCID: PMC4816515 DOI: 10.1371/journal.pone.0152422] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Accepted: 03/14/2016] [Indexed: 02/07/2023] Open
Abstract
Estrogen-receptor-positive (ER+) tumors employ complex signaling that engages in crosstalk with multiple pathways through genomic and non-genomic regulation. A greater understanding of these pathways is important for developing improved biomarkers that can better determine treatment choices, risk of recurrence and cancer progression. Deficiencies in DNA repair capacity (DRC) is a hallmark of breast cancer (BC); therefore, in this work we tested whether ER signaling influences DRC. We analyzed the association between ER positivity (% receptor activation) and DRC in 270 BC patients, then further stratified our analysis by HER2 receptor status. Our results show that among HER2 negative, the likelihood of having low DRC values among ER- women is 1.92 (95% CI: 1.03, 3.57) times the likelihood of having low DRC values among ER+ women, even adjusting for different potential confounders (p<0.05); however, a contrary pattern was observed among HER2 positives women. In conclusion, there is an association between DRC levels and ER status, and this association is modified by HER2 receptor status. Adding a DNA repair capacity test to hormone receptor testing may provide new information on defective DNA repair phenotypes, which could better stratify BC patients who have ER+ tumors. ER+/HER2- tumors are heterogeneous, incompletely defined, and clinically challenging to treat; the addition of a DRC test could better characterize and classify these patients as well as help clinicians select optimal therapies, which could improve outcomes and reduce recurrences.
Collapse
Affiliation(s)
- Jaime Matta
- Department of Basic Sciences, Division of Pharmacology & Toxicology, Ponce Health Sciences University-School of Medicine, Ponce Research Institute, Ponce, Puerto Rico, United States of America
- Department of Basic Sciences, Division of Cancer Biology, Ponce Health Sciences University-School of Medicine, Ponce Research Institute, Ponce, Puerto Rico, United States of America
- * E-mail:
| | - Luisa Morales
- Public Health Program, Ponce Health Sciences University, Ponce, Puerto Rico, United States of America
| | - Carmen Ortiz
- Department of Basic Sciences, Division of Pharmacology & Toxicology, Ponce Health Sciences University-School of Medicine, Ponce Research Institute, Ponce, Puerto Rico, United States of America
- Department of Basic Sciences, Division of Cancer Biology, Ponce Health Sciences University-School of Medicine, Ponce Research Institute, Ponce, Puerto Rico, United States of America
| | - Damian Adams
- Department of Basic Sciences, Division of Pharmacology & Toxicology, Ponce Health Sciences University-School of Medicine, Ponce Research Institute, Ponce, Puerto Rico, United States of America
- Department of Basic Sciences, Division of Cancer Biology, Ponce Health Sciences University-School of Medicine, Ponce Research Institute, Ponce, Puerto Rico, United States of America
| | - Wanda Vargas
- Department of Basic Sciences, Division of Pharmacology & Toxicology, Ponce Health Sciences University-School of Medicine, Ponce Research Institute, Ponce, Puerto Rico, United States of America
- Department of Basic Sciences, Division of Cancer Biology, Ponce Health Sciences University-School of Medicine, Ponce Research Institute, Ponce, Puerto Rico, United States of America
| | - Patricia Casbas
- Department of Basic Sciences, Division of Cancer Biology, Ponce Health Sciences University-School of Medicine, Ponce Research Institute, Ponce, Puerto Rico, United States of America
- Department of Basic Sciences, Division of Biochemistry, Ponce Health Sciences University-School of Medicine, Ponce Research Institute, Ponce, Puerto Rico, United States of America
| | - Julie Dutil
- Department of Basic Sciences, Division of Cancer Biology, Ponce Health Sciences University-School of Medicine, Ponce Research Institute, Ponce, Puerto Rico, United States of America
- Department of Basic Sciences, Division of Biochemistry, Ponce Health Sciences University-School of Medicine, Ponce Research Institute, Ponce, Puerto Rico, United States of America
| | - Miguel Echenique
- Auxilio Mutuo Hospital, San Juan, Puerto Rico, United States of America
| | - Erick Suárez
- Department of Biostatistics and Epidemiology, Graduate School of Public Health, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico, United States of America
| |
Collapse
|
40
|
Akt-mediated phosphorylation of XLF impairs non-homologous end-joining DNA repair. Mol Cell 2015; 57:648-661. [PMID: 25661488 DOI: 10.1016/j.molcel.2015.01.005] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 10/14/2014] [Accepted: 12/29/2014] [Indexed: 01/01/2023]
Abstract
Deficiency in repair of damaged DNA leads to genomic instability and is closely associated with tumorigenesis. Most DNA double-strand-breaks (DSBs) are repaired by two major mechanisms, homologous-recombination (HR) and non-homologous-end-joining (NHEJ). Although Akt has been reported to suppress HR, its role in NHEJ remains elusive. Here, we report that Akt phosphorylates XLF at Thr181 to trigger its dissociation from the DNA ligase IV/XRCC4 complex, and promotes its interaction with 14-3-3β leading to XLF cytoplasmic retention, where cytosolic XLF is subsequently degraded by SCF(β-TRCP) in a CKI-dependent manner. Physiologically, upon DNA damage, XLF-T181E expressing cells display impaired NHEJ and elevated cell death. Whereas a cancer-patient-derived XLF-R178Q mutant, deficient in XLF-T181 phosphorylation, exhibits an elevated tolerance of DNA damage. Together, our results reveal a pivotal role for Akt in suppressing NHEJ and highlight the tight connection between aberrant Akt hyper-activation and deficiency in timely DSB repair, leading to genomic instability and tumorigenesis.
Collapse
|
41
|
Pérez-Tenorio G, Karlsson E, Stål O. Clinical value of isoform-specific detection and targeting of AKT1, AKT2 and AKT3 in breast cancer. BREAST CANCER MANAGEMENT 2014. [DOI: 10.2217/bmt.14.35] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
SUMMARY Overactivation of the PI3K/AKT signaling pathway is frequently reported in breast cancer, consequently inhibitors targeting this pathway are clinically useful. AKT constitutes a hub in the regulation of several cancer hallmarks, such as proliferation, survival and migration. Three AKT isoforms, named AKT1, AKT2 and AKT3, are identified in humans. AKT alterations, mainly upregulation of phosphorylated AKT in tumors may have prognostic and predictive value. Moreover, the AKT isoforms may possess partly divergent cellular functions and be upregulated in certain breast cancer subtypes, suggesting the importance of isoform-specific analyses. In conclusion, AKT isoform-specific detection and targeting in different tumor subtypes will hopefully result into a further developed personalized medicine.
Collapse
Affiliation(s)
- Gizeh Pérez-Tenorio
- Department of Clinical & Experimental Medicine & Department of Oncology, Linköping University, Linköping, SE-58185, Sweden
| | - Elin Karlsson
- Department of Clinical & Experimental Medicine & Department of Oncology, Linköping University, Linköping, SE-58185, Sweden
| | - Olle Stål
- Department of Clinical & Experimental Medicine & Department of Oncology, Linköping University, Linköping, SE-58185, Sweden
| |
Collapse
|
42
|
DNA damage response and prostate cancer: defects, regulation and therapeutic implications. Oncogene 2014; 34:2815-22. [PMID: 25132269 PMCID: PMC4333141 DOI: 10.1038/onc.2014.238] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 06/25/2014] [Accepted: 06/25/2014] [Indexed: 02/07/2023]
Abstract
DNA damage response (DDR) includes the activation of numerous cellular activities that prevent duplication of DNA lesions and maintain genomic integrity, which is critical for the survival of normal and cancer cells. Specific genes involved in the DDR such as BRCA1/2 and P53 are mutated during prostate cancer progression, while various oncogenic signaling such as Akt and c-Myc are activated, enhancing the replication stress and increasing the genomic instability of cancer cells. These events may render prostate cancer cells particularly sensitive to inhibition of specific DDR pathways, such as PARP in homologous recombination (HR) DNA repair and Chk1 in cell cycle checkpoint and DNA repair, creating opportunities for synthetic lethality or synergistic cytotoxicity. Recent reports highlight the critical role of androgen receptor (AR) as a regulator of DDR genes, providing a rationale for combining DNA-damaging agents or targeted DDR inhibitors with hormonal manipulation or AR inhibition as treatment for aggressive disease. The aims of this review are to discuss specific DDR defects in prostate cancer that occur during disease progression, to summarize recent advances in understanding the regulation of DDR in prostate cancer, and to present potential therapeutic opportunities through combinational targeting of the intact components of DDR signaling pathways.
Collapse
|
43
|
Retarding the senescence of human vascular endothelial cells induced by hydrogen peroxide: effects of 17beta-estradiol (E2) mediated mitochondria protection. Biogerontology 2014; 15:367-75. [DOI: 10.1007/s10522-014-9507-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 05/30/2014] [Indexed: 01/26/2023]
|
44
|
Li Z, Chen K, Jiao X, Wang C, Willmarth NE, Casimiro MC, Li W, Ju X, Kim SH, Lisanti MP, Katzenellenbogen JA, Pestell RG. Cyclin D1 integrates estrogen-mediated DNA damage repair signaling. Cancer Res 2014; 74:3959-70. [PMID: 24830723 DOI: 10.1158/0008-5472.can-13-3137] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The cyclin D1 gene encodes the regulatory subunit of a holoenyzme that phosphorylates the retinoblastoma protein (pRb) and nuclear respiratory factor (NRF1) proteins. The abundance of cyclin D1 determines estrogen-dependent gene expression in the mammary gland of mice. Using estradiol (E2) and an E2-dendrimer conjugate that is excluded from the nucleus, we demonstrate that E2 delays the DNA damage response (DDR) via an extranuclear mechanism. The E2-induced DDR required extranuclear cyclin D1, which bound ERα at the cytoplasmic membrane and augmented AKT phosphorylation (Ser473) and γH2AX foci formation. In the nucleus, E2 inhibited, whereas cyclin D1 enhanced homology-directed DNA repair. Cyclin D1 was recruited to γH2AX foci by E2 and induced Rad51 expression. Cyclin D1 governs an essential role in the E2-dependent DNA damage response via a novel extranuclear function. The dissociable cytoplasmic function to delay the E2-mediated DDR together with the nuclear enhancement of DNA repair uncovers a novel extranuclear function of cyclin D1 that may contribute to the role of E2 in breast tumorigenesis.
Collapse
Affiliation(s)
- Zhiping Li
- Authors' Affiliations: Departments of Cancer Biology and Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania; and
| | - Ke Chen
- Authors' Affiliations: Departments of Cancer Biology and Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania; and
| | - Xuanmao Jiao
- Authors' Affiliations: Departments of Cancer Biology and Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania; and
| | - Chenguang Wang
- Authors' Affiliations: Departments of Cancer Biology and Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania; and
| | - Nicole E Willmarth
- Authors' Affiliations: Departments of Cancer Biology and Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania; and
| | - Mathew C Casimiro
- Authors' Affiliations: Departments of Cancer Biology and Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania; and
| | - Weihua Li
- Authors' Affiliations: Departments of Cancer Biology and Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania; and
| | - Xiaoming Ju
- Authors' Affiliations: Departments of Cancer Biology and Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania; and
| | - Sung Hoon Kim
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Michael P Lisanti
- Stem Cell Biology and Regenerative Medicine, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania; and
| | | | - Richard G Pestell
- Authors' Affiliations: Departments of Cancer Biology and Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania; and
| |
Collapse
|
45
|
Caldon CE. Estrogen signaling and the DNA damage response in hormone dependent breast cancers. Front Oncol 2014; 4:106. [PMID: 24860786 PMCID: PMC4030134 DOI: 10.3389/fonc.2014.00106] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 04/28/2014] [Indexed: 12/21/2022] Open
Abstract
Estrogen is necessary for the normal growth and development of breast tissue, but high levels of estrogen are a major risk factor for breast cancer. One mechanism by which estrogen could contribute to breast cancer is via the induction of DNA damage. This perspective discusses the mechanisms by which estrogen alters the DNA damage response (DDR) and DNA repair through the regulation of key effector proteins including ATM, ATR, CHK1, BRCA1, and p53 and the feedback on estrogen receptor signaling from these proteins. We put forward the hypothesis that estrogen receptor signaling converges to suppress effective DNA repair and apoptosis in favor of proliferation. This is important in hormone-dependent breast cancer as it will affect processing of estrogen-induced DNA damage, as well as other genotoxic insults. DDR and DNA repair proteins are frequently mutated or altered in estrogen responsive breast cancer, which will further change the processing of DNA damage. Finally, the action of estrogen signaling on DNA damage is also relevant to the therapeutic setting as the suppression of a DDR by estrogen has the potential to alter the response of cancers to anti-hormone treatment or chemotherapy that induces DNA damage.
Collapse
Affiliation(s)
- C Elizabeth Caldon
- Genome and Replication Stability Group, The Kinghorn Cancer Centre, Garvan Institute of Medical Research , Sydney, NSW , Australia ; St Vincent's Clinical School, Faculty of Medicine, UNSW Australia , Sydney, NSW , Australia
| |
Collapse
|
46
|
Liu Q, Turner KM, Alfred Yung WK, Chen K, Zhang W. Role of AKT signaling in DNA repair and clinical response to cancer therapy. Neuro Oncol 2014; 16:1313-23. [PMID: 24811392 DOI: 10.1093/neuonc/nou058] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Effective cancer treatment has been limited by the emergence of resistant cancer cells. The results of many studies indicate that AKT activation plays an important role in the acquisition of resistance to anticancer therapy. AKT is a critical effector serine/threonine kinase in the receptor tyrosine kinase/phosphatase and tensin homolog/phospho-inositide 3-kinase pathway and controls a myriad of cellular functions. Activation of AKT not only supports tumor growth and progression but also contributes to tumor-cell evasion of the cytotoxic effects of cancer therapy through many avenues including the promotion of anti-apoptosis, proliferation, and migration and regulation of the cell cycle. Accumulating evidence has implicated AKT as a direct participant in the DNA damage response and repair induced by commonly used genotoxic agents. In this review, we discuss the molecular mechanisms by which genotoxic agents activate AKT and therefore contribute to resistance to cancer therapeutics, with particular emphasis on DNA repair.
Collapse
Affiliation(s)
- Qun Liu
- Department of Neuro-Oncology, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China (Q.L.); Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas (K.M.T., W.Z.); Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas (W.K.A.Y.); Department of Epidemiology and Biostatistics, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China (K.C.)
| | - Kristen M Turner
- Department of Neuro-Oncology, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China (Q.L.); Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas (K.M.T., W.Z.); Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas (W.K.A.Y.); Department of Epidemiology and Biostatistics, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China (K.C.)
| | - W K Alfred Yung
- Department of Neuro-Oncology, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China (Q.L.); Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas (K.M.T., W.Z.); Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas (W.K.A.Y.); Department of Epidemiology and Biostatistics, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China (K.C.)
| | - Kexin Chen
- Department of Neuro-Oncology, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China (Q.L.); Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas (K.M.T., W.Z.); Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas (W.K.A.Y.); Department of Epidemiology and Biostatistics, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China (K.C.)
| | - Wei Zhang
- Department of Neuro-Oncology, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China (Q.L.); Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas (K.M.T., W.Z.); Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas (W.K.A.Y.); Department of Epidemiology and Biostatistics, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China (K.C.)
| |
Collapse
|
47
|
Levin ER. Translating extranuclear steroid receptor signaling to clinical medicine. Discov Oncol 2014; 5:140-5. [PMID: 24752388 DOI: 10.1007/s12672-014-0179-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 04/07/2014] [Indexed: 12/20/2022] Open
Abstract
The existence and function of extranuclear steroid receptors (SR) to rapidly modulate signal transduction is now acknowledged as present in cells and organs throughout the body. Work over the past 15 years has defined key mechanisms that are required for sex steroid receptors to traffic to the plasma membrane, but mechanisms of localization in other cell organelles such as mitochondria is still unclear. Signaling by membrane-localized SR has now been reported to impact many aspects of adult organ functions, while the roles in organ development are under investigation. In hormone-responsive cancers, both extranuclear and nuclear sex steroid receptors appear to collaborate in the regulation of some key genes that promote malignancy. Here, I review what is understood about the impact of extranuclear steroid receptor signaling to mitigate or promote disease processes.
Collapse
Affiliation(s)
- Ellis R Levin
- Division of Endocrinology, Departments of Medicine, University of California, Irvine, CA, 92717, USA,
| |
Collapse
|
48
|
Regulation of estrogen receptor signaling in breast carcinogenesis and breast cancer therapy. Cell Mol Life Sci 2014; 71:1549. [PMID: 25031550 PMCID: PMC3962223 DOI: 10.1007/s00018-013-1376-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 05/15/2013] [Accepted: 05/16/2013] [Indexed: 12/19/2022]
Abstract
Estrogen and estrogen receptors (ERs) are critical regulators of breast epithelial cell proliferation, differentiation, and apoptosis. Compromised signaling vis-à-vis the estrogen receptor is believed to be a major contributing factor in the malignancy of breast cells. Targeting the ER signaling pathway has been a focal point in the development of breast cancer therapy. Although approximately 75 % of breast cancer patients are classified as luminal type (ER(+)), which predicts for response to endocrine-based therapy; however, innate or acquired resistance to endocrine-based drugs remains a serious challenge. The complexity of regulation for estrogen signaling coupled with the crosstalk of other oncogenic signaling pathways is a reason for endocrine therapy resistance. Alternative strategies that target novel molecular mechanisms are necessary to overcome this current and urgent gap in therapy. A thorough analysis of estrogen-signaling regulation is critical. In this review article, we will summarize current insights into the regulation of estrogen signaling as related to breast carcinogenesis and breast cancer therapy.
Collapse
|
49
|
Savage KI, Matchett KB, Barros EM, Cooper KM, Irwin GW, Gorski JJ, Orr KS, Vohhodina J, Kavanagh JN, Madden AF, Powell A, Manti L, McDade SS, Park BH, Prise KM, McIntosh SA, Salto-Tellez M, Richard DJ, Elliott CT, Harkin DP. BRCA1 deficiency exacerbates estrogen-induced DNA damage and genomic instability. Cancer Res 2014; 74:2773-2784. [PMID: 24638981 DOI: 10.1158/0008-5472.can-13-2611] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Germline mutations in BRCA1 predispose carriers to a high incidence of breast and ovarian cancers. BRCA1 functions to maintain genomic stability through critical roles in DNA repair, cell-cycle arrest, and transcriptional control. A major question has been why BRCA1 loss or mutation leads to tumors mainly in estrogen-regulated tissues, given that BRCA1 has essential functions in all cell types. Here, we report that estrogen and estrogen metabolites can cause DNA double-strand breaks (DSB) in estrogen receptor-α-negative breast cells and that BRCA1 is required to repair these DSBs to prevent metabolite-induced genomic instability. We found that BRCA1 also regulates estrogen metabolism and metabolite-mediated DNA damage by repressing the transcription of estrogen-metabolizing enzymes, such as CYP1A1, in breast cells. Finally, we used a knock-in human cell model with a heterozygous BRCA1 pathogenic mutation to show how BRCA1 haploinsufficiency affects these processes. Our findings provide pivotal new insights into why BRCA1 mutation drives the formation of tumors in estrogen-regulated tissues, despite the general role of BRCA1 in DNA repair in all cell types.
Collapse
Affiliation(s)
- Kienan I Savage
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Rd, Belfast BT9 7BL, UK
| | - Kyle B Matchett
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Rd, Belfast BT9 7BL, UK
| | - Eliana M Barros
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Rd, Belfast BT9 7BL, UK
| | - Kevin M Cooper
- Institute for Global Food Security, Queen's University Belfast, 30 Malone Rd, Belfast BT9 5BN, UK
| | - Gareth W Irwin
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Rd, Belfast BT9 7BL, UK
| | - Julia J Gorski
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Rd, Belfast BT9 7BL, UK
| | - Katy S Orr
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Rd, Belfast BT9 7BL, UK
| | - Jekaterina Vohhodina
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Rd, Belfast BT9 7BL, UK
| | - Joy N Kavanagh
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Rd, Belfast BT9 7BL, UK
| | - Angelina F Madden
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Rd, Belfast BT9 7BL, UK
| | - Alexander Powell
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Rd, Belfast BT9 7BL, UK.,Institute for Global Food Security, Queen's University Belfast, 30 Malone Rd, Belfast BT9 5BN, UK
| | - Lorenzo Manti
- Radiation Biophysics Laboratory, Department of Physics, University of Naples Federico II, Via Cinthia-80126 Naples, Italy
| | - Simon S McDade
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Rd, Belfast BT9 7BL, UK
| | - Ben Ho Park
- The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Kevin M Prise
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Rd, Belfast BT9 7BL, UK
| | - Stuart A McIntosh
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Rd, Belfast BT9 7BL, UK
| | - Manuel Salto-Tellez
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Rd, Belfast BT9 7BL, UK
| | - Derek J Richard
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, 4059, Brisbane, Australia
| | - Christopher T Elliott
- Institute for Global Food Security, Queen's University Belfast, 30 Malone Rd, Belfast BT9 5BN, UK
| | - D Paul Harkin
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Rd, Belfast BT9 7BL, UK
| |
Collapse
|
50
|
The effect of tributyltin chloride on Caenorhabditis elegans germline is mediated by a conserved DNA damage checkpoint pathway. Toxicol Lett 2014; 225:413-21. [DOI: 10.1016/j.toxlet.2014.01.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 01/03/2014] [Accepted: 01/07/2014] [Indexed: 11/22/2022]
|