1
|
Ali K, Mishra P, Kumar A, Reddy DN, Chowdhury S, Panda G. Reactivity vs. selectivity of quinone methides: synthesis of pharmaceutically important molecules, toxicity and biological applications. Chem Commun (Camb) 2022; 58:6160-6175. [PMID: 35522910 DOI: 10.1039/d2cc00838f] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Quinone methides (QMs) are considered to be highly reactive intermediates because of their aromatization both in chemical and biological systems. Being highly accessible, quinone methides (QMs) have been widely exploited and their concurrent use has been manifested for the synthesis of tertiary and quaternary carbon centers of bioactives, drugs and drug-like molecules. In this feature article, the synthetic routes, structure-reactivity relationships and synthetic applications of quinone methides are discussed. Formation of the intermediates during bioactivation of different chemical entities and possible chemical manifestations leading to their toxicity in biological systems are also covered.
Collapse
Affiliation(s)
- Kasim Ali
- Medicinal & Process Chemistry Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Sector 10, Lucknow 226031, UP, India. .,Academy of Scientific & Industrial Research (AcSIR), Ghaziabad, Uttar Pradesh-201 002, India
| | - Prajjval Mishra
- Medicinal & Process Chemistry Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Sector 10, Lucknow 226031, UP, India.
| | - Awnish Kumar
- Medicinal & Process Chemistry Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Sector 10, Lucknow 226031, UP, India.
| | - Damodara N Reddy
- Medicinal & Process Chemistry Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Sector 10, Lucknow 226031, UP, India. .,Academy of Scientific & Industrial Research (AcSIR), Ghaziabad, Uttar Pradesh-201 002, India
| | - Sushobhan Chowdhury
- Medicinal & Process Chemistry Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Sector 10, Lucknow 226031, UP, India.
| | - Gautam Panda
- Medicinal & Process Chemistry Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Sector 10, Lucknow 226031, UP, India. .,Academy of Scientific & Industrial Research (AcSIR), Ghaziabad, Uttar Pradesh-201 002, India
| |
Collapse
|
2
|
Zhou W, Jiang Y, Xu Y, Wang Y, Ma X, Zhou L, Lin Y, Wang Y, Wu Z, Li M, Yin W, Lu J. Comparison of adverse drug reactions between tamoxifen and toremifene in breast cancer patients with different CYP2D6 genotypes: a propensity-score matched cohort study. Int J Cancer 2021; 150:1664-1676. [PMID: 34957551 DOI: 10.1002/ijc.33919] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 11/18/2021] [Accepted: 12/10/2021] [Indexed: 11/06/2022]
Abstract
CYP2D6 gene polymorphism had a profound impact upon the effect of tamoxifen as adjuvant endocrine therapy in breast cancers. However, it had never been reported whether the adverse drug reactions vary by CYP2D6 metabolic status for patients treated with tamoxifen or toremifene. We conducted an retrospective study in breast cancer patients to investigate the impact of CYP2D6 metabolizers on liver dysfunction events, gynecological events, and dyslipidemia events. According to CYP2D6*10 (100C → T) genotype, the enrolled patients were further categorized into four cohorts (extensive metabolizers taking tamoxifen [EM + TAM], extensive metabolizers taking toremifene [EM + TOR], intermediate metabolizers taking tamoxifen [IM + TAM], intermediate metabolizers taking toremifene cohort [IM + TOR]). A total of 192 patients were included into the study, with a median follow-up time of 26.2 months. In EM + TAM cohort, the risks of liver dysfunction events (P = 0.004) and gynecological events (P = 0.004) were significantly higher compared with EM + TOR cohort. In IM + TAM cohort, the risks of liver dysfunction events (P = 0.14) and gynecological events (P = 0.99) were not significantly different from IM + TOR cohort. Significant decrease of total cholesterol was observed in EM + TAM cohort around 1 year after taking tamoxifen (P < 0.001). Significant interactions between CYP2D6 metabolic status and endocrine agents were observed in terms of liver dysfunction events (p-interaction = 0.007) and gynecological events (p-interaction = 0.026). These findings suggested that CYP2D6 gene polymorphism played a significant role in predicting liver dysfunction, gynecological diseases and lipid metabolism changes among patients taking tamoxifen or toremifene. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Weihang Zhou
- Department of Breast Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yiwei Jiang
- Department of Breast Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yaqian Xu
- Department of Breast Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yaohui Wang
- Department of Breast Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaowei Ma
- Department of Clinical Laboratory, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Liheng Zhou
- Department of Breast Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yanping Lin
- Department of Breast Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yan Wang
- Department of Breast Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ziping Wu
- Department of Breast Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Min Li
- Department of Clinical Laboratory, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wenjin Yin
- Department of Breast Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jinsong Lu
- Department of Breast Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
3
|
E Owumi S, K Olusola J, O Arunsi U, K Oyelere A. Chlorogenic acid abates oxido-inflammatory and apoptotic responses in the liver and kidney of Tamoxifen-treated rats. Toxicol Res (Camb) 2021; 10:345-353. [PMID: 33884184 PMCID: PMC8045591 DOI: 10.1093/toxres/tfab002] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/18/2020] [Accepted: 01/08/2021] [Indexed: 12/12/2022] Open
Abstract
Plant-derived phenolics are utilized as chemopreventive agents to abate adverse toxic responses associated with drug-induced damages. Tamoxifen (TAM)-a chemotherapeutic agent-is used in managing all stages of hormone-dependent breast cancer. Notwithstanding TAM's clinical side effect-including hepatic toxicity-its use is commonplace. The present study investigates the effect of Chlorogenic acid (CGA: 25 and 50 mg kg-1; per os (p.o)) reported to exhibit various beneficial properties, including antioxidative effect against TAM (50 mg/kg; p.o.)-induced hepatorenal toxicities in rats treated as follows: Control, CGA, or TAM alone, and rats co-treated with CGA and TAM for 2 weeks. Biomarkers of hepatorenal function, oxido-inflammatory stress, and hepatorenal histopathology were performed. We observed that TAM alone decreased relative organ weights (ROW), marginally impacted rat's survivability, and significantly (P < 0.05) increased hepatorenal toxicities and reactive oxygen and nitrogen species (RONS). TAM decreased (P < 0.05) antioxidant, anti-inflammatory cytokine (IL-10), besides increase in (P < 0.05) lipid peroxidation (LPO), pro-inflammatory cytokines (IL-1β, TNF-α), nitric oxide (NO), xanthine oxidase (XO), myeloperoxidase (MPO), and apoptotic caspases (Casp-3 and -9) levels. These biochemical alterations were accompanied by morphological lesions in experimental rats' liver and kidney. Conversely, that CGA dose-dependently relieved TAM-mediated toxic responses, restored antioxidants capacities, reduced oxidative stress, pro-inflammatory cytokines levels, and Casp-3 and -9 activities in experimental rats. Furthermore, CGA protected against lesions observed in the liver and kidney of rats treated with TAM alone. Overall, CGA blocked TAM-mediated hepatorenal injuries associated with pro-oxidative, inflammatory, and apoptotic mechanisms. CGA may serve as a chemoprotective agent boosting patients prognosis undergoing TAM chemotherapy.
Collapse
Affiliation(s)
- Solomon E Owumi
- Cancer Research and Molecular Biology Laboratories, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan 200004, Nigeria
| | - Joseph K Olusola
- Cancer Research and Molecular Biology Laboratories, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan 200004, Nigeria
| | - Uche O Arunsi
- Department of Cancer Immunology and Biotechnology, School of Medicine, University of Nottingham, NG7 2RD, UK
| | - Adegboyega K Oyelere
- School of Chemistry & Biochemistry, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332-0400, USA
| |
Collapse
|
4
|
Owumi SE, Anaikor RA, Arunsi UO, Adaramoye OA, Oyelere AK. Chlorogenic acid co-administration abates tamoxifen-mediated reproductive toxicities in male rats: An experimental approach. J Food Biochem 2021; 45:e13615. [PMID: 33491243 DOI: 10.1111/jfbc.13615] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/18/2020] [Accepted: 01/04/2021] [Indexed: 12/16/2022]
Abstract
Reports over the years have demonstrated toxic side effect-including reproductive toxicity- of tamoxifen (TAM), a drug of choice in the management of primary breast cancer. Chlorogenic acid (CGA), a dietary polyphenol, reportedly elicits beneficial pharmacological effects. However, the impact of CGA on TAM-associated reproductive toxicity is absent in the literature. We, therefore, experimented on CGA's effect and TAM-mediated reproductive toxicity in rats. Cohorts of rats were treated with TAM (50 mg/kg) or co-treated with CGA (25 or 50 mg/kg) for 14 consecutive days. The result showed that treatment of CGA significantly increases testosterone, LH, and FSH levels compared to the TAM group. However, prolactin level was markedly decreased after pretreatment of CGA in TAM-treated rats. CGA abated TAM-induced decreases acid phosphatase, alkaline phosphatase, and antioxidant enzymes in the testis. CGA alleviated TAM-facilitated surges of reactive oxygen and nitrogen species, myeloperoxidase, nitric oxide, interleukin-1β, and tumor necrosis factor-alpha in rats epididymis and testes. Additionally, CGA increased anti-inflammatory cytokine -interleukin-10-, suppressed caspase-3 activity, and reduced pathological lesions in the examined organs of rats co-treated with CGA and TAM. CGA phytoprotective effect improved reproductive function occasioned by TAM-mediated toxicities in rats, by abating oxido-inflammatory damages and downregulating apoptotic responses. PRACTICAL APPLICATIONS: CGA protects against the damaging oxido-inflammatory responses incumbent on TAM metabolism. As an antioxidant abundant in plant-derived foods, CGA reportedly protects against inflammatory damage, hypertension, and neurodegenerative diseases. We present evidence that CGA ameliorates TAM-induced reproductive dysfunction by suppressing oxidative and inflammation stress downregulate apoptosis and improve reproductive function biomarker in rats.
Collapse
Affiliation(s)
- Solomon E Owumi
- Cancer Research and Molecular Biology Laboratories, Biochemistry Department, Faculty of Basic Medical Sciences, University of Ibadan, Ibadan, Nigeria
| | - Ruth A Anaikor
- Cancer Research and Molecular Biology Laboratories, Biochemistry Department, Faculty of Basic Medical Sciences, University of Ibadan, Ibadan, Nigeria
| | - Uche O Arunsi
- Cancer Immunology and Biotechnology Center, The University of Nottingham, Nottingham, UK
| | - Oluwatosin A Adaramoye
- Molecular Drug Metabolism and Toxicology Research Laboratories, Biochemistry Department, Faculty of Basic Medical Sciences, University of Ibadan, Ibadan, Nigeria
| | - Adegboyega K Oyelere
- School of Chemistry & Biochemistry, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
5
|
Wang L, Sharma A. The Quest for Orally Available Selective Estrogen Receptor Degraders (SERDs). ChemMedChem 2020; 15:2072-2097. [PMID: 32916035 DOI: 10.1002/cmdc.202000473] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/31/2020] [Indexed: 01/10/2023]
Abstract
Estrogen receptor-alpha (ERα) is the target of endocrine therapies for the treatment of more than 70 % of ERα-positive breast cancers. Selective estrogen receptor degraders (SERDs) antagonize estrogen binding and target the receptor for degradation, representing the last line of treatment for resistant metastatic breast cancer patients. However, the clinical efficacy of the lone clinically approved SERD (Fulvestrant) is limited by its poor oral bioavailability. Recently, several analogues of GW5638, an acrylic acid-based ERα ligand developed by Glaxo Research Institute in 1994, have been reported as promising orally bioavailable SERDs. Some of these compounds are currently in clinical trials, while various other structurally novel SERDs have also been reported by pharma as well as academic research groups. This review provides a critical analysis of the recent developments in orally available SERDs, with a focus on the structure-activity relationships, binding interactions and pharmacokinetic properties of these compounds.
Collapse
Affiliation(s)
- Lucia Wang
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, New Jersey, 07030, USA
| | - Abhishek Sharma
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, New Jersey, 07030, USA
| |
Collapse
|
6
|
Sanchez-Spitman A, Swen J, Dezentje V, Moes D, Gelderblom H, Guchelaar H. Clinical pharmacokinetics and pharmacogenetics of tamoxifen and endoxifen. Expert Rev Clin Pharmacol 2019; 12:523-536. [DOI: 10.1080/17512433.2019.1610390] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- A.B. Sanchez-Spitman
- Leiden Network for Personalised Therapeutics, Leiden University Medical Center, Leiden, The Netherlands
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - J.J. Swen
- Leiden Network for Personalised Therapeutics, Leiden University Medical Center, Leiden, The Netherlands
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - V.O. Dezentje
- Department of Medical Oncology, Netherlands Cancer Institute/Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - D.J.A.R. Moes
- Leiden Network for Personalised Therapeutics, Leiden University Medical Center, Leiden, The Netherlands
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - H. Gelderblom
- Leiden Network for Personalised Therapeutics, Leiden University Medical Center, Leiden, The Netherlands
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - H.J. Guchelaar
- Leiden Network for Personalised Therapeutics, Leiden University Medical Center, Leiden, The Netherlands
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
7
|
Okamoto Y, Shibutani S. Development of novel and safer anti-breast cancer agents, SS1020 and SS5020, based on a fundamental carcinogenic research. Genes Environ 2019; 41:9. [PMID: 30976361 PMCID: PMC6437986 DOI: 10.1186/s41021-019-0124-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 03/11/2019] [Indexed: 12/24/2022] Open
Abstract
Tamoxifen (TAM) has been prescribed worldwide to patients with and women at high-risk of breast cancer. However, long-term use of TAM increases the incidence of endometrial cancer. The carcinogenic mechanisms of TAM have been extensively investigated. TAM is hydroxylated and sulfonated at α-carbon to form α-hydroxytamoxifen-O-sulfonate. This metabolite readily reacts with genomic DNA, particularly with 2′-deoxyguanosine, leading to DNA replication error. TAM also exerts estrogenic activity at endometrial tissue to induce endometrial hyperplasia. Therefore, our efforts focused on the development of novel and safer anti-estrogens to diminish carcinogenic potential of TAM based on chemical modifications. In this review, we describe a crucial idea of our drug design and introduce our compounds SS1020 and SS5020, possessing high effectiveness, and no genotoxic and estrogenic activities.
Collapse
Affiliation(s)
- Yoshinori Okamoto
- 1Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya, 468-8503 Japan
| | - Shinya Shibutani
- 2Department of Pharmacological Sciences, State University of New York at Stony Brook, Stony Brook, 11794-8651 New York USA
| |
Collapse
|
8
|
Bremer S, Brittebo E, Dencker L, Knudsen LE, Mathisien L, Olovsson M, Pazos P, Pellizzer C, Paulesu LR, Schaefer W, Schwarz M, Staud F, Stavreus-Evers A, Vähänkangas K. In Vitro Tests for Detecting Chemicals Affecting the Embryo Implantation Process. Altern Lab Anim 2019; 35:421-39. [PMID: 17850188 DOI: 10.1177/026119290703500407] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Susanne Bremer
- ECVAM, Institute for Health and Consumer Protection, European Commission Joint Research Centre, Ispra, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Filardo EJ. A role for G-protein coupled estrogen receptor (GPER) in estrogen-induced carcinogenesis: Dysregulated glandular homeostasis, survival and metastasis. J Steroid Biochem Mol Biol 2018; 176:38-48. [PMID: 28595943 DOI: 10.1016/j.jsbmb.2017.05.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 05/09/2017] [Accepted: 05/16/2017] [Indexed: 12/18/2022]
Abstract
Mechanisms of carcinogenesis by estrogen center on its mitogenic and genotoxic potential on tumor target cells. These models suggest that estrogen receptor (ER) signaling promotes expansion of the transformed population and that subsequent accumulation of somatic mutations that drive cancer progression occur via metabolic activation of cathecol estrogens or by epigenetic mechanisms. Recent findings that GPER is linked to obesity, vascular pathology and immunosuppression, key events in the development of metabolic syndrome and intra-tissular estrogen synthesis, provides an alternate view of estrogen-induced carcinogenesis. Consistent with this concept, GPER is directly associated with clinicopathological indices that predict cancer progression and poor survival in breast and gynecological cancers. Moreover, GPER manifests cell biological responses and a microenvironment conducive for tumor development and cancer progression, regulating cellular responses associated with glandular homeostasis and survival, invading surrounding tissue and attracting a vascular supply. Thus, the cellular actions attributed to GPER fit well with the known molecular mechanisms of G-protein coupled receptors, GPCRs, namely, their ability to transactivate integrins and EGF receptors and alter the interaction between glandular epithelia and their extracellular environment, affecting epithelial-to-mesenchymal transition (EMT) and allowing for tumor cell survival and dissemination. This perspective reviews the molecular and cellular responses manifested by GPER and evaluates its contribution to female reproductive cancers as diseases that progress as a result of dysregulated glandular homeostasis resulting in chronic inflammation and metastasis. This review is organized in sections as follows: I) a brief synopsis of the current state of knowledge regarding estrogen-induced carcinogenesis, II) a review of evidence from clinical and animal-based studies that support a role for GPER in cancer progression, and III) a mechanistic framework describing how GPER-mediated estrogen action may influence the tumor and its microenvironment.
Collapse
Affiliation(s)
- Edward J Filardo
- Division of Hematology & Oncology, The Warren Alpert School of Medicine, Brown University, Providence, RI 02818, United States.
| |
Collapse
|
10
|
Synergistic anticancer effects of a bioactive subfraction of Strobilanthes crispus and tamoxifen on MCF-7 and MDA-MB-231 human breast cancer cell lines. Altern Ther Health Med 2014; 14:252. [PMID: 25034326 PMCID: PMC4223515 DOI: 10.1186/1472-6882-14-252] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2014] [Accepted: 07/11/2014] [Indexed: 12/21/2022]
Abstract
Background Development of tumour resistance to chemotherapeutic drugs and concerns over their toxic effects has led to the increased use of medicinal herbs or natural products by cancer patients. Strobilanthes crispus is a traditional remedy for many ailments including cancer. Its purported anticancer effects have led to the commercialization of the plant leaves as medicinal herbal tea, although the scientific basis for its use has not been established. We previously reported that a bioactive subfraction of Strobilanthes crispus leaves (SCS) exhibit potent cytotoxic activity against human breast cancer cell lines. The current study investigates the effect of this subfraction on cell death activities induced by the antiestrogen drug, tamoxifen, in estrogen receptor-responsive and nonresponsive breast cancer cells. Methods Cytotoxic activity of SCS and tamoxifen in MCF-7 and MDA-MB-231 human breast cancer cells was determined using lactate dehydrogenase release assay and synergism was evaluated using the CalcuSyn software. Apoptosis was quantified by flow cytometry following Annexin V and propidium iodide staining. Cells were also stained with JC-1 dye to determine changes in the mitochondrial membrane potential. Fluorescence imaging using FAM-FLICA assay detects caspase-8 and caspase-9 activities. DNA damage in the non-malignant breast epithelial cell line, MCF-10A, was evaluated using Comet assay. Results The combined SCS and tamoxifen treatment displayed strong synergistic inhibition of MCF-7 and MDA-MB-231 cell growth at low doses of the antiestrogen. SCS further promoted the tamoxifen-induced apoptosis that was associated with modulation of mitochondrial membrane potential and activation of caspase-8 and caspase-9, suggesting the involvement of intrinsic and extrinsic signaling pathways. Interestingly, the non-malignant MCF-10A cells displayed no cytotoxicity or DNA damage when treated with either SCS or SCS-tamoxifen combination. Conclusions The combined use of SCS and lower tamoxifen dose could potentially reduce the side effects/toxicity of the drug. However, further studies are needed to determine the effectiveness and safety of the combination treatment in vivo.
Collapse
|
11
|
Nagy E, Gajjar KB, Patel II, Taylor S, Martin-Hirsch PL, Stringfellow HF, Martin FL, Phillips DH. MGMT promoter hypermethylation and K-RAS, PTEN and TP53 mutations in tamoxifen-exposed and non-exposed endometrial cancer cases. Br J Cancer 2014; 110:2874-80. [PMID: 24853176 PMCID: PMC4056065 DOI: 10.1038/bjc.2014.263] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 04/23/2014] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Tamoxifen has anti-oestrogenic and anti-tumour activity in the breast, but is oestrogenic and carcinogenic in the endometrium. It can induce experimental tumours by both hormonal and DNA-damaging mechanisms, but its carcinogenic mode of action in human endometrium remains unclear. METHODS We investigated whether an epigenetic mechanism, involving promoter hypermethylation of the gene for the DNA repair enzyme MGMT (O6-methylguanine DNA methyltransferase), was associated with K-RAS, TP53 and PTEN mutations in endometrial tumours from women treated with tamoxifen (TAM, n=30) or unexposed to the drug (EC, n=38). RESULTS There were significant (P<0.05) differences in tumour grade between the TAM and EC groups, with more favourable morphology in the latter. K-RAS mutations, predominantly G>A, occurred in small numbers in both groups. TP53 mutations were of mainly A>G, C>T and indel modifications in both groups, but more frequent in TAM cases. PTEN mutations dominated in EC tumours and were of the type that has large impact on protein function, such as indel or nonsense mutations. These observations alongside the mutational spectrum in PTEN suggest that the malignancies arise from different backgrounds, hence pointing to an effect of tamoxifen. Both groups displayed MGMT promoter hypermethylation. This coincided with mutations more frequently in the TAM (78%) than in the EC (50%) group, even though there were significantly (P<0.05) fewer mutations and methylations in TAM cases. CONCLUSIONS Although the difference in coincidence did not reach significance with the current sample size, the findings suggest that epigenetic processes may play a role in the way tamoxifen induces endometrial cancer.
Collapse
Affiliation(s)
- E Nagy
- Analytical and Environmental Sciences Division, King's College London, London SE1 9NH, UK
| | - K B Gajjar
- Centre for Biophotonics, Lancaster Environment Centre, Lancaster University, Lancaster LA1 4YQ, UK
| | - I I Patel
- Cavendish Laboratories, JJ Thomson Avenue, University of Cambridge, Cambridge CB3 0HE, UK
| | - S Taylor
- Liverpool Women's Hospital NHS Foundation Trust, Department of Gynaecology, Crown Street, Liverpool, Merseyside L8 7SS, UK
| | - P L Martin-Hirsch
- Centre for Biophotonics, Lancaster Environment Centre, Lancaster University, Lancaster LA1 4YQ, UK
- Lancashire Teaching Hospitals NHS Trust, Royal Preston Hospital, Sharoe Green Lane North, Preston, Lancashire PR2 9HT, UK
| | - H F Stringfellow
- Lancashire Teaching Hospitals NHS Trust, Royal Preston Hospital, Sharoe Green Lane North, Preston, Lancashire PR2 9HT, UK
| | - F L Martin
- Centre for Biophotonics, Lancaster Environment Centre, Lancaster University, Lancaster LA1 4YQ, UK
| | - D H Phillips
- Analytical and Environmental Sciences Division, King's College London, London SE1 9NH, UK
| |
Collapse
|
12
|
Yaacob NS, Ismail NF. Comparison of cytotoxicity and genotoxicity of 4-hydroxytamoxifen in combination with Tualang honey in MCF-7 and MCF-10A cells. Altern Ther Health Med 2014; 14:106. [PMID: 24646375 PMCID: PMC3994783 DOI: 10.1186/1472-6882-14-106] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2013] [Accepted: 03/12/2014] [Indexed: 02/07/2023]
Abstract
Background The Malaysian Tualang honey (TH) is not only cytotoxic to human breast cancer cell lines but it has recently been reported to promote the anticancer activity induced by tamoxifen in MCF-7 and MDA-MB-231 cells suggesting its potential as an adjuvant for the chemotherapeutic agent. However, tamoxifen produces adverse effects that could be due to its ability to induce cellular DNA damage. Therefore, the study is undertaken to determine the possible modulation of the activity of 4-hydroxytamoxifen (OHT), an active metabolite of tamoxifen, by TH in non-cancerous epithelial cell line, MCF-10A, in comparison with MCF-7 cells. Methods MCF-7 and MCF-10A cells were treated with TH, OHT or the combination of both and cytotoxicity and antiproliferative activity were determined using LDH and MTT assays, respectively. The effect on cellular DNA integrity was analysed by comet assay and the expression of DNA repair enzymes was determined by Western blotting. Results OHT exposure was cytotoxic to both cell lines whereas TH was cytotoxic to MCF-7 cells only. TH also significantly decreased the cytotoxic effect of OHT in MCF-10A but not in MCF-7 cells. TH induced proliferation of MCF10A cells but OHT caused growth inhibition that was abrogated by the concomitant treatment with TH. While TH enhanced the OHT-induced DNA damage in the cancer cells, it dampened the genotoxic effect of OHT in the non-cancerous cells. This was supported by the increased expression of DNA repair proteins, Ku70 and Ku80, in MCF-10A cells by TH. Conclusion The findings indicate that TH could afford protection of non-cancerous cells from the toxic effects of tamoxifen by increasing the efficiency of DNA repair mechanism in these cells.
Collapse
|
13
|
Abstract
The formation of quinone methides (QMs) from either direct 2-electron oxidation of 2- or 4-alkylphenols, isomerization of o-quinones, or elimination of a good leaving group could explain the cytotoxic/cytoprotective effects of several drugs, natural products, as well as endogenous compounds. For example, the antiretroviral drug nevirapine and the antidiabetic agent troglitazone both induce idiosyncratic hepatotoxicity through mechanisms involving quinone methide formation. The anesthetic phencyclidine induces psychological side effects potentially through quinone methide mediated covalent modification of crucial macromolecules in the brain. Selective estrogen receptor modulators (SERMs) such as tamoxifen, toremifene, and raloxifene are metabolized to quinone methides which could potentially contribute to endometrial carcinogenic properties and/or induce detoxification enzymes and enhance the chemopreventive effects of these SERMs. Endogenous estrogens and/or estrogens present in estrogen replacement formulations are also metabolized to catechols and further oxidized to o-quinones which can isomerize to quinone methides. Both estrogen quinoids could cause DNA damage which could enhance hormone dependent cancer risk. Natural products such as the food and flavor agent eugenol can be directly oxidized to a quinone methide which may explain the toxic effects of this natural compound. Oral toxicities associated with chewing areca quid could be the result of exposure to hydroxychavicol through initial oxidation to an o-quinone which isomerizes to a p-quinone methide. Similar o-quinone to p-quinone methide isomerization reactions have been reported for the ubiquitous flavonoid quercetin which needs to be taken into consideration when evaluating risk-benefit assessments of these natural products. The resulting reaction of these quinone methides with proteins, DNA, and/or resulting modulation of gene expression may explain the toxic and/or beneficial effects of the parent compounds.
Collapse
Affiliation(s)
- Judy L. Bolton
- Department of Medicinal Chemistry and Pharmacognosy (M/C 781) College of Pharmacy University of Illinois at Chicago 833 S. Wood Street Chicago, Illinois 60612-7231
| |
Collapse
|
14
|
Tualang honey promotes apoptotic cell death induced by tamoxifen in breast cancer cell lines. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:989841. [PMID: 23476711 PMCID: PMC3586458 DOI: 10.1155/2013/989841] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Accepted: 01/07/2013] [Indexed: 12/31/2022]
Abstract
Tualang honey (TH) is rich in flavonoids and phenolic acids and has significant anticancer activity against breast cancer cells comparable to the effect of tamoxifen (TAM), in vitro. The current study evaluated the effects of TH when used in combination with TAM on MCF-7 and MDA-MB-231 cells. We observed that TH promoted the anticancer activity of TAM in both the estrogen receptor-(ER-)responsive and ER-nonresponsive human breast cancer cell lines. Flow cytometric analyses indicated accelerated apoptosis especially in MDA-MB-231 cells and with the involvement of caspase-3/7, -8 and -9 activation as shown by fluorescence microscopy. Depolarization of the mitochondrial membrane was also increased in both cell lines when TH was used in combination with TAM compared to TAM treatment alone. TH may therefore be a potential adjuvant to be used with TAM for reducing the dose of TAM, hence, reducing TAM-induced adverse effects.
Collapse
|
15
|
Wexselblatt E, Gibson D. What do we know about the reduction of Pt(IV) pro-drugs? J Inorg Biochem 2012; 117:220-9. [DOI: 10.1016/j.jinorgbio.2012.06.013] [Citation(s) in RCA: 236] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Revised: 06/23/2012] [Accepted: 06/25/2012] [Indexed: 10/28/2022]
|
16
|
CYP1B1 and hormone-induced cancer. Cancer Lett 2012; 324:13-30. [DOI: 10.1016/j.canlet.2012.04.021] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2012] [Revised: 04/21/2012] [Accepted: 04/24/2012] [Indexed: 12/11/2022]
|
17
|
Cyril V, Muller MT. A solid phase assay for topoisomerase I interfacial poisons and catalytic inhibitors. Anal Biochem 2012; 421:607-16. [DOI: 10.1016/j.ab.2011.12.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Revised: 12/05/2011] [Accepted: 12/08/2011] [Indexed: 10/14/2022]
|
18
|
Williams-Brown MY, Salih SM, Xu X, Veenstra TD, Saeed M, Theiler SK, Diaz-Arrastia CR, Salama SA. The effect of tamoxifen and raloxifene on estrogen metabolism and endometrial cancer risk. J Steroid Biochem Mol Biol 2011; 126:78-86. [PMID: 21600284 PMCID: PMC3421458 DOI: 10.1016/j.jsbmb.2011.05.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Revised: 03/22/2011] [Accepted: 05/03/2011] [Indexed: 11/18/2022]
Abstract
Selective estrogen receptor modulators (SERMs) demonstrate differential endometrial cancer (EC) risk. While tamoxifen (TAM) use increases the risk of endometrial hyperplasia and malignancy, raloxifene (RAL) has neutral effects on the uterus. How TAM increases the risk of EC and why TAM and RAL differentially modulate the risk for EC, however, remain elusive. Here, we tested the hypothesis that TAM increases the risk for EC, at least in part, by enhancing the local estrogen biosynthesis and directing estrogen metabolism towards the formation of genotoxic and hormonally active estrogen metabolites. In addition, the differential effects of TAM and RAL in EC risk are attributed to their differential effect on estrogen metabolism/metabolites. The endometrial cancer cell line (Ishikawa cells) and the nonmalignant immortalized human endometrial glandular cell line (EM1) were used for the study. The profile of estrogen/estrogen metabolites (EM), depurinating estrogen-DNA adducts, and the expression of estrogen-metabolizing enzymes in cells treated with 17β-estradiol (E2) alone or in combination with TAM or RAL were investigated using high performance liquid chromatography-electrospray ionization-tandem mass spectrometry (HPLC-ESI-MS(2)), ultraperformance liquid chromatography/tandem mass spectrometry (UPLC-MS/MS), and Western blot analysis, respectively. TAM significantly increased the total EM and enhanced the formation of hormonally active and carcinogenic estrogen metabolites, 4-hydroxestrone (4-OHE1) and 16α-hydroxyestrone, with concomitant reduction in the formation of antiestrogenic and anticarcinogenic 2-hydroxyestradiol and 2-methoxyestradiol. Furthermore, TAM increased the formation of depurinating estrogen-DNA adducts 4-OHE1 [2]-1-N7Guanine and 4-OHE1 [2]-1-N3 Adenine. TAM-induced alteration in EM and depurinating DNA adduct formation is associated with altered expression of estrogen metabolizing enzymes CYP1A1, CYP1B1, COMT, NQO1, and SF-1 as revealed by Western blot analysis. In contrast to TAM, RAL has minimal effect on EM, estrogen-DNA adduct formation, or estrogen-metabolizing enzymes expression. These data show that TAM perturbs the balance of estrogen-metabolizing enzymes and alters the disposition of estrogen metabolites, which can explain, at least in part, the mechanism for TAM-induced EC. These results also implicate the differential effect of TAM and RAL on estrogen metabolism/metabolites as a potential mechanism for their disparate effects on the endometrium.
Collapse
Affiliation(s)
- Marian Y Williams-Brown
- Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, United States.
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Suzuki N, Liu X, Laxmi YRS, Okamoto K, Kim HJ, Zhang G, Chen JJ, Okamoto Y, Shibutani S. Anti-breast cancer potential of SS5020, a novel benzopyran antiestrogen. Int J Cancer 2011; 128:974-82. [PMID: 20824696 PMCID: PMC3011858 DOI: 10.1002/ijc.25659] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2010] [Accepted: 08/26/2010] [Indexed: 12/21/2022]
Abstract
Treatment with tamoxifen (TAM) increases the risk of developing endometrial cancer in women. The carcinogenic effect is thought to involve initiation and/or promotion resulting from DNA damage induced by TAM as well as its estrogenic action. To minimize this serious side-effect while increasing the anti-breast cancer potential, a new benzopyran antiestrogen, 2E-3-{4-[(7-hydroxy-2-oxo-3-phenyl-2H-chromen-4-yl)-methyl]-phenyl}-acrylic acid (SS5020), was synthesized. Unlike TAM, SS5020 exhibits no genotoxic activity to damage DNA. Furthermore, SS5020 does not present significant uterotrophic potential in rats; in contrast, the structurally related compounds, TAM, toremifene, raloxifene (RAL) and SP500263 all have uterotrophic activity. At the human equivalent molar dose of TAM (0.33 or 1.0 mg/kg), SS5020 had much stronger antitumor potential than those same antiestrogens against 7,12-dimethylbenz(a)anthracene-induced mammary carcinoma in rats. The growth of human MCF-7 breast cancer xenograft implanted into athymic nude mice was also effectively suppressed by SS5020. SS5020, lacking genotoxic and estrogenic actions, could be a safer and stronger antiestrogen alternative to TAM and RAL for breast cancer therapy and prevention.
Collapse
Affiliation(s)
- Naomi Suzuki
- Department of Pharmacological Sciences, State University of New York at Stony Brook, Stony Brook, New York 11794-8651
| | - Xiaoping Liu
- Department of Pharmacological Sciences, State University of New York at Stony Brook, Stony Brook, New York 11794-8651
| | - Y. R. Santosh Laxmi
- Department of Pharmacological Sciences, State University of New York at Stony Brook, Stony Brook, New York 11794-8651
| | - Kanako Okamoto
- Department of Pharmacological Sciences, State University of New York at Stony Brook, Stony Brook, New York 11794-8651
| | - Hyo Jeong Kim
- Department of Pharmacological Sciences, State University of New York at Stony Brook, Stony Brook, New York 11794-8651
| | - Guangxiang Zhang
- Department of Preventive Medicine, State University of New York at Stony Brook, Stony Brook, New York 11794-8651
| | - John J. Chen
- Department of Preventive Medicine, State University of New York at Stony Brook, Stony Brook, New York 11794-8651
| | - Yoshinori Okamoto
- Department of Pharmacological Sciences, State University of New York at Stony Brook, Stony Brook, New York 11794-8651
| | - Shinya Shibutani
- Department of Pharmacological Sciences, State University of New York at Stony Brook, Stony Brook, New York 11794-8651
| |
Collapse
|
20
|
Effects of tamoxifen on the endometrium and its mechanism of carcinogenicity. Hum Cell 2011; 24:65-73. [PMID: 21547354 DOI: 10.1007/s13577-010-0005-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2010] [Accepted: 10/22/2010] [Indexed: 10/18/2022]
Abstract
This study was conducted to clarify the clinicopathological characteristics of tamoxifen-associated endometrial carcinomas and its mechanisms of carcinogenesis. Seven patients with tamoxifen-associated endometrial carcinomas (TAM group) and 28 with sporadic endometrioid adenocarcinomas (EMC group) were included in the study. The clinicopathological factors, such as FIGO stage, histological type, grade, lymph node metastases, vascular invasion and the coexistence of hyperplasia, were investigated in both groups. The protein expression of p53, PTEN, hMLH1 and hMSH2 was investigated by immunohistochemistry. Microsatellite instability (MSI), k-ras and p53 mutation were also examined. In the TAM group, the histological types included five endometrioid, one endometrioid combined with serous and one clear cell type. The rates of coexistence with hyperplasia (five of seven cases) and vascular invasion (four cases) were significantly higher in the TAM group. The rates of stage III/IV (four cases) and lymph node metastasis (three cases) tended to be higher in the TAM group. Although there were no significant differences in PTEN, hMLH1 and hMSH2 expression between the two groups, p53 mutation was more frequent in three out of five cases (60%) in the TAM group compared with 2 of 15 cases in the EMC group (13.3%). No significant differences were observed concerning MSI and k-ras mutation in either group. These results suggested that TAM-associated endometrial carcinomas have overlapping biological characteristics of type I and type II endometrial carcinomas. This might explain the somewhat worse prognosis of these tumors than sporadic endometrioid carcinomas.
Collapse
|
21
|
Zhang Y, Saha S, Rosenfeld G, Gonzalez J, Pepeljugoski KP, Peeva E. Raloxifene modulates estrogen-mediated B cell autoreactivity in NZB/W F1 mice. J Rheumatol 2010; 37:1646-57. [PMID: 20551107 DOI: 10.3899/jrheum.090911] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Estrogen has been found to exacerbate disease activity in murine lupus and to induce a lupus-like syndrome in nonspontaneously autoimmune mice. This has led to the consideration that estrogen may be a risk factor for the development of systemic lupus erythematosus (SLE), and selective estrogen receptor modulators (SERM) may serve to ameliorate lupus activity. We evaluated the effects and mechanism of action of the SERM raloxifene in murine lupus. METHODS Effects of raloxifene on the development of lupus in NZB/W F1 mice were evaluated in the presence and absence of estrogen by assessing the serum DNA reactivity, glomerular IgG deposition and kidney damage, B cell maturation and selection, and activation status of marginal zone and follicular B cells. RESULTS Compared to estradiol-treated mice, mice treated with estradiol and raloxifene had significantly lower serum anti-DNA antibody levels and less kidney damage. These effects of raloxifene were due, at least in part, to antagonism of the influence of estrogen on DNA-reactive B cells. Raloxifene was found to prevent estrogen-mediated suppression of autoreactive B cell elimination at the T1/T2 selection checkpoint, to reduce estrogen-induced CD40 overexpression on follicular B cells, making them less responsive to T cell costimulation, and to ameliorate estrogen-mediated CD22 downregulation on marginal zone B cells, thereby decreasing their responsiveness to B cell antigen receptor-mediated stimuli. CONCLUSION Raloxifene suppressed estrogen-mediated effects on the survival, maturation, and activation of autoreactive B cells in NZB/W F1 mice.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Medicine and Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA.
| | | | | | | | | | | |
Collapse
|
22
|
Moore CD, Reilly CA, Yost GS. CYP3A4-Mediated oxygenation versus dehydrogenation of raloxifene. Biochemistry 2010; 49:4466-75. [PMID: 20405834 DOI: 10.1021/bi902213r] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Raloxifene was approved in 2007 by the FDA for the chemoprevention of breast cancer in postmenopausal women at high risk for invasive breast cancer. Approval was based in part on the improved safety profile for raloxifene relative to the standard treatment of tamoxifen. However, recent studies have demonstrated the ability of raloxifene to form reactive intermediates and act as a mechanism-based inhibitor of cytochrome P450 3A4 (CYP3A4) by forming adducts with the apoprotein. However, previous studies could not differentiate between dehydrogenation to a diquinone methide and the more common oxygenation pathway to an arene oxide as the most likely intermediate to inactivate CYP3A4. In the current work, (18)O-incorporation studies were utilized to carefully elucidate CYP3A4-mediated oxygenation versus dehydrogenation of raloxifene. These studies established that 3'-hydroxyraloxifene is produced exclusively via CYP3A4-mediated oxygenation and provide convincing evidence for the mechanism of CYP3A4-mediated dehydrogenation of raloxifene to a reactive diquinone methide, while excluding the alternative arene oxide pathway. Furthermore, it was demonstrated that 7-hydroxyraloxifene, which was previously believed to be a typical O(2)-derived metabolite of CYP3A4, is in fact produced by a highly unusual hydrolysis pathway from a putative ester, formed by the conjugation of raloxifene diquinone methide with a carboxylic acid moiety of CYP3A4, or other proteins in the reconstituted system. These findings not only confirm CYP3A4-mediated dehydrogenation of raloxifene to a reactive diquinone methide but also suggest a novel route of raloxifene toxicity.
Collapse
Affiliation(s)
- Chad D Moore
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah 84112, USA
| | | | | |
Collapse
|
23
|
Laxmi YS, Liu X, Suzuki N, Kim SY, Okamoto K, Kim HJ, Zhang G, Chen JJ, Okamoto Y, Shibutani S. Anti-breast cancer potential of SS1020, a novel antiestrogen lacking estrogenic and genotoxic actions. Int J Cancer 2010; 127:1718-26. [DOI: 10.1002/ijc.25167] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
24
|
Andersson H, Helmestam M, Zebrowska A, Olovsson M, Brittebo E. Tamoxifen-Induced Adduct Formation and Cell Stress in Human Endometrial Glands. Drug Metab Dispos 2009; 38:200-7. [DOI: 10.1124/dmd.109.029488] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
25
|
|
26
|
Okamoto Y, Liu X, Suzuki N, Okamoto K, Sekimoto M, Laxmi YS, Shibutani S. Increased antitumor potential of the raloxifene prodrug, raloxifene diphosphate. Int J Cancer 2008; 122:2142-7. [DOI: 10.1002/ijc.23362] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
27
|
Overk CR, Peng KW, Asghodom RT, Kastrati I, Lantvit DD, Qin Z, Frasor J, Bolton JL, Thatcher GRJ. Structure–Activity Relationships for a Family of Benzothiophene Selective Estrogen Receptor Modulators Including Raloxifene and Arzoxifene. ChemMedChem 2007; 2:1520-6. [PMID: 17654759 DOI: 10.1002/cmdc.200700104] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The search for the "ideal" selective estrogen receptor modulator (SERM) as a substitute for hormone replacement therapy (HRT) or use in cancer chemoprevention has focused on optimization of estrogen receptor (ER) ligand binding. Based on the clinical and preclinical benzothiophene SERMs, raloxifene and arzoxifene, a family of SERMs has been developed to modulate activity and oxidative lability. Antiestrogenic potency measured in human endometrial and breast cancer cells, and ER ligand binding data were correlated and seen to provide a guide to SERM design only when viewed in toto. The in vitro studies were extended to the juvenile rat model, in which the desired antiestrogenic profile and putative cardiovascular benefits of SERMs were observed.
Collapse
Affiliation(s)
- Cassia R Overk
- Department of Medicinal Chemistry & Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, 833 S Wood St, Chicago, IL 60612, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Matsumoto J, Iwano H, Inoue H, Iwano N, Yamashiki N, Yokota H. Metabolic Barrier against Bisphenol A in Rat Uterine Endometrium. Toxicol Sci 2007; 99:118-25. [PMID: 17567591 DOI: 10.1093/toxsci/kfm148] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Exposure to environmental chemicals with estrogenic activity during the early stage of pregnancy can seriously affect embryonic development and the maintenance of pregnancy. To estimate the metabolism and pharmacodynamics of a xenoestrogen, bisphenol A, in a reproductive organ, the metabolite of bisphenol A was analyzed after incubating a rat uterine sac in buffer solutions containing the chemical. When the inner or the outer side of the uterine sac was exposed to bisphenol A, the concentration of the parent chemical was decreased in buffer solution and then, only one metabolite, bisphenol A-glucuronide, was observed only in the outer, that is, the maternal, side. A small amount of the parent chemical could pass through the uterine sac without being modified. Uridine diphosphate (UDP)-glucuronosyltransferase (UGT) was shown by immunohistochemical staining analysis to be distributed in epithelial cells of the endometrium, oviduct, and uterine glands. Based on measurements of enzyme activity and on Western blot analysis, UGT activity toward bisphenol A and UGT protein were identified in the microsomal fractions prepared from rat uterus. UGT isoforms, such as UGT1A1, 1A2, 1A5, 1A6, and 1A7, were expressed, and MRP-1 (multidrug resistance-associated protein) and MRP-3, which are well-known to be transporters of various drug-glucuronides, were detected in the rat uterus by reverse transcription-PCR. These results elucidate the rat uterine barrier system by showing that most bisphenol A perfused into the uterus was glucuronidated in the epithelium, resulting in transport of glucuronides to the maternal side.
Collapse
Affiliation(s)
- Junya Matsumoto
- Department of Veterinary Biochemistry, School of Veterinary Medicine, Faculty of Environmental System, Rakuno Gakuen University, Hokkaido, Japan
| | | | | | | | | | | |
Collapse
|
29
|
Dowers TS, Qin ZH, Thatcher GRJ, Bolton JL. Bioactivation of Selective Estrogen Receptor Modulators (SERMs). Chem Res Toxicol 2006; 19:1125-37. [PMID: 16978016 PMCID: PMC2517576 DOI: 10.1021/tx060126v] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Tamara S Dowers
- Department of Medicinal Chemistry and Pharmacognosy (M/C 781), College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, Illinois 60612-7231, USA
| | | | | | | |
Collapse
|
30
|
Kim SY, Suzuki N, Santosh Laxmi YR, Umemoto A, Matsuda T, Shibutani S. Antiestrogens and the formation of DNA damage in rats: a comparison. Chem Res Toxicol 2006; 19:852-8. [PMID: 16780365 PMCID: PMC2533849 DOI: 10.1021/tx060052n] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Tamoxifen (TAM) has been used as an agent for the treatment and prevention of breast cancer. However, long-term treatment of TAM in women increases the risk of developing endometrial cancer. The secondary cancer may be due to the genotoxicity of TAM. To find safer alternatives, four selective estrogen receptor modulators (SERMs), 4-hydroxytamoxifen (4-OHTAM), toremifene (TOR), raloxifene (RAL), and ICI 182,780, were administered to rats with an equimolar dose of TAM [54 micromol/kg (20 mg/kg)/day, p.o. for 7 days]. To evaluate the genotoxicity of each SERM, the presence of bulky DNA adducts was determined by (32)P-postlabeling/polyacrylamide gel electrophoresis and (32)P-postlabeling/high-performance liquid chromatography. The formation of 7,8-dihydro-8-oxodeoxyguanosine (8-oxodG) was analyzed as a marker of typical oxidative damage, using liquid chromatography electrospray tandem mass spectrometry. Among the SERMs, bulky DNA adducts were detected in the livers of rats treated with TAM; the total amount of TAM-DNA adducts was 26.1 adducts/10(7) nucleotides. However, with a detection limit of approximately 2 adducts/10(9) nucleotides, no bulky DNA adducts were observed with 4-OHTAM, TOR, RAL, or ICI 182,780. In addition, no significant increase of hepatic 8-oxodG lesions was detected in rats treated with any of the antiestrogens. Therefore, TOR, RAL, and ICI 182,780 are likely to be less genotoxic than TAM.
Collapse
Affiliation(s)
| | | | | | | | | | - Shinya Shibutani
- *To whom correspondence should be addressed. Tel: 631-444-7849/Fax: 631-444-3218., E-mail:
| |
Collapse
|
31
|
Yasui M, Suzuki N, Laxmi YRS, Shibutani S. Translesion synthesis past tamoxifen-derived DNA adducts by human DNA polymerases eta and kappa. Biochemistry 2006; 45:12167-74. [PMID: 17002316 PMCID: PMC2593916 DOI: 10.1021/bi0608461] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The long-term treatment of tamoxifen (TAM), widely used for adjuvant chemotherapy and chemoprevention for breast cancer, increases a risk of developing endometrial cancer. A high frequency of K-ras mutations has been observed in the endometrium of women treated with TAM. Human DNA polymerase (pol) eta and pol kappa are highly expressed in the reproductive organs and are associated with translesion synthesis past bulky DNA adducts. To explore the miscoding properties of alpha-(N2-deoxyguanosinyl)tamoxifen (dG-N2-TAM), a major TAM-DNA adduct, site-specifically modified oligodeoxynucleotides containing a single diastereoisomer of trans or cis forms of dG-N2-TAM were prepared by phosphoramidite chemical procedure and used as templates. The primer extension reaction catalyzed by pol kappa deltaC, a truncated form of pol kappa, extended more efficiently past the adduct than that of pol eta by incorporating dCMP, a correct base, opposite the adduct. With pol eta, all diastereoisomers of dG-N2-TAM promoted small amounts of direct incorporation of dAMP and deletions. With pol kappa deltaC, dG-N2-TAM promoted small amounts of dTMP and/or dAMP incorporations and deletions. The miscoding properties varied depending on the diastereoisomer of dG-N2-TAM adducts and the DNA pol used. Steady-state kinetic studies were also performed using either the nonspecific sequence or the K-ras gene sequence containing a single dG-N2-TAM at the second base of codon 12. With pol eta, the bypass frequency past the dA x dG-N2-TAM pair positioned in the K-ras sequence was only 2.3 times lower than that for the dC x dG-N2-TAM pair, indicating that dG-N2-TAM in the K-ras sequence has higher miscoding potential than that in the nonspecific sequence. However, with pol kappa deltaC, the bypass frequency past the dC x dG-N2-TAM pair was higher than that of the dT x dG-N2-TAM pair in both sequences. The properties of pol eta and pol kappa are consistent with the mutagenic events attributed to TAM-DNA adducts.
Collapse
Affiliation(s)
| | | | | | - Shinya Shibutani
- To whom correspondence should be addressed. Phone: 631−444−7849 Fax: 631−444−3218 E-mail: . State University of New York at Stony Brook
| |
Collapse
|
32
|
Ravazoula P, Androutsopoulos G, Zyli P, Michail G, Kardamakis DM, Kourounis G. A Clinicopathologic Study on Patients with Endometrial Cancer after Adjuvant Tamoxifen Treatment for Breast Cancer: A Single Center Experience. Breast J 2006; 12:578-84. [PMID: 17238994 DOI: 10.1111/j.1524-4741.2006.00353.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
33
|
Kim SY, Suzuki N, Laxmi YRS, Shibutani S. Inefficient repair of tamoxifen-DNA adducts in rats and mice. Drug Metab Dispos 2005; 34:311-7. [PMID: 16299164 DOI: 10.1124/dmd.105.007013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
A long-term treatment with tamoxifen (TAM) to women increases the risk of developing endometrial cancer. The cancer may result from genotoxic damage induced by this drug. In fact, TAM-DNA adducts were detected in the liver of rats treated with TAM and initiated to develop hepatocellular carcinomas. To explore the distribution and repair rate of TAM-DNA adducts, the level of TAM-DNA adducts in all tissues of rats and mice was monitored for 28 days and 7 days, respectively, after the termination of TAM treatment, using 32P-postlabeling/polyacrylamide gel electrophoresis and 32P-postlabeling/HPLC analyses. TAM-DNA adducts were formed specifically in the liver of rodents. In rats, the level of hepatic TAM-DNA adducts was decreased only to 43% in 28 days, indicating that the half-life of adducts was approximately 25 days. Among trans [fraction (fr)-1 and fr-2]- and cis (fr-3 and fr-4)-isoforms of TAM-DNA adducts, a trans-form (fr-1) was removed much more slowly than other adducts, indicating that the repair rate of TAM-DNA adducts varied depending on the structure of isoforms. The repair rate of TAM-DNA adducts was also compared between nucleotide excision repair-deficient (Xpc knockout) and wild mice. Although the level of hepatic TAM-DNA adducts observed with Xpc knockout mice was slightly higher than that of the wild type, the removal of TAM-DNA adducts in both mice was only 20% in 7 days. Thus, TAM-DNA adducts are not efficiently repaired from the targeted tissue, leading to the development of cancer.
Collapse
Affiliation(s)
- Sung Yeon Kim
- Department of Pharmacological Sciences, State University of New York at Stony Brook, 1 Nicolls Road, Stony Brook, NY 11794-8651, USA
| | | | | | | |
Collapse
|
34
|
Garuti G, Cellani F, Centinaio G, Sita G, Nalli G, Luerti M. Histopathologic behavior of endometrial hyperplasia during tamoxifen therapy for breast cancer. Gynecol Oncol 2005; 101:269-73. [PMID: 16297970 DOI: 10.1016/j.ygyno.2005.10.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2005] [Revised: 10/09/2005] [Accepted: 10/14/2005] [Indexed: 10/25/2022]
Abstract
OBJECTIVE The purpose of the present study is to prospectively evaluate the effects of tamoxifen on the pathological behavior of endometrial hyperplasias without atypia, diagnosed before the start of adjuvant endocrine therapy, in menopausal patients suffering from breast cancer. METHODS Twenty-six patients suffering from estrogen-receptor positive breast cancer and candidate to receive adjuvant tamoxifen, were found to be affected by endometrial hyperplasias before the start of endocrine therapy. All women showed a baseline endometrial stripe, measured by transvaginal ultrasonography, thicker than 4 mm and the diagnosis of endometrial hyperplasia was made by hysteroscopy and endometrial biopsy. Two patients showing complex atypical hyperplasia underwent vaginal hysterectomy, whereas the remaining 24 patients, suffering from endometrial hyperplasia without atypia, were followed on a yearly basis during the period of tamoxifen intake, by hysteroscopy and endometrial biopsy. RESULTS Baseline histopathology showed simple and complex hyperplasia in 20 and in 4 patients, respectively. The median follow-up period was 38 months; in particular, all patients underwent endometrial assessment after 12 months, while 22, 16, 10 and 4 patients were followed-up after 24, 36, 48 and 60 months of tamoxifen therapy, respectively. Progression from complex hyperplasia to complex atypical hyperplasia (1 patient) and from complex and simple hyperplasia to adenocarcinomas (2 patients) was found within 24 months of tamoxifen intake in 3 patients (12.5%). In 2 patients (8.3%), a progression from simple to complex hyperplasia was detected within 36 months of tamoxifen treatment. In 13 patients (54.1%), stable histology of simple or complex hyperplasia was found, whereas 6 patients (25.0%), with focal hyperplasia harbored in an endometrial polyp, showed a normalization of endometrial histology after polyp resection. CONCLUSIONS Endometrial hyperplasias without atypia diagnosed before endocrine therapy for breast cancer in menopausal patients show an early and high progression-rate to atypical lesions under tamoxifen influence.
Collapse
Affiliation(s)
- Giancarlo Garuti
- Department of Obstetrics and Gynecology, Lodi Hospital, via Savoia no 1, 26900-Lodi, Italy.
| | | | | | | | | | | |
Collapse
|
35
|
Kim SY, Laxmi YRS, Suzuki N, Ogura K, Watabe T, Duffel MW, Shibutani S. Formation of tamoxifen-DNA adducts via O-sulfonation, not O-acetylation, of alpha-hydroxytamoxifen in rat and human livers. Drug Metab Dispos 2005; 33:1673-8. [PMID: 16099924 DOI: 10.1124/dmd.105.005330] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Tamoxifen (TAM) is used as the standard endocrine therapy for breast cancer patients and as a chemopreventive agent for women at high risk for this disease. Unfortunately, treatment of TAM increases the incidence of endometrial cancer; this may be due to the genotoxic damage induced by TAM metabolites. Formation of TAM-DNA adducts in rat liver correlates with the development of hepatocarcinoma. TAM-DNA adducts are proposed to be formed through O-sulfonation and/or O-acetylation of alpha-hydroxylated TAM and its metabolites. However, the role of O-sulfonation and O-acetylation in the formation of TAM-DNA adducts has not been extensively investigated. Rat or human hydroxysteroid sulfotransferases (HST), acetyltransferases, and liver cytosol were incubated with calf thymus DNA, alpha-OHTAM, and either 3'-phosphoadenosine 5'-phosphosulfate (PAPS) or acetyl coenzyme A (acetyl-CoA) as a cofactor and analyzed for TAM-DNA adduct formation, using 32P postlableling/polyacrylamide gel electrophoresis analysis. TAM-DNA adduct was formed when PAPS, not acetyl-CoA, was used. No TAM-DNA adducts were produced using human N-acetyltransferase I and II. HST antibody inhibited approximately 90% of TAM-DNA adduct formation generated by the cytosol or HST, suggesting that HST is primarily involved in the formation of TAM-DNA adducts. The formation of TAM-DNA adducts with rat liver cytosol and HST was much higher than that of human liver cytosol and HST. Our results indicate that TAM-DNA adducts are formed via O-sulfonation, not O-acetylation, of alpha-hydroxylated TAM and its metabolites.
Collapse
Affiliation(s)
- Sung Yeon Kim
- Laboratory of Chemical Biology, Department of Pharmacological Sciences, State University of New York at Stony Brook, NY 11794-8651, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Laxmi YRS, Suzuki N, Kim SY, Shibutani S. Synthesis of oligodeoxynucleotides containing a single diastereoisomer of alpha-(N2-2'-deoxyguanosinyl)-N-desmethyltamoxifen. Bioorg Med Chem Lett 2005; 14:4051-4. [PMID: 15225724 DOI: 10.1016/j.bmcl.2004.05.030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2004] [Revised: 05/13/2004] [Accepted: 05/14/2004] [Indexed: 11/28/2022]
Abstract
A phosphoramidite chemical synthesis of oligodeoxynucleotides containing a diastereoisomer of (E)-alpha-(N(2)-deoxyguanosinyl)-N-desmethyltamoxifen, a major tamoxifen (TAM)-derived DNA adduct in animal and women treated with TAM, was described. The site-specifically modified oligodeoxynucleotide can be used for mutagenesis, DNA repair, and 3D structural studies and also as standard for quantitative analysis of TAM-DNA adducts in animal and human.
Collapse
Affiliation(s)
- Y R Santosh Laxmi
- Laboratory of Chemical Biology, Department of Pharmacological Sciences, State University of New York at Stony Brook, Stony Brook, New York 11794-8651, USA
| | | | | | | |
Collapse
|
37
|
Schild LJ, Phillips DH, Osborne MR, Hewer A, Beland FA, Churchwell MI, Brown K, Gaskell M, Wright E, Poirier MC. Hepatic DNA adduct dosimetry in rats fed tamoxifen: a comparison of methods. Mutagenesis 2005; 20:115-24. [PMID: 15755801 DOI: 10.1093/mutage/gei015] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Liver homogenates from rats fed tamoxifen (TAM) in the diet were shared among four different laboratories. TAM-DNA adducts were assayed by high pressure liquid chromatography-electrospray tandem mass spectrometry (HPLC-ES-MS/MS), TAM-DNA chemiluminescence immunoassay (TAM-DNA CIA), and (32)P-postlabeling with either thin layer ((32)P-P-TLC) or liquid chromatography ((32)P-P-HPLC) separation. In the first study, rats were fed a diet containing 500 p.p.m. TAM for 2 months, and the values for measurements of the (E)-alpha-(deoxyguanosin-N(2)-yl)-tamoxifen (dG-N(2)-TAM) adduct in replicate rat livers varied by 3.5-fold when quantified using 'in house' TAM-DNA standards, or other approaches where appropriate. In the second study, rats were fed 0, 50, 250 or 500 p.p.m. TAM for 2 months, and TAM-DNA values were quantified using both 'in house' approaches as well as a newly synthesized [N-methyl-(3)H]TAM-DNA standard that was shared among all the participating groups. In the second study, the total TAM-DNA adduct values varied by 2-fold, while values for the dG-N(2)-TAM varied by 2.5-fold. Ratios of dG-N(2)-TAM:(E)-alpha-(deoxyguanosin-N(2)-yl)-N-desmethyltamoxifen (dG-N(2)-N-desmethyl-TAM) in the second study were approximately 1:1 over the range of doses examined. The study demonstrated a remarkably good agreement for TAM-DNA adduct measurements among the diverse methods employed.
Collapse
Affiliation(s)
- Laura J Schild
- Carcinogen-DNA Interactions Section, National Cancer Institute, Building 37, Room 4032 NIH, 37 Convent Drive MSC-4255, Bethesda, MD 20892-4255, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Echchgadda I, Song CS, Oh TS, Cho SH, Rivera OJ, Chatterjee B. Gene regulation for the senescence marker protein DHEA-sulfotransferase by the xenobiotic-activated nuclear pregnane X receptor (PXR). Mech Ageing Dev 2004; 125:733-45. [PMID: 15541768 DOI: 10.1016/j.mad.2004.08.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Dehydroepiandrosterone (DHEA)-sulfotransferase (SULT2A1) is a phase II metabolizing/detoxifying enzyme with substrate preference for physiological hydroxysteroids, diverse drugs and other xenobiotics. The first-pass tissues (liver and intestine) express SULT2A1 at high levels. In senescent male rodents, Sult2A1 gene transcription in the liver is markedly enhanced and calorie restriction retards this increase. Age-associated loss of the liver expression of androgen receptor in part explains the up-regulation of Sult2A1 expression at late life, since androgen receptor is a negative regulator of this gene. In line with its role in xenobiotic metabolism, the Sult2A1 gene is induced by the pregnane X receptor (PXR). PXR is a xenosensing nuclear receptor that is activated by endobiotic (natural steroids) and xenobiotic (therapeutic drugs and environmental chemicals) molecules. An inverted-repeat arrangement (IR0) of the consensus half site binding sequence for nuclear receptors mediates the xenobiotic induction of the Sult2A1 promoter. The IR0 element is a specific binding site for PXR and its heterodimer partner retinoid X receptor (RXR-alpha) and it directs PXR-mediated induction of a heterologous promoter. In contrast to the loss of androgen receptor expression, PXR and RXR-alpha mRNA expression is invariant during aging. Repression by the androgen receptor and induction by PXR may act coordinately to cause the senescence associated and xenobiotic mediated stimulation of Sult2A1 transcription. Increased Sult2A1 expression appears to be an adaptive response to ensure optimal metabolism of Sult2A1 substrates at old age.
Collapse
Affiliation(s)
- Ibtissam Echchgadda
- Department of Molecular Medicine, University of Texas Health Science Center, 15355 Lambda Drive, San Antonio, TX 78245, USA
| | | | | | | | | | | |
Collapse
|