1
|
Zuzak T, Bogaczyk A, Krata AA, Kamiński R, Paneth P, Kluz T. Isotopic Composition of C, N, and S as an Indicator of Endometrial Cancer. Cancers (Basel) 2024; 16:3169. [PMID: 39335141 PMCID: PMC11430076 DOI: 10.3390/cancers16183169] [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: 08/16/2024] [Revised: 09/02/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
OBJECTIVES The metabolic pathway of cancerous tissue differs from healthy tissue, leading to the unique isotopic composition of stable isotopes at their natural abundance. We have studied if these changes can be developed into diagnostic or prognostic tools in the case of endometrial cancer. METHODS Measurements of stable isotope ratios were performed using isotope ratio mass spectrometry for nitrogen, carbon, and sulfur isotopic assessment. Uterine tissue and serum samples were collected from patients and the control group. RESULTS At a natural abundance, the isotopic compositions of all three of the studied elements of uterus cancerous and healthy tissues are different. However, no correlation of the isotopic composition of the tissues with that of serum was found. CONCLUSIONS Differences in the isotopic composition of the tissues might be a potential prognostic tool. However, the lack of a correlation between the differences in the isotopic composition of the tissues and serum seems to exclude their application as diagnostic biomarkers, which, however, might be possible if a position-specific isotopic analysis is performed.
Collapse
Affiliation(s)
- Tomasz Zuzak
- Department of Gynecology, Gynecology Oncology and Obstetrics, Fryderyk Chopin University Hospital, Szopena 2, 35-055 Rzeszow, Poland
| | - Anna Bogaczyk
- Department of Gynecology, Gynecology Oncology and Obstetrics, Fryderyk Chopin University Hospital, Szopena 2, 35-055 Rzeszow, Poland
| | - Agnieszka Anna Krata
- Institute of Applied Radiation Chemistry, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland
| | - Rafał Kamiński
- Institute of Applied Radiation Chemistry, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland
| | - Piotr Paneth
- Institute of Applied Radiation Chemistry, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland
| | - Tomasz Kluz
- Department of Gynecology, Gynecology Oncology and Obstetrics, Fryderyk Chopin University Hospital, Szopena 2, 35-055 Rzeszow, Poland
- Institute of Medical Sciences, Medical College of Rzeszow University, 35-959 Rzeszow, Poland
| |
Collapse
|
2
|
da Costa RSS, Souza NDA, Zukeram KDA, Freire C, Jácome GPO, Koifman RJ, Cardoso CC, Santos SDS. Pon1 and Sult1a1 Polymorphisms and Breast Cancer Among Young Women in Brazil. J Adolesc Young Adult Oncol 2024. [PMID: 39046919 DOI: 10.1089/jayao.2023.0176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024] Open
Abstract
Purpose: To investigate the association of genetic polymorphisms Gln192Arg and Leu55Met of Paraoxonase 1 (PON1) gene, and Arg213His of Sulfotransferase 1A1 (SUT1A1) gene with occurrence of breast cancer among young women living in Rio de Janeiro city. Methods: This is a hospital-based case-control study including 265 women aged 18-35 years, diagnosed with breast cancer at National Cancer Institute; and 277 controls in the same age group selected among women patients and companions of three general hospitals from Rio de Janeiro public health network. Polymorphisms genotyping was performed using the PCR-RFLP technique. Results: For PON1 gene, breast cancer women had a greater chance of being homozygote for Leu55Met polymorphism (ORadjusted = 1.42, 95% CI= 0.67-3.00, recessive model) and a lower chance of having at least one allele of Gln192Arg polymorphism (ORadjusted = 0.75, 95% CI = 0.50-1.13, dominant model), but without statistical significance. Accordingly, frequency of the haplotype Met55/Arg192 was lower among breast cancer women, but no statistically significant association was observed (ORadjusted = 0.85; 95% CI = 0.48-1.51). SULT1A1 His/His genotype was significantly associated with a protective effect for breast cancer (OR adjusted = 0.51, 95% CI = 0.28-0.91, recessive model). Conclusion: Arg213His polymorphism of SUT1A1 gene showed a protective effect against breast cancer among Brazilian young women. More studies with different designs are needed to understand the role of PON1 and SULT1A1 polymorphisms in breast cancer development in young Brazilian women.
Collapse
Affiliation(s)
- Rafaela Soares Senra da Costa
- Public Health and Environment Post-Graduation Program (PPGSPMA, Programa de Pós-Graduação em Saúde Pública e Meio Ambiente). National Public Health School (ENSP, Escola Nacional de Saúde Pública), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
| | - Nara de Almeida Souza
- Public Health and Environment Post-Graduation Program (PPGSPMA, Programa de Pós-Graduação em Saúde Pública e Meio Ambiente). National Public Health School (ENSP, Escola Nacional de Saúde Pública), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
| | - Ketiuce de Azevedo Zukeram
- Department of Epidemiology and Quantitative Methods in Health (DEMQS, Departamento de Epidemiologia e Métodos Quantitativos em Saúde), National School of Public Health (ENSP), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
| | - Carmen Freire
- Department of Legal Medicine and Toxicology, CIBER of Epidemiology and Public Health, University of Granada, Biosanitary Research Institute ibs.granada, Granada, Spain
| | | | - Rosalina Jorge Koifman
- Public Health and Environment Post-Graduation Program (PPGSPMA, Programa de Pós-Graduação em Saúde Pública e Meio Ambiente). National Public Health School (ENSP, Escola Nacional de Saúde Pública), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
- Department of Epidemiology and Quantitative Methods in Health (DEMQS, Departamento de Epidemiologia e Métodos Quantitativos em Saúde), National School of Public Health (ENSP), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
| | - Cynthia Chester Cardoso
- Laboratório de Virologia Molecular, Instituto de Biologia, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Sabrina da Silva Santos
- Public Health and Environment Post-Graduation Program (PPGSPMA, Programa de Pós-Graduação em Saúde Pública e Meio Ambiente). National Public Health School (ENSP, Escola Nacional de Saúde Pública), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
- Department of Epidemiology and Quantitative Methods in Health (DEMQS, Departamento de Epidemiologia e Métodos Quantitativos em Saúde), National School of Public Health (ENSP), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
| |
Collapse
|
3
|
Zhao Y, Tan H, Zhang J, Zhan D, Yang B, Hong S, Pan B, Wang N, Chen T, Shi Y, Wang Z. Developing liver-targeted naringenin nanoparticles for breast cancer endocrine therapy by promoting estrogen metabolism. J Nanobiotechnology 2024; 22:122. [PMID: 38504208 PMCID: PMC10953142 DOI: 10.1186/s12951-024-02356-0] [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: 12/31/2023] [Accepted: 02/20/2024] [Indexed: 03/21/2024] Open
Abstract
Endocrine therapy is standard for hormone receptor-positive (HR+) breast cancer treatment. However, current strategies targeting estrogen signaling pay little attention to estradiol metabolism in the liver and is usually challenged by treatment failure. In a previous study, we demonstrated that the natural compound naringenin (NAR) inhibited HR+ breast cancer growth by activating estrogen sulfotransferase (EST) expression in the liver. Nevertheless, the poor water solubility, low bio-barrier permeability, and non-specific distribution limited its clinical application, particularly for oral administration. Here, a novel nano endocrine drug NAR-cell penetrating peptide-galactose nanoparticles (NCG) is reported. We demonstrated that NCG presented specific liver targeting and increased intestinal barrier permeability in both cell and zebrafish xenotransplantation models. Furthermore, NCG showed liver targeting and enterohepatic circulation in mouse breast cancer xenografts following oral administration. Notably, the cancer inhibition efficacy of NCG was superior to that of both NAR and the positive control tamoxifen, and was accompanied by increased hepatic EST expression and reduced estradiol levels in the liver, blood, and tumor tissue. Moreover, few side effects were observed after NCG treatment. Our findings reveal NCG as a promising candidate for endocrine therapy and highlight hepatic EST targeting as a novel therapeutic strategy for HR+ breast cancer.
Collapse
Affiliation(s)
- Yuying Zhao
- State Key Laboratory of Dampness, Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Hanxu Tan
- The Research Center for Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Juping Zhang
- State Key Laboratory of Dampness, Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Dandan Zhan
- State Key Laboratory of Dampness, Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Bowen Yang
- State Key Laboratory of Dampness, Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Shicui Hong
- State Key Laboratory of Dampness, Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Bo Pan
- State Key Laboratory of Dampness, Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Neng Wang
- The Research Center for Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Tongkai Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| | - Yafei Shi
- The Research Center for Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| | - Zhiyu Wang
- State Key Laboratory of Dampness, Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
- Guangdong Provincial Key Laboratory of Clinical Research On Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong, China.
| |
Collapse
|
4
|
Wang J, Feng Y, Liu B, Xie W. Estrogen sulfotransferase and sulfatase in steroid homeostasis, metabolic disease, and cancer. Steroids 2024; 201:109335. [PMID: 37951289 PMCID: PMC10842091 DOI: 10.1016/j.steroids.2023.109335] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/26/2023] [Accepted: 11/06/2023] [Indexed: 11/13/2023]
Abstract
Sulfation and desulfation of steroids are opposing processes that regulate the activation, metabolism, excretion, and storage of steroids, which account for steroid homeostasis. Steroid sulfation and desulfation are catalyzed by cytosolic sulfotransferase and steroid sulfatase, respectively. By modifying and regulating steroids, cytosolic sulfotransferase (SULT) and steroid sulfatase (STS) are also involved in the pathophysiology of steroid-related diseases, such as hormonal dysregulation, metabolic disease, and cancer. The estrogen sulfotransferase (EST, or SULT1E1) is a typical member of the steroid SULTs. This review is aimed to summarize the roles of SULT1E1 and STS in steroid homeostasis and steroid-related diseases.
Collapse
Affiliation(s)
- Jingyuan Wang
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Ye Feng
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Endocrinology and Metabolic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Brian Liu
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Wen Xie
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| |
Collapse
|
5
|
Bencharski C, Soria EA, Falchini GE, Pasqualini ME, Perez RD. Study of anti-tumorigenic actions of essential fatty acids in a murine mammary gland adenocarcinoma by micro-XRF. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2023; 15:2044-2051. [PMID: 37073557 DOI: 10.1039/d2ay02094g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
In the present work, a statistical experiment based on the microscopy X-ray fluorescence technique was developed to evaluate the effect of diets rich in ω-3 and ω-6 polyunsaturated fatty acids on tumour tissues. Relative variations on the local content of P, S, Ca, Fe, Cu and Zn were analysed in the experiment. Neoplastic tissues were obtained from mammary gland adenocarcinomas inoculated in mice belonging to three different dietary groups: normal, rich in ω-3 and in ω-6 polyunsaturated fatty acids. Slices of 30 microns thick sections of these samples were scanned in the air atmosphere in areas of 5 mm × 5 mm with a spatial resolution of 50 microns using synchrotron radiation. Principal component analysis was employed to analyse the correlation between the X-ray fluorescence signals of P, S, Ca, Fe, Cu and Zn. The subsequent application of the K-means clustering was used for the automatic segmentation of the image scans. By comparison with conventional histological analysis, the clusters were positively identified as tumour parenchyma, transition and necrotic region. The calculation of the mean content of P, S, Ca, Fe, Cu, and Zn in these regions showed that dietary polyunsaturated fatty acids modify elemental content of tumour parenchyma, suggesting its involvement in the antitumour effects of chia oil and protumour effects of safflower oil.
Collapse
Affiliation(s)
- C Bencharski
- IFEG (Physics Institute Enrique Gaviola), CONICET (National Research Council Scientific and Technical), Córdoba, Argentina.
| | - Elio A Soria
- National University of Córdoba, Córdoba, Argentina
| | | | | | - Roberto Daniel Perez
- IFEG (Physics Institute Enrique Gaviola), CONICET (National Research Council Scientific and Technical), Córdoba, Argentina.
| |
Collapse
|
6
|
Liu YZ, Yang K, Zhang W, Zhang Q, Liu TF, Xu T, Li Y, Ran RX, Yang K, Cao YF, Fang ZZ. Inhibition of human sulfotransferases (SULTs) by per- and polyfluoroalkyl substances (PFASs) and structure-activity relationship. Food Chem Toxicol 2023; 174:113664. [PMID: 36775137 DOI: 10.1016/j.fct.2023.113664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 01/15/2023] [Accepted: 02/09/2023] [Indexed: 02/12/2023]
Abstract
Per- and polyfluoroalkyl substances (PFASs) are a family of highly fluorinated aliphatic substances widely used in industrial and commercial applications. This study aims to determine the inhibition of PFASs towards sulfotransferases (SULTs) activity, and trying to explain the toxicity mechanism of PFASs. In vitro recombinant SULTs-catalyzed sulfation of p-nitrophenol (PNP) was utilized as a probe reaction. The incubation system was consisted of PFASs, SULTs, PNP, 3'-phosphoadenosine-5'-phosphosulfate, MgCl2 and Tris-HCl buffer. Ultra-performance liquid chromatography was employed for analysis of the metabolites. All tested PFASs showed inhibition towards SULTs. The longer the carbon chain length of the PFASs terminated with -COOH, the higher is its capability of inhibiting SULT1A3. PFASs with -SO3H had a relatively higher ability to inhibit SULT1A3 activity than those with -COOH, -I and -OH. The inhibition kinetic parameter was 2.16 and 1.42 μM for PFOS-SULT1A1, PFTA-SULT1B1. In vitro in vivo extrapolation showed that the concentration of PFOS and PFTA in human matrices might be higher than the threshold for inducing inhibition of SULTs. Therefore, PFASs could interfere with the metabolic pathways catalyzed by SULTs in vivo. All these results will help to understand the toxicity of PFASs from the perspective of metabolism.
Collapse
Affiliation(s)
- Yong-Zhe Liu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, 300070, China; Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, Hebei, 050000, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, 300070, China; National Demonstration Center for Experimental Preventive Medicine Education, Tianjin Medical University, Tianjin, 300070, China; Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin, 300070, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institute, Hefei, 230032, China
| | - Kai Yang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| | - Wei Zhang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| | - Qian Zhang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, 300070, China; Baoding First Central Hospital, Baoding, 071000, China
| | - Tong-Feng Liu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| | - Tong Xu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| | - Yang Li
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| | - Rui-Xue Ran
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Kun Yang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, 300070, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, 300070, China; National Demonstration Center for Experimental Preventive Medicine Education, Tianjin Medical University, Tianjin, 300070, China; Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin, 300070, China
| | - Yun-Feng Cao
- Shanghai Institute for Biomedical and Pharmaceutical Technologies, NHC Key Laboratory of Reproduction Regulation, ShangHai, 200032, China.
| | - Zhong-Ze Fang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, 300070, China; Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, Hebei, 050000, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, 300070, China; National Demonstration Center for Experimental Preventive Medicine Education, Tianjin Medical University, Tianjin, 300070, China; Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin, 300070, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institute, Hefei, 230032, China.
| |
Collapse
|
7
|
Shi M, Yin L, Sun Y, Wang C, Cai L, Zhang T, Zhou X, Fawcett JP, Gao X, Gu J. Pharmacokinetic Study of Conjugated Equine Estrogens in Healthy Chinese Postmenopausal Women Using a Parallel Two-Column LC-MS/MS Method. Clin Pharmacokinet 2022; 61:1571-1583. [PMID: 36173592 DOI: 10.1007/s40262-022-01177-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND AND OBJECTIVE: Postmenopausal women often require estrogen supplementation to improve menopausal and postmenopausal vasomotor symptoms and maintain hormonal balance. Conjugated equine estrogens extracted from the urine of pregnant mares are commonly used to provide this estrogen replacement therapy. The complex composition of this mixture of animal sulfated metabolites makes its bioanalysis challenging such that its detailed pharmacokinetics has not been fully characterized. The purpose of this work is to reveal the pharmacokinetic behavior of conjugated equine estrogens in healthy Chinese postmenopausal women by a parallel two-column LC-MS/MS method. METHODS An open-label study was carried out in 35 Chinese healthy postmenopausal women who received a single dose of Premarin® 0.625 mg. A high-throughput column-switching liquid chromatography-tandem mass spectrometry method was developed to determine four conjugated estrogens and two unconjugated estrogens formed by hydrolysis in vivo. The method multiplexes two high-performance liquid chromatography systems into one mass spectrometer and incorporates the positive/negative ion switching acquisition mode of mass spectrometry to significantly increase analysis efficiency. Pharmacokinetics was determined using non-compartmental methods. RESULTS Both conjugated and unconjugated estrogens can be analyzed simultaneously in a single run with an analysis time of 13.0 minutes in the column-switching liquid chromatography-tandem mass spectrometry method as opposed to 23.0 minutes in a single-column liquid chromatography-tandem mass spectrometry system. The exposures (maximum concentration and area under the curve) of estrone and equilin in Chinese women were higher than those in the North American women. CONCLUSIONS The fully validated assay was successfully applied to a pharmacokinetic study in healthy postmenopausal Chinese women after oral administration of a conjugated equine estrogen tablet. This study suggests that Chinese postmenopausal women achieve the same level of unconjugated estrogens in plasma at a lower dose of conjugated equine estrogens than North American women.
Collapse
Affiliation(s)
- Meiyun Shi
- Research Center for Drug Metabolism, School of Life Science, Jilin University, 2699 Qianjin Street, Changchun, 130012, People's Republic of China.,School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, People's Republic of China
| | - Lei Yin
- Research Center for Drug Metabolism, School of Life Science, Jilin University, 2699 Qianjin Street, Changchun, 130012, People's Republic of China.,School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, People's Republic of China
| | - Yantong Sun
- School of Pharmaceutical Sciences, Jilin University, Changchun, People's Republic of China
| | - Can Wang
- Research Center for Drug Metabolism, School of Life Science, Jilin University, 2699 Qianjin Street, Changchun, 130012, People's Republic of China
| | - Lanlan Cai
- Research Center for Drug Metabolism, School of Life Science, Jilin University, 2699 Qianjin Street, Changchun, 130012, People's Republic of China
| | - Tinglan Zhang
- Research Center for Drug Metabolism, School of Life Science, Jilin University, 2699 Qianjin Street, Changchun, 130012, People's Republic of China
| | - Xiaotong Zhou
- Research Center for Drug Metabolism, School of Life Science, Jilin University, 2699 Qianjin Street, Changchun, 130012, People's Republic of China
| | - J Paul Fawcett
- Research Center for Drug Metabolism, School of Life Science, Jilin University, 2699 Qianjin Street, Changchun, 130012, People's Republic of China
| | - Xiaoli Gao
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830011, People's Republic of China. .,Xinjiang Key Laboratory of Active Components and Drug Release Technology of Natural Drugs, Urumqi, People's Republic of China. .,Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, Beijing, People's Republic of China.
| | - Jingkai Gu
- Research Center for Drug Metabolism, School of Life Science, Jilin University, 2699 Qianjin Street, Changchun, 130012, People's Republic of China.
| |
Collapse
|
8
|
Jovanovic F, Sudhakar A, Knezevic NN. The Kynurenine Pathway and Polycystic Ovary Syndrome: Inflammation as a Common Denominator. Int J Tryptophan Res 2022; 15:11786469221099214. [PMID: 35620306 PMCID: PMC9128055 DOI: 10.1177/11786469221099214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 04/20/2022] [Indexed: 11/17/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is a complex metabolic disorder commonly seen in females of reproductive age. The pathophysiology of PCOS is multifactorial and includes dysfunction in ovarian steroidogenesis and folliculogenesis, impaired gonadotropin levels, insulin resistance, gut microbiota imbalance, genetic predisposition, and lifestyle preferences. Low-grade inflammatory conditions such as obesity and impaired glucose tolerance are common metabolic disturbances in women with PCOS. A growing body of literature suggests strong evidence rendering PCOS in close proximity with chronic inflammation as documented by high levels of serum white blood cells, C-reactive protein, and various proinflammatory cytokines seen in this condition. Inflammation seems to be the most common metabolic denominator between the kynurenine pathway and PCOS. The association of tryptophan and kynurenine pathway has already been well documented in mood disorders, neurodegenerative diseases, chronic pain conditions, and different inflammatory states. In this manuscript, we describe the influence of sex steroid hormones on different enzymes of the KP; inflammatory nature of PCOS and CRP as a marker of IDO/TDO activity; and the effects of altered gut flora in women with PCOS. This review provides a novel view of the available evidence of tryptophan and downstream metabolites in PCOS in the context of underlying inflammation.
Collapse
Affiliation(s)
- Filip Jovanovic
- Department of Internal Medicine, Merit Health Wesley, Hattiesburg, MS, USA
| | - Aboorva Sudhakar
- Department of Internal Medicine, Merit Health Wesley, Hattiesburg, MS, USA
| | - Nebojsa Nick Knezevic
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, Chicago, USA
- Department of Anesthesiology, University of Illinois, Chicago, USA
- Department of Surgery, University of Illinois, Chicago, USA
| |
Collapse
|
9
|
Estrogen Sulfotransferase is Highly Expressed in Vascular Endothelial Cells Overlying Atherosclerotic Plaques. Protein J 2022; 41:179-188. [DOI: 10.1007/s10930-022-10042-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2022] [Indexed: 12/15/2022]
|
10
|
Sato A, Watanabe H, Yamazaki M, Sakurai E, Ebina K. Interaction of Native- and Oxidized-Low-Density Lipoprotein with Human Estrogen Sulfotransferase. Protein J 2021; 40:192-204. [PMID: 33665770 DOI: 10.1007/s10930-021-09971-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/19/2021] [Indexed: 12/11/2022]
Abstract
Cytosolic estrogen sulfotransferase (SULT1E) mainly catalyzes the sulfate conjugation of estrogens, which decrease atherosclerosis progression. Recently we reported that a YKEG sequence in human SULT1E1 (hSULT1E1) corresponding to residues 61-64 can bind specifically to oxidized low-density lipoprotein (Ox-LDL), which plays a major role in the pathogenesis of atherosclerosis; its major oxidative lipid component lysophosphatidylcholine (LPC), and its structurally similar lipid, platelet-activating factor (PAF). In this study, we investigated the effect of Ox-LDL on the sulfating activity of hSULT1E1. In vivo experiments using a mouse model of atherosclerosis showed that the protein expression of SULT1E1 was higher in the aorta of mice with atherosclerosis compared with that in control animals. Results from a sulfating activity assay of hSULT1E1 using 1-hydroxypyrene as the substrate demonstrated that Ox-LDL, LPC, and PAF markedly decreased the sulfating activity of hSULT1E1, whereas native LDL and 1-palmitoyl-2-(5'-oxo-valeroyl)-sn-glycero-3-phosphocholine (POVPC) as one of the oxidized phosphatidylcholines showed the opposite effect. The sulfating activity greatly changed in the presence of LPC, PAF, and POVPC in their concentration-dependen manner (especially above their critical micelle concentrations). Moreover, Ox-LDL specifically recognized dimeric hSULT1E1. These results suggest that the effects of Ox-LDL and native LDL on the sulfating activity of hSULT1E1 might be helpful in elucidating the novel mechanism underlying the pathogenesis of atherosclerosis, involving the relationship between estrogen metabolism, LDL, and Ox-LDL.
Collapse
Affiliation(s)
- Akira Sato
- Department of Pharmaceutical Health Science, Faculty of Pharmacy, Iryo Sosei University, 5-5-1, Chuodai-Iino, Iwaki, Fukushima, 970-8551, Japan. .,Graduate School of Life Science and Technology, Iryo Sosei University, 5-5-1, Chuodai-Iino, Iwaki, Fukushima, 970-8551, Japan.
| | - Hinako Watanabe
- Department of Pharmaceutical Health Science, Faculty of Pharmacy, Iryo Sosei University, 5-5-1, Chuodai-Iino, Iwaki, Fukushima, 970-8551, Japan
| | - Miyuki Yamazaki
- Department of Pharmaceutical Health Science, Faculty of Pharmacy, Iryo Sosei University, 5-5-1, Chuodai-Iino, Iwaki, Fukushima, 970-8551, Japan
| | - Eiko Sakurai
- Department of Pharmaceutical Health Science, Faculty of Pharmacy, Iryo Sosei University, 5-5-1, Chuodai-Iino, Iwaki, Fukushima, 970-8551, Japan.,Graduate School of Life Science and Technology, Iryo Sosei University, 5-5-1, Chuodai-Iino, Iwaki, Fukushima, 970-8551, Japan
| | - Keiichi Ebina
- Department of Pharmaceutical Health Science, Faculty of Pharmacy, Iryo Sosei University, 5-5-1, Chuodai-Iino, Iwaki, Fukushima, 970-8551, Japan.,Graduate School of Life Science and Technology, Iryo Sosei University, 5-5-1, Chuodai-Iino, Iwaki, Fukushima, 970-8551, Japan
| |
Collapse
|
11
|
Jackson SS, Van De Wyngard V, Pfeiffer RM, Cook P, Hildesheim A, Pinto LA, Jackson SH, Choi K, Verdugo RA, Cuevas M, Yáñez C, Tobar-Calfucoy E, Retamales-Ortega R, Araya JC, Ferreccio C, Koshiol J. Inflammatory profiles in Chilean Mapuche and non-Mapuche women with gallstones at risk of developing gallbladder cancer. Sci Rep 2021; 11:3686. [PMID: 33574564 PMCID: PMC7878792 DOI: 10.1038/s41598-021-83300-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 02/01/2021] [Indexed: 01/11/2023] Open
Abstract
Chile has high incidence rates of gallbladder cancer globally, particularly among Amerindian women, who also have a high prevalence of gallstones. We examined differences in inflammatory biomarkers between Mapuche and non-Mapuche women from the Chile Biliary Longitudinal Study, a cohort of women with ultrasound-detected gallstones. We randomly selected 200 Mapuche women frequency matched to non-Mapuche women on age and statin use Inflammatory biomarkers were analyzed using a multiplex assay and linear regression to assess associations of a priori markers (CCL20, CXCL10, IL-6, and IL-8) with ethnicity. Novel biomarkers were analyzed using exploratory factor analysis (EFA) and sufficient dimension reduction (SDR) to identify correlated marker groups, followed by linear regression to examine their association with ethnicity. The mean values of IL-8 were higher in Mapuche than non-Mapuche women (P = 0.04), while CCL20, CXCL10, and IL-6 did not differ significantly by ethnicity. EFA revealed two marker groups associated with ethnicity (P = 0.03 and P < 0.001). SDR analysis confirmed correlation between the biomarkers and ethnicity. We found higher IL-8 levels among Mapuche than non-Mapuche women. Novel inflammatory biomarkers were correlated with ethnicity and should be studied further for their role in gallbladder disease. These findings may elucidate underlying ethnic disparities in gallstones and carcinogenesis among Amerindians.
Collapse
Affiliation(s)
- Sarah S Jackson
- Infections and Immunoepidemiology Branch, Division of Cancer Epidemiology and Genetics, National Institutes of Health, National Cancer Institute, Rockville, MD, USA.
| | - Vanessa Van De Wyngard
- Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), FONDAP, Santiago, Chile
| | - Ruth M Pfeiffer
- Infections and Immunoepidemiology Branch, Division of Cancer Epidemiology and Genetics, National Institutes of Health, National Cancer Institute, Rockville, MD, USA
| | - Paz Cook
- Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), FONDAP, Santiago, Chile
| | - Allan Hildesheim
- Infections and Immunoepidemiology Branch, Division of Cancer Epidemiology and Genetics, National Institutes of Health, National Cancer Institute, Rockville, MD, USA
| | - Ligia A Pinto
- Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Sharon H Jackson
- Division of Intramural Research, National Institute on Minority Health and Health Disparities, Bethesda, MD, USA
| | - Kelvin Choi
- Division of Intramural Research, National Institute on Minority Health and Health Disparities, Bethesda, MD, USA
| | - Ricardo A Verdugo
- Programa de Genética Human, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Departamento de Oncología Básico Clínica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Mara Cuevas
- Programa de Genética Human, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Cristian Yáñez
- Programa de Genética Human, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Eduardo Tobar-Calfucoy
- Programa de Genética Human, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Rocío Retamales-Ortega
- Programa de Genética Human, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Juan Carlos Araya
- Advanced Center for Chronic Diseases (ACCDiS), FONDAP, Santiago, Chile.,Hospital Dr. Hernan Henríquez Aravena, Temuco, Chile.,Department of Pathology, Faculty of Medicine, Universidad de la Frontera, Temuco, Chile
| | - Catterina Ferreccio
- Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), FONDAP, Santiago, Chile
| | - Jill Koshiol
- Infections and Immunoepidemiology Branch, Division of Cancer Epidemiology and Genetics, National Institutes of Health, National Cancer Institute, Rockville, MD, USA
| |
Collapse
|
12
|
Xie Y, Xie W. The Role of Sulfotransferases in Liver Diseases. Drug Metab Dispos 2020; 48:742-749. [PMID: 32587100 PMCID: PMC7469250 DOI: 10.1124/dmd.120.000074] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 06/08/2020] [Indexed: 12/19/2022] Open
Abstract
The cytosolic sulfotransferases (SULTs) are phase II conjugating enzymes that catalyze the transfer of a sulfonate group from the universal sulfate donor 3'-phosphoadenosine-5'-phosphosulfate to a nucleophilic group of their substrates to generate hydrophilic products. Sulfation has a major effect on the chemical and functional homeostasis of substrate chemicals. SULTs are widely expressed in metabolically active or hormonally responsive tissues, including the liver and many extrahepatic tissues. The expression of SULTs exhibits isoform-, tissue-, sex-, and development-specific regulations. SULTs display a broad range of substrates including xenobiotics and endobiotics. The expression of SULTs has been shown to be transcriptionally regulated by members of the nuclear receptor superfamily, such as the peroxisome proliferator-activated receptors, pregnane X receptor, constitutive androstane receptor, vitamin D receptor, liver X receptors, farnesoid X receptor, retinoid-related orphan receptors, estrogen-related receptors, and hepatocyte nuclear factor 4α These nuclear receptors can be activated by numerous xenobiotics and endobiotics, such as fatty acids, bile acids, and oxysterols, many of which are substrates of SULTs. Due to their metabolism of xenobiotics and endobiotics, SULTs and their regulations are implicated in the pathogenesis of many diseases. This review is aimed to summarize the central role of major SULTs, including the SULT1 and SULT2 subfamilies, in the pathophysiology of liver and liver-related diseases. SIGNIFICANCE STATEMENT: Sulfotransferases (SULTs) are indispensable in the homeostasis of xenobiotics and endobiotics. Knowing SULTs and their regulations are implicated in human diseases, it is hoped that genetic or pharmacological manipulations of the expression and/or activity of SULTs can be used to affect the clinical outcome of diseases.
Collapse
Affiliation(s)
- Yang Xie
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania (Y.X., W.X.) and Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania (W.X.)
| | - Wen Xie
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania (Y.X., W.X.) and Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania (W.X.)
| |
Collapse
|
13
|
Sato A, Yamazaki M, Watanabe H, Sakurai E, Ebina K. Human estrogen sulfotransferase and its related fluorescently labeled decapeptides specifically interact with oxidized low-density lipoprotein. J Pept Sci 2020; 26:e3274. [PMID: 32633098 DOI: 10.1002/psc.3274] [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: 04/06/2020] [Revised: 05/16/2020] [Accepted: 06/12/2020] [Indexed: 12/24/2022]
Abstract
Estrogen sulfotransferase (SULT1E) mainly catalyzes the sulfation of estrogens, which are known to prevent the pathogenesis of atherosclerosis. Recently, we found that peptides with a YKDG sequence specifically bind to oxidized low-density lipoprotein (Ox-LDL), which plays a major role in the pathogenesis of atherosclerosis. Here, we investigated the interaction between human SULT1E1 (hSULT1E1), which has a YKEG sequence (residues 61-64) unlike other human SULTs, and Ox-LDL. Results from polyacrylamide gel electrophoresis and western blotting demonstrated that hSULT1E1 specifically binds to Ox-LDL and its major lipid component (lysophosphatidylcholine; LPC), and platelet-activating factor (PAF), which bears a marked resemblance to LPC in terms of structure and activity. Moreover, an N-terminally fluorescein isothiocyanate (FITC)-labeled decapeptide (MIYKEGDVEK; FITC-hSULT1E1-P10) corresponding to residues 59-68 of hSULT1E1 specifically binds to Ox-LDL, LPC, and PAF. Unveiling the specific interaction between hSULT1E1 and Ox-LDL, LPC, and PAF provides important information regarding the mechanisms underlying various diseases caused by Ox-LDL, LPC, and PAF, such as atherosclerosis. In addition, FITC-hSULT1E1-P10 could be used as an efficient fluorescent probe for the detection of Ox-LDL, LPC, and PAF, which could facilitate the mechanistic study, identification, diagnosis, prevention, and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Akira Sato
- Department of Pharmaceutical Health Science, Faculty of Pharmacy, Iryo Sosei University, Iwaki, Fukushima, Japan.,Graduate School of Life Science and Technology, Iryo Sosei University, Iwaki, Fukushima, Japan
| | - Miyuki Yamazaki
- Department of Pharmaceutical Health Science, Faculty of Pharmacy, Iryo Sosei University, Iwaki, Fukushima, Japan
| | - Hinako Watanabe
- Department of Pharmaceutical Health Science, Faculty of Pharmacy, Iryo Sosei University, Iwaki, Fukushima, Japan
| | - Eiko Sakurai
- Department of Pharmaceutical Health Science, Faculty of Pharmacy, Iryo Sosei University, Iwaki, Fukushima, Japan.,Graduate School of Life Science and Technology, Iryo Sosei University, Iwaki, Fukushima, Japan
| | - Keiichi Ebina
- Department of Pharmaceutical Health Science, Faculty of Pharmacy, Iryo Sosei University, Iwaki, Fukushima, Japan.,Graduate School of Life Science and Technology, Iryo Sosei University, Iwaki, Fukushima, Japan
| |
Collapse
|
14
|
Nazmeen A, Chen G, Ghosh TK, Maiti S. Breast cancer pathogenesis is linked to the intra-tumoral estrogen sulfotransferase (hSULT1E1) expressions regulated by cellular redox dependent Nrf-2/NF κβ interplay. Cancer Cell Int 2020; 20:70. [PMID: 32158360 PMCID: PMC7057506 DOI: 10.1186/s12935-020-1153-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 02/24/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Estrogen sulfotransferase catalyzes conjugation of sulfuryl-group to estradiol/estrone and regulates E2 availability/activity via estrogen-receptor or non-receptor mediated pathways. Sulfoconjugated estrogen fails to bind estrogen-receptor (ER). High estrogen is a known carcinogen in postmenopausal women. Reports reveal a potential redox-regulation of hSULT1E1/E2-signalling. Further, oxidatively-regulated nuclear-receptor-factor 2 (Nrf2) and NFκβ in relation to hSULT1E1/E2 could be therapeutic-target via cellular redox-modification. METHODS Here, oxidative stress-regulated SULT1E1-expression was analyzed in human breast carcinoma-tissues and in rat xenografted with human breast-tumor. Tumor and its surrounding tissues were obtained from the district-hospital. Intracellular redox-environment of tumors was screened with some in vitro studies. RT-PCR and western blotting was done for SULT1E1 expression. Immunohistochemistry was performed to analyze SULT1E1/Nrf2/NFκβ localization. Tissue-histoarchitecture/DNA-stability (comet assay) studies were done. RESULTS Oxidative-stress induces SULT1E1 via Nrf2/NFκβ cooperatively in tumor-pathogenesis to maintain the required proliferative-state under enriched E2-environment. Higher malondialdehyde/non-protein-soluble-thiol with increased superoxide-dismutase/glutathione-peroxidase/catalase activities was noticed. SULT1E1 expression and E2-level were increased in tumor-tissue compared to their corresponding surrounding-tissues. CONCLUSIONS It may be concluded that tumors maintain a sustainable oxidative-stress through impaired antioxidants as compared to the surrounding. Liver-tissues from xenografted rat manifested similar E2/antioxidant dysregulations favoring pre-tumorogenic environment.
Collapse
Affiliation(s)
- Aarifa Nazmeen
- Dept. of Biochemistry, Cell & Molecular Therapeutics Lab, Oriental Institute of Science & Technology, Midnapore, 721101 India
| | - Guangping Chen
- Venture I OSU Laboratory, Oklahoma Technology & Research Park, 1110 S. Innovation Way, Stillwater, OK 74074 USA
| | - Tamal Kanti Ghosh
- Special Secretary, Higher Medical Education, Health and Family Welfare Dept, Govt. of West Bengal, Salt Lake, Calcutta, India
| | - Smarajit Maiti
- Dept. of Biochemistry, Cell & Molecular Therapeutics Lab, Oriental Institute of Science & Technology, Midnapore, 721101 India
- Department of Biochemistry and Biotechnology, Cell & Molecular Therapeutics Lab, OIST, Midnapore, 721102 India
| |
Collapse
|
15
|
Di Nisio A, Rocca MS, Sabovic I, De Rocco Ponce M, Corsini C, Guidolin D, Zanon C, Acquasaliente L, Carosso AR, De Toni L, Foresta C. Perfluorooctanoic acid alters progesterone activity in human endometrial cells and induces reproductive alterations in young women. CHEMOSPHERE 2020; 242:125208. [PMID: 31896193 DOI: 10.1016/j.chemosphere.2019.125208] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 10/10/2019] [Accepted: 10/23/2019] [Indexed: 06/10/2023]
Abstract
Female fecundity is finely regulated by hormonal signaling, representing a potential target for endocrine-disrupting chemicals. Among the chemicals of most concern are the perfluoroalkyl substances (PFAS), widely used in consumer goods, that are associated with adverse effects on reproductive health. In this context, the endometrium clearly represents an important fertility determining factor. The aim of this study was to investigate PFAS interference on hormonal endometrial regulation. This study was performed within a screening protocol to evaluate reproductive health in high schools. We studied a cohort of 146 exposed females aged 18-21 from the Veneto region in Italy, one of the four areas worldwide heavily polluted with PFAS, and 1080 non-exposed controls. In experiments on Ishikawa cells included UV-Vis spectroscopy, microarray analysis and qPCR. We report a significant dysregulation of the genetic cascade leading to embryo implantation and endometrial receptivity. The most differentially-expressed genes upon PFOA coincubation were ITGB8, KLF5, WNT11, SULT1E1, ALPPL2 and G0S2 (all p < 0.01). By qPCR, we confirmed an antagonistic effect of PFOA on all these genes, which was reversed at higher progesterone levels. Molecular interference of PFOA on progesterone was confirmed by an increase in the intensity of absorption spectra at 250 nm in a dose-dependent manner, but not in the presence of β-estradiol. Age at menarche (+164 days, p = 0.006) and the frequency of girls with irregular periods (29.5% vs 21.5%, p = 0.022) were significantly higher in the exposed group. Our results are indicative of endocrine-disrupting activity of PFAS on progesterone-mediated endometrial function.
Collapse
Affiliation(s)
- A Di Nisio
- Department of Medicine, Operative Unit of Andrology and Medicine of Human Reproduction, University of Padova, Via Giustiniani, 2, 35128, Padova, Italy
| | - M S Rocca
- Familial Cancer Clinic, Veneto Institute of Oncology (IOV-IRCSS), 35128, Padova, Italy
| | - I Sabovic
- Department of Medicine, Operative Unit of Andrology and Medicine of Human Reproduction, University of Padova, Via Giustiniani, 2, 35128, Padova, Italy
| | - M De Rocco Ponce
- Department of Medicine, Operative Unit of Andrology and Medicine of Human Reproduction, University of Padova, Via Giustiniani, 2, 35128, Padova, Italy
| | - C Corsini
- Department of Medicine, Operative Unit of Andrology and Medicine of Human Reproduction, University of Padova, Via Giustiniani, 2, 35128, Padova, Italy
| | - D Guidolin
- Department of Neurosciences, University of Padova, Via Gabelli 65, 35128, Padova, Italy
| | - C Zanon
- Istituto di Ricerca Pediatrica (IRP) - Fondazione Città della Speranza, Corso Stati Uniti 4, Padova, Italy
| | - L Acquasaliente
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine 1100 South Grand Blvd. St. Louis, MO, 63104, USA
| | - A R Carosso
- Department of Surgical Sciences, Gynecology and Obstetrics 1, Physiopathology of Reproduction and IVF Unit, S. Anna Hospital, University of Torino, 10126, Torino, Italy
| | - L De Toni
- Department of Medicine, Operative Unit of Andrology and Medicine of Human Reproduction, University of Padova, Via Giustiniani, 2, 35128, Padova, Italy
| | - C Foresta
- Department of Medicine, Operative Unit of Andrology and Medicine of Human Reproduction, University of Padova, Via Giustiniani, 2, 35128, Padova, Italy.
| |
Collapse
|
16
|
Aversa J, Song M, Shimazu T, Inoue M, Charvat H, Yamaji T, Sawada N, Pfeiffer RM, Karimi P, Dawsey SM, Rabkin CS, Tsugane S, Camargo MC. Prediagnostic circulating inflammation biomarkers and esophageal squamous cell carcinoma: A case-cohort study in Japan. Int J Cancer 2019; 147:686-691. [PMID: 31671219 DOI: 10.1002/ijc.32763] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/14/2019] [Accepted: 10/15/2019] [Indexed: 01/05/2023]
Abstract
Esophageal squamous cell carcinoma (ESCC) is the predominant histologic subtype of esophageal cancer worldwide. Measurements of circulating inflammation-related biomarkers may inform etiology or provide noninvasive signatures for early diagnosis. We therefore examined levels of inflammation molecules for associations with ESCC risk. Using a case-cohort study designed within the Japan Public Health Center-based Prospective Study, we measured baseline plasma levels of 92 biomarkers using a multiplex assay in a subcohort of 410 randomly selected participants and 66 participants with incident ESCC (including four cases that occurred in the subcohort). ESCC hazard ratios (HRs) were calculated for 2-4 quantiles of each biomarker by Cox proportional hazards regression models with age as the time metric, adjusted for sex, smoking and alcohol use. Twenty analytes were undetectable in nearly all samples. Of the remaining 72, 12 biomarkers (FGF19, ST1A1, STAMBP, AXIN1, CASP8, NT3, CD6, CDCP1, CD5, SLAMF1, OPG and CSF1) were associated with increased ESCC risk (ptrend < 0.05) with HRs per quantile 1.28-1.65. Seven biomarkers (CXCL6, CCL23, CXCL5, TGFA, CXCL1, OSM and CCL4) were inversely associated with HRs 0.57-0.72. FGF19, CASP8, STAMBP, ST1A1 and CCL-4 met statistical significance with false discovery rate correction. Associations did not differ <5 vs. ≥5 years between blood collection and ESCC diagnosis. CASP8, STAMBP and ST1A1 were strongly correlated (p < 0.05). Our study expands the range of inflammation molecules associated with the development of this highly lethal neoplasia. Correlations among these novel biomarkers suggest a possible shared pathway. These findings need replication and could further delineate ESCCs molecular mechanisms of carcinogenesis.
Collapse
Affiliation(s)
- John Aversa
- Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Minkyo Song
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Taichi Shimazu
- Epidemiology and Prevention Group, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - Manami Inoue
- Epidemiology and Prevention Group, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - Hadrien Charvat
- Epidemiology and Prevention Group, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - Taiki Yamaji
- Epidemiology and Prevention Group, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - Norie Sawada
- Epidemiology and Prevention Group, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - Ruth M Pfeiffer
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Parisa Karimi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Sanford M Dawsey
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Charles S Rabkin
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Shoichiro Tsugane
- Epidemiology and Prevention Group, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - M Constanza Camargo
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
17
|
Mao X, Dong B, Gao M, Ruan G, Huang M, Braicu EI, Sehouli J, Sun P. Dual targeting of estrogen receptor α and estrogen-related receptor α: a novel endocrine therapy for endometrial cancer. Onco Targets Ther 2019; 12:6757-6767. [PMID: 31686835 PMCID: PMC6709363 DOI: 10.2147/ott.s216146] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 07/16/2019] [Indexed: 11/24/2022] Open
Abstract
Background Endometrial cancer (EC) is a hormone dependent carcinoma that may involve complex molecular mechanisms. Endocrine therapy by blocking the estrogen and estrogen receptor α (ERα) has been effective in breast cancer, while it is still controversial in EC. Recently, estrogen-related receptor α (ERRα) was proven to be another endocrine therapy target. Methods The anti-tumor effect of selective estrogen receptor modulators (SERMs) and XCT790 (XCT) used alone or in combination were evaluated in both of ERα-positive (ERα+) and ERα-negative (ERα-) EC cells. ERα and ERRα mRNA were tested by qPCR, while the protein was detected by Western blot. The proliferation was tested by MTS and cell cycle, apoptosis rate were analyzed by flow cytometry. Results A relatively high dose (10 μM) of tamoxifen (TAM) suppressed the expression of ERα and ERRα in two types of EC cells. However, 10 μM raloxifene (RAL) exhibited no effect on ERα and ERRα, while 10 μM XCT down regulated ERRα specifically, but not ERα in all EC cells. When dual targeting on ERα and ERRα by combining TAM with XCT, the proliferation inhibitory effect and apoptosis reached the strongest in all EC cells (P<0.05). Moreover, the inhibitory effect of proliferation was attributed significantly to the G0/G1 arrest (P<0.05). Interestingly, the apoptosis induced by combining TAM with XCT were obviously higher in ERα+ EC cells than ERα- EC cells (P<0.05). Conclusion Taken together, the results indicate that dual targeting on ERα and ERRα represents a better anti-tumor effect, which provides a novel endocrine based therapy strategy for EC.
Collapse
Affiliation(s)
- XiaoDan Mao
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, People's Republic of China
| | - Binhua Dong
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, People's Republic of China
| | - Min Gao
- Department of Gynecology Oncology, Peking University Cancer Hospital, Beijing 100142, People's Republic of China
| | - GuanYu Ruan
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, People's Republic of China
| | - MeiMei Huang
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, People's Republic of China
| | - Elena Ioana Braicu
- Department of Gynecology, Campus Virchow-Klinikum, Charité Universitätmedizin Berlin, Berlin D-13353, Germany
| | - Jalid Sehouli
- Department of Gynecology, Campus Virchow-Klinikum, Charité Universitätmedizin Berlin, Berlin D-13353, Germany
| | - PengMing Sun
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, People's Republic of China
| |
Collapse
|
18
|
Ancuceanu R, Dinu M, Dinu-Pirvu C, Anuţa V, Negulescu V. Pharmacokinetics of B-Ring Unsubstituted Flavones. Pharmaceutics 2019; 11:E370. [PMID: 31374885 PMCID: PMC6723510 DOI: 10.3390/pharmaceutics11080370] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 07/23/2019] [Accepted: 07/23/2019] [Indexed: 02/07/2023] Open
Abstract
B-ring unsubstituted flavones (of which the most widely known are chrysin, baicalein, wogonin, and oroxylin A) are 2-phenylchromen-4-one molecules of which the B-ring is devoid of any hydroxy, methoxy, or other substituent. They may be found naturally in a number of herbal products used for therapeutic purposes, and several have been designed by researchers and obtained in the laboratory. They have generated interest in the scientific community for their potential use in a variety of pathologies, and understanding their pharmacokinetics is important for a grasp of their optimal use. Based on a comprehensive survey of the relevant literature, this paper examines their absorption (with deglycosylation as a preliminary step) and their fate in the body, from metabolism to excretion. Differences among species (inter-individual) and within the same species (intra-individual) variability have been examined based on the available data, and finally, knowledge gaps and directions of future research are discussed.
Collapse
Affiliation(s)
- Robert Ancuceanu
- Department of Pharmaceutical Botany and Cell Biology, Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Mihaela Dinu
- Department of Pharmaceutical Botany and Cell Biology, Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania.
| | - Cristina Dinu-Pirvu
- Department of Physical Chemistry and Colloidal Chemistry, Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 020956 Bucharest 020956, Romania
| | - Valentina Anuţa
- Department of Physical Chemistry and Colloidal Chemistry, Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 020956 Bucharest 020956, Romania
| | - Vlad Negulescu
- Department of Toxicology, Clinical Pharmacology and Psychopharmacology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| |
Collapse
|
19
|
Barbosa ACS, Feng Y, Yu C, Huang M, Xie W. Estrogen sulfotransferase in the metabolism of estrogenic drugs and in the pathogenesis of diseases. Expert Opin Drug Metab Toxicol 2019; 15:329-339. [PMID: 30822161 DOI: 10.1080/17425255.2019.1588884] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Biotransformation is important in the metabolism of endobiotics and xenobiotics. This process comprises the activity of phase I and phase II enzymes. Estrogen sulfotransferase (SULT1E1 or EST) is a phase II conjugating enzyme that belongs to the family of cytosolic sulfotransferases. The expression of SULT1E1 can be detected in many tissues, including the liver. SULT1E1 catalyzes the transfer of a sulfate group from 3'-phosphoadenosine-5'-phosphosulfate (PAPS) to any available hydroxyl group in estrogenic molecules. The substrates of SULT1E1 include the endogenous and synthetic estrogens. Upon SULT1E1-mediated sulfation, the hydrosolubility of estrogens increases, preventing the binding between the sulfated estrogens and the estrogen receptor (ER). This sulfated state of the estrogens is not irreversible, as the steroid sulfatase (STS) can convert sulfoconjugated estrogens to free estrogens. The expression of SULT1E1 is inducible by several diseases that involve tissue inflammation, such as type 2 diabetes, sepsis, and ischemia-reperfusion injury. Areas covered: This systematic literature review aims to summarize the role of SULT1E1 in the metabolism of estrogenic drugs and xenobiotics, and the role of SULT1E1 in the pathogenesis of several diseases, including cancer, metabolic disease, sepsis, liver injury, and cystic fibrosis. Meanwhile, ablation or pharmacological inhibition of SULT1E1 can affect the outcomes of the aforementioned diseases. Expert opinion: In addition to its role in metabolizing estrogenic drugs, SULT1E1 is unexpectedly being unveiled as a mediator for the disease effect on estrogen metabolism and homeostasis. Meanwhile, because the expression and activity of SULT1E1 can affect the outcome of diseases, the same sulfotransferase and the reversing enzymes STS can be potential therapeutic targets to prevent or manage diseases. Accumulating evidence suggest that the physiological and pathophysiological effects of SULT1E1 can be estrogen-independent and it is necessary to elucidate what other possible substrates may be recognized by the enzyme. Moreover, human studies are paramount to confirm the human relevance of the animal studies.
Collapse
Affiliation(s)
- Anne Caroline S Barbosa
- a Center for Pharmacogenetics and Department of Pharmaceutical Sciences , University of Pittsburgh , Pittsburgh , PA , USA
| | - Ye Feng
- a Center for Pharmacogenetics and Department of Pharmaceutical Sciences , University of Pittsburgh , Pittsburgh , PA , USA.,b Department of Endocrinology and Metabolic Disease , The First Affiliated Hospital, Zhejiang University School of Medicine , Hangzhou , China
| | - Chaohui Yu
- c Department of Gastroenterology , The First Affiliated Hospital, Zhejiang University School of Medicine , Hangzhou , China
| | - Min Huang
- d Institute of Clinical Pharmacology and Guangdong Provincial Key Laboratory of New Drug Design and Evaluation , Sun Yat-Sen University , Guangzhou , China
| | - Wen Xie
- a Center for Pharmacogenetics and Department of Pharmaceutical Sciences , University of Pittsburgh , Pittsburgh , PA , USA.,e Department of Pharmacology and Chemical Biology , University of Pittsburgh , Pittsburgh , PA , USA
| |
Collapse
|
20
|
Bousset L, Rambur A, Fouache A, Bunay J, Morel L, Lobaccaro JMA, Baron S, Trousson A, de Joussineau C. New Insights in Prostate Cancer Development and Tumor Therapy: Modulation of Nuclear Receptors and the Specific Role of Liver X Receptors. Int J Mol Sci 2018; 19:E2545. [PMID: 30154328 PMCID: PMC6164771 DOI: 10.3390/ijms19092545] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/08/2018] [Accepted: 08/09/2018] [Indexed: 12/14/2022] Open
Abstract
Prostate cancer (PCa) incidence has been dramatically increasing these last years in westernized countries. Though localized PCa is usually treated by radical prostatectomy, androgen deprivation therapy is preferred in locally advanced disease in combination with chemotherapy. Unfortunately, PCa goes into a castration-resistant state in the vast majority of the cases, leading to questions about the molecular mechanisms involving the steroids and their respective nuclear receptors in this relapse. Interestingly, liver X receptors (LXRα/NR1H3 and LXRβ/NR1H2) have emerged as new actors in prostate physiology, beyond their historical roles of cholesterol sensors. More importantly LXRs have been proposed to be good pharmacological targets in PCa. This rational has been based on numerous experiments performed in PCa cell lines and genetic animal models pointing out that using selective liver X receptor modulators (SLiMs) could actually be a good complementary therapy in patients with a castration resistant PCa. Hence, this review is focused on the interaction among the androgen receptors (AR/NR3C4), estrogen receptors (ERα/NR3A1 and ERβ/NR3A2), and LXRs in prostate homeostasis and their putative pharmacological modulations in parallel to the patients' support.
Collapse
Affiliation(s)
- Laura Bousset
- Université Clermont Auvergne, GReD, CNRS UMR 6293, INSERM U1103, 28, place Henri Dunant, BP38, F63001 Clermont-Ferrand, France.
- Centre de Recherche en Nutrition Humaine d'Auvergne, 58 Boulevard Montalembert, F-63009 Clermont-Ferrand, France.
| | - Amandine Rambur
- Université Clermont Auvergne, GReD, CNRS UMR 6293, INSERM U1103, 28, place Henri Dunant, BP38, F63001 Clermont-Ferrand, France.
- Centre de Recherche en Nutrition Humaine d'Auvergne, 58 Boulevard Montalembert, F-63009 Clermont-Ferrand, France.
| | - Allan Fouache
- Université Clermont Auvergne, GReD, CNRS UMR 6293, INSERM U1103, 28, place Henri Dunant, BP38, F63001 Clermont-Ferrand, France.
- Centre de Recherche en Nutrition Humaine d'Auvergne, 58 Boulevard Montalembert, F-63009 Clermont-Ferrand, France.
| | - Julio Bunay
- Université Clermont Auvergne, GReD, CNRS UMR 6293, INSERM U1103, 28, place Henri Dunant, BP38, F63001 Clermont-Ferrand, France.
- Centre de Recherche en Nutrition Humaine d'Auvergne, 58 Boulevard Montalembert, F-63009 Clermont-Ferrand, France.
| | - Laurent Morel
- Université Clermont Auvergne, GReD, CNRS UMR 6293, INSERM U1103, 28, place Henri Dunant, BP38, F63001 Clermont-Ferrand, France.
- Centre de Recherche en Nutrition Humaine d'Auvergne, 58 Boulevard Montalembert, F-63009 Clermont-Ferrand, France.
| | - Jean-Marc A Lobaccaro
- Université Clermont Auvergne, GReD, CNRS UMR 6293, INSERM U1103, 28, place Henri Dunant, BP38, F63001 Clermont-Ferrand, France.
- Centre de Recherche en Nutrition Humaine d'Auvergne, 58 Boulevard Montalembert, F-63009 Clermont-Ferrand, France.
| | - Silvère Baron
- Université Clermont Auvergne, GReD, CNRS UMR 6293, INSERM U1103, 28, place Henri Dunant, BP38, F63001 Clermont-Ferrand, France.
- Centre de Recherche en Nutrition Humaine d'Auvergne, 58 Boulevard Montalembert, F-63009 Clermont-Ferrand, France.
| | - Amalia Trousson
- Université Clermont Auvergne, GReD, CNRS UMR 6293, INSERM U1103, 28, place Henri Dunant, BP38, F63001 Clermont-Ferrand, France.
- Centre de Recherche en Nutrition Humaine d'Auvergne, 58 Boulevard Montalembert, F-63009 Clermont-Ferrand, France.
| | - Cyrille de Joussineau
- Université Clermont Auvergne, GReD, CNRS UMR 6293, INSERM U1103, 28, place Henri Dunant, BP38, F63001 Clermont-Ferrand, France.
- Centre de Recherche en Nutrition Humaine d'Auvergne, 58 Boulevard Montalembert, F-63009 Clermont-Ferrand, France.
| |
Collapse
|
21
|
Sun P, Mao X, Gao M, Huang M, Chen L, Ruan G, Huang W, Braicu EI, Sehouli J. Novel endocrine therapeutic strategy in endometrial carcinoma targeting estrogen-related receptor α by XCT790 and siRNA. Cancer Manag Res 2018; 10:2521-2535. [PMID: 30127640 PMCID: PMC6089116 DOI: 10.2147/cmar.s168043] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Purpose To explore the targeted therapy of estrogen-related receptor α (ERRα) in endometrial cancer (EC) cells and its potential mechanisms. Methods The mRNA and protein expression levels of ERRα and estrogen receptor α (ERα) were detected by qPCR and Western blotting in RL-952, AN3-CA, HEC-1A, and HEC-1B EC cell lines. After treatment with the ERRα-specific antagonist XCT790 or infection with lentivirus-mediated small interfering RNA (siRNA) targeting the ERRα (siRNA-ERRα), cell proliferation and apoptosis were evaluated by MTS assay and flow cytometry. After treatment with siRNA-ERRα, the expression profiles of transcription factors (TFs) were analyzed by protein/DNA arrays in EC cells. Results The relative mRNA levels of ERRa in RL-952 (1±0.0831) and AN3-CA (1.162±0.0325) were significantly higher than those in HEC-1A (0.3081±0.0339) and HEC-1B (0.1119±0.0091) (P<0.05), and similar results were observed for ERRα protein levels. A higher ratio of ERa/ERRa was observed in ERα-positive RL-952 (10-fold) and ANC-3A (8.5-fold) cells, whereas a lower ratio was observed in ERα-negative HEC-1A (3.75-fold) and HEC-1B cells (0-fold). Both – exogenous XCT790 and endogenous siRNA-ERRα – can decrease the expression of ERRα, thereby inhibiting proliferation but promoting apoptosis in both ERα-positive and -negative EC cells. The XCT790 presented higher proliferation-inhibition and apoptosis rates in the ERα-positive than ERα-negative cells, whereas the siRNA-ERRα exhibited higher proliferation-inhibition and apoptosis rates in the ERα-negative than in ERα-positive cells. In total, 3 upregulated and 17 downregulated TFs were screened out by knocked-down expression of ERRα in all EC cells. Among them, the upregulated TFs organic cation transporter 3/4(Oct3/4), hepatic nuclear factor 4 (HNF4), HNF4 and chicken ovalbumin upstream TF (COUP-TF) as well as downregulated transcription factor EB (TFEB) were found to be statistically significant (P<0.05). Conclusion Targeting ERRα provides a promising novel endocrine therapeutic strategy.
Collapse
Affiliation(s)
- PengMing Sun
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, People's Republic of China,
| | - XiaoDan Mao
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, People's Republic of China,
| | - Min Gao
- Department of Gynecology Oncology, Beijing Cancer Hospital, Beijing 100142, People's Republic of China
| | - MeiMei Huang
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, People's Republic of China,
| | - LiLi Chen
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, People's Republic of China,
| | - GuanYu Ruan
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, People's Republic of China,
| | - WeiYi Huang
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, People's Republic of China,
| | - Elena Ioana Braicu
- Department of Gynecology, Campus Virchow Clinic, Charité Medical University Berlin, Berlin, Germany
| | - Jalid Sehouli
- Department of Gynecology, Campus Virchow Clinic, Charité Medical University Berlin, Berlin, Germany
| |
Collapse
|
22
|
Schuler G, Dezhkam Y, Tenbusch L, Klymiuk MC, Zimmer B, Hoffmann B. SULFATION PATHWAYS: Formation and hydrolysis of sulfonated estrogens in the porcine testis and epididymis. J Mol Endocrinol 2018; 61:M13-M25. [PMID: 29467139 DOI: 10.1530/jme-17-0245] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 02/21/2018] [Indexed: 12/22/2022]
Abstract
Boars exhibit high concentrations of sulfonated estrogens (SE) mainly originating from the testicular-epididymal compartment. Intriguingly, in porcine Leydig cells, sulfonation of estrogens is colocalized with aromatase and steroid sulfatase (STS), indicating that de novo synthesis of unconjugated estrogens (UE), their sulfonation and hydrolysis of SE occur within the same cell type. So far in boars no plausible concept concerning the role of SE has been put forward. To obtain new information on SE formation and hydrolysis, the porcine testicular-epididymal compartment was screened for the expression of the estrogen-specific sulfotransferase SULT1E1 and STS applying real-time RT-qPCR, Western blot and immunohistochemistry. The epididymal head was identified as the major site of SULT1E1 expression, whereas in the testis, it was virtually undetectable. However, SE tissue concentrations are clearly consistent with the testis as the predominant site of estrogen sulfonation. Results from measurements of estrogen sulfotransferase activity indicate that in the epididymis, SULT1E1 is the relevant enzyme, whereas in the testis, estrogens are sulfonated by a different sulfotransferase with a considerably lower affinity. STS expression and activity was high in the testis (Leydig cells, rete testis epithelium) but also present throughout the epididymis. In the epididymis, SULT1E1 and STS were colocalized in the ductal epithelium, and there was evidence for their apocrine secretion into the ductal lumen. The results suggest that in porcine Leydig cells, SE may be produced as a reservoir to support the levels of bioactive UE via the sulfatase pathway during periods of low activity of the pulsatile testicular steroidogenesis.
Collapse
Affiliation(s)
- G Schuler
- Veterinary Clinic for Obstetrics, Gynecology and Andrology, Faculty of Veterinary Medicine, Justus-Liebig-University, Giessen, Germany
| | - Y Dezhkam
- Veterinary Clinic for Obstetrics, Gynecology and Andrology, Faculty of Veterinary Medicine, Justus-Liebig-University, Giessen, Germany
| | - L Tenbusch
- Veterinary Clinic for Obstetrics, Gynecology and Andrology, Faculty of Veterinary Medicine, Justus-Liebig-University, Giessen, Germany
| | - M C Klymiuk
- Veterinary Clinic for Obstetrics, Gynecology and Andrology, Faculty of Veterinary Medicine, Justus-Liebig-University, Giessen, Germany
| | - B Zimmer
- Veterinary Clinic for Obstetrics, Gynecology and Andrology, Faculty of Veterinary Medicine, Justus-Liebig-University, Giessen, Germany
| | - B Hoffmann
- Veterinary Clinic for Obstetrics, Gynecology and Andrology, Faculty of Veterinary Medicine, Justus-Liebig-University, Giessen, Germany
| |
Collapse
|
23
|
Zimmer B, Tenbusch L, Klymiuk MC, Dezhkam Y, Schuler G. SULFATION PATHWAYS: Expression of SULT2A1, SULT2B1 and HSD3B1 in the porcine testis and epididymis. J Mol Endocrinol 2018; 61:M41-M55. [PMID: 29588428 DOI: 10.1530/jme-17-0277] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 03/27/2018] [Indexed: 01/01/2023]
Abstract
In the porcine testis, in addition to estrogen sulfates, the formation of numerous sulfonated neutral hydroxysteroids has been observed. However, their functions and the underlying synthetic pathways are still widely unclear. To obtain further information on their formation in postpubertal boars, the expression of sulfotransferases considered relevant for neutral hydroxysteroids (SULT2A1, SULT2B1) was investigated in the testis and defined segments of the epididymis applying real-time RT-qPCR, Western blot and immunohistochemistry (IHC). Sulfotransferase activities were assessed in tissue homogenates or cytosolic preparations applying dehydroepiandrosterone and pregnenolone as substrates. A high SULT2A1 expression was confirmed in the testis and localized in Leydig cells by IHC. In the epididymis, SULT2A1 expression was virtually confined to the body. SULT2B1 expression was absent or low in the testis but increased significantly along the epididymis. Immunohistochemical observations indicate that both enzymes are secreted into the ductal lumen via an apocrine mechanism. The results from the characterization of expression patterns and activity measurements suggest that SULT2A1 is the prevailing enzyme for the sulfonation of hydroxysteroids in the testis, whereas SULT2B1 may catalyze the formation of sterol sulfates in the epididymis. In order to obtain information on the overall steroidogenic capacity of the porcine epididymis, the expression of important steroidogenic enzymes (CYP11A1, CYP17A1, CYP19, HSD3B1, HSD17B3, SRD5A2) was monitored in the defined epididymal segments applying real-time RT-qPCR. Surprisingly, in addition to a high expression of SRD5A2 in the epididymal head, a substantial expression of HSD3B1 was detected, which increased along the organ.
Collapse
Affiliation(s)
- B Zimmer
- Veterinary Clinic for Obstetrics, Gynecology and Andrology, Justus-Liebig-University, Giessen, Germany
| | - L Tenbusch
- Veterinary Clinic for Obstetrics, Gynecology and Andrology, Justus-Liebig-University, Giessen, Germany
| | - M C Klymiuk
- Veterinary Clinic for Obstetrics, Gynecology and Andrology, Justus-Liebig-University, Giessen, Germany
| | - Y Dezhkam
- Veterinary Clinic for Obstetrics, Gynecology and Andrology, Justus-Liebig-University, Giessen, Germany
| | - G Schuler
- Veterinary Clinic for Obstetrics, Gynecology and Andrology, Justus-Liebig-University, Giessen, Germany
| |
Collapse
|
24
|
Poschner S, Maier-Salamon A, Zehl M, Wackerlig J, Dobusch D, Meshcheryakova A, Mechtcheriakova D, Thalhammer T, Pachmann B, Jäger W. Resveratrol Inhibits Key Steps of Steroid Metabolism in a Human Estrogen-Receptor Positive Breast Cancer Model: Impact on Cellular Proliferation. Front Pharmacol 2018; 9:742. [PMID: 30042681 PMCID: PMC6048268 DOI: 10.3389/fphar.2018.00742] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 06/19/2018] [Indexed: 12/12/2022] Open
Abstract
The role of resveratrol (RES) in preventing breast cancer is controversial, as low concentrations may stimulate the proliferation of estrogen-receptor alpha positive (ERα+) breast cancer cells. As metabolism is the key factor in altering cellular estrogens, thereby influencing breast tumor growth, we investigated the effects of RES on the formation of estrogen metabolites, namely 4-androstene-3,17-dione (AD), dehydroepiandrosterone (DHEA), dehydroepiandrosterone-3-O-sulfate (DHEA-S), estrone (E1), estrone-3-sulfate (E1-S), 17β-estradiol (E2), 17β-estradiol-3-O-(β-D-glucuronide) (E2-G), 17β-estradiol-3-O-sulfate (E2-S), 16α-hydroxy-17β-estradiol (estriol, E3), and testosterone (T) in ERα- MDA-MB-231 and ERα+ MCF-7 cells. Incubation of both of the cell lines with the hormone precursors DHEA and E1 revealed that sulfation and glucuronidation were preferred metabolic pathways for DHEA, E1 and E2 in MCF-7 cells, compared with in MDA-MB-231 cells, as the Vmax values were significantly higher (DHEA-S: 2873.0 ± 327.4 fmol/106 cells/h, E1-S: 30.4 ± 2.5 fmol/106 cells/h, E2-S: 24.7 ± 4.9 fmol/106 cells/h, E2-G: 7.29 ± 1.36 fmol/106 cells/h). RES therefore significantly inhibited DHEA-S, E1-S, E2-S and E2-G formation in MCF-7, but not in MDA-MB-231 cells (Kis: E2-S, 0.73 ± 0.07 μM < E1-S, 0.94 ± 0.03 μM < E2-G, 7.92 ± 0.24 μM < DHEA-S, 13.2 ± 0.2 μM). Suppression of these metabolites subsequently revealed twofold higher levels of active E2, concomitant with an almost twofold increase in MCF-7 cell proliferation, which was the most pronounced upon the addition of 5 μM RES. As the content of RES in food is relatively low, an increased risk of breast cancer progression in women is likely to only be observed following the continuous consumption of high-dose RES supplements. Further long-term human studies simultaneously monitoring free estrogens and their conjugates are therefore highly warranted to evaluate the efficacy and safety of RES supplementation, particularly in patients diagnosed with ERα+ breast cancer.
Collapse
Affiliation(s)
- Stefan Poschner
- Division of Clinical Pharmacy and Diagnostics, Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Alexandra Maier-Salamon
- Division of Clinical Pharmacy and Diagnostics, Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Martin Zehl
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Judith Wackerlig
- Division of Drug Design and Medicinal Chemistry, Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Daniel Dobusch
- Division of Drug Design and Medicinal Chemistry, Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Anastasia Meshcheryakova
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Diana Mechtcheriakova
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Theresia Thalhammer
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Bettina Pachmann
- Division of Clinical Pharmacy and Diagnostics, Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Walter Jäger
- Division of Clinical Pharmacy and Diagnostics, Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria.,Vienna Metabolomics Center, University of Vienna, Vienna, Austria
| |
Collapse
|
25
|
Klymiuk MC, Neunzig J, Bernhardt R, Sánchez-Guijo A, Hartmann MF, Wudy SA, Schuler G. Efficiency of the sulfate pathway in comparison to the Δ4- and Δ5-pathway of steroidogenesis in the porcine testis. J Steroid Biochem Mol Biol 2018; 179:64-72. [PMID: 29107177 DOI: 10.1016/j.jsbmb.2017.10.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 10/12/2017] [Accepted: 10/24/2017] [Indexed: 12/20/2022]
Abstract
Sulfonated steroids are increasingly recognized as a circulating reservoir of precursors for the local production of active steroids in certain target tissues. As an alternative to sulfonation of unconjugated steroids by cytosolic sulfotransferases, their direct formation from sulfonated precursors has been described. However, productivity and physiological relevance of this sulfate pathway of steroidogenesis are still widely unclear. Applying the porcine testis as a model, conversion of pregnenolone sulfate (P5S, sulfate pathway) by CYP17A1 was assessed in comparison to the parallel conversions of pregnenolone (P5, Δ5-pathway) and progesterone (P4, Δ4-pathway). To characterize conversions in the virtual absence of competing enzyme activities, in a first series of experiments porcine recombinant CYP17A1 was incubated with the respective substrate in the presence of bovine recombinant cytochrome P450 oxidoreductase (CPR) and cytochrome b5 (b5). Moreover, porcine testicular microsomal fractions were used as a source of homologous CYP17A1, CPR and b5. Invariably 17α-hydroxylation of P5S was, if at all, only minimal and no formation of dehydroepiandrosterone sulfate from P5S was detectable. Consistent with earlier studies porcine CYP17A1 efficiently metabolized P4 and P5 in both assay systems. Metabolism of P4 and P5 by testicular microsomal protein varied substantially between the five animals tested. In conclusion, a physiologically relevant sulfate pathway for the production of C19-steroids from P5S via CYP17A1 is very unlikely in the porcine testis.
Collapse
Affiliation(s)
- M C Klymiuk
- Veterinary Clinic for Obstetrics, Gynecology and Andrology, Justus-Liebig-University, Giessen, Germany
| | - J Neunzig
- Department of Biochemistry, Faculty of Technical and Natural Sciences III, Saarland University, Saarbruecken, Germany
| | - R Bernhardt
- Department of Biochemistry, Faculty of Technical and Natural Sciences III, Saarland University, Saarbruecken, Germany
| | - A Sánchez-Guijo
- Steroid Research & Mass Spectrometry Unit, Division of Pediatric Endocrinology & Diabetology, Center of Child and Adolescent Medicine, Justus-Liebig University, Giessen, Germany
| | - M F Hartmann
- Steroid Research & Mass Spectrometry Unit, Division of Pediatric Endocrinology & Diabetology, Center of Child and Adolescent Medicine, Justus-Liebig University, Giessen, Germany
| | - S A Wudy
- Steroid Research & Mass Spectrometry Unit, Division of Pediatric Endocrinology & Diabetology, Center of Child and Adolescent Medicine, Justus-Liebig University, Giessen, Germany
| | - G Schuler
- Veterinary Clinic for Obstetrics, Gynecology and Andrology, Justus-Liebig-University, Giessen, Germany.
| |
Collapse
|
26
|
Blaschka C, Schuler G, Sánchez-Guijo A, Zimmer B, Feller S, Kotarski F, Wudy SA, Wrenzycki C. Occurrence of sulfonated steroids and ovarian expression of steroid sulfatase and SULT1E1 in cyclic cows. J Steroid Biochem Mol Biol 2018; 179:79-87. [PMID: 29262378 DOI: 10.1016/j.jsbmb.2017.12.010] [Citation(s) in RCA: 5] [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: 04/10/2017] [Revised: 12/07/2017] [Accepted: 12/14/2017] [Indexed: 12/28/2022]
Abstract
Historically sulfonated steroids were primarily considered as inactive metabolites destined for elimination. However, more recently they have been increasingly recognized as precursors for the production of bioactive steroids in target tissues and as functional molecules without preceding hydrolysis. In order to comprehensively characterize their occurrence in cyclic cows and their formation and hydrolysis in bovine ovarian steroidogenesis, ovaries from cyclic cows were screened for the expression of oestrogen sulfotransferase (SULTE1) and steroid sulfatase (STS) by Western blot and immunohistochemistry. Moreover, a broad spectrum of 13 sulfonated steroids was measured applying liquid chromatography-tandem mass spectrometry (LC-MS/MS) in blood samples collected from three cycling heifers during defined stages of the ovarian cycle and in fluid obtained from ovarian follicles of different size. SULT1E1 was undetectable in ovarian tissues. For STS only a weak immunostaining was found predominantly in granulosa cells of larger follicles. However, no specific band occurred in Western blot. In blood, concentrations of all sulfonated steroids investigated were below the limit of quantification (LOQ). In follicular fluid, only cholesterol sulfate was measured in considerable concentrations (328.3 ± 63.8 ng/ml). However, the role of cholesterol sulfate in bovine follicular steroidogenesis remains unclear as concentrations were obviously unrelated to follicular size. The remaining sulfonated steroids investigated were undetectable or only slightly exceeded LOQ in a minor proportion of samples. The results are clearly contrary to a role of sulfonated steroids as important precursors, intermediates or products of bovine ovarian steroidogenesis.
Collapse
Affiliation(s)
- Carina Blaschka
- Clinic for Veterinary Obstetrics, Gynaecology and Andrology, Department of Molecular Reproductive Medicine, Faculty of Veterinary Medicine, Justus-Liebig-University Giessen, Frankfurter Strasse 106, D-35392 Giessen, Germany
| | - Gerhard Schuler
- Clinic for Veterinary Obstetrics, Gynaecology and Andrology, Department of Molecular Reproductive Medicine, Faculty of Veterinary Medicine, Justus-Liebig-University Giessen, Frankfurter Strasse 106, D-35392 Giessen, Germany
| | - Alberto Sánchez-Guijo
- Steroid Research and Mass Spectrometry Unit, Center of Child and Adolescent Medicine, Faculty of Medicine, Justus-Liebig-University Giessen, Feulgenstrasse 10-12, D-35392 Giessen, Germany
| | - Bettina Zimmer
- Clinic for Veterinary Obstetrics, Gynaecology and Andrology, Department of Molecular Reproductive Medicine, Faculty of Veterinary Medicine, Justus-Liebig-University Giessen, Frankfurter Strasse 106, D-35392 Giessen, Germany
| | - Sabine Feller
- Clinic for Veterinary Obstetrics, Gynaecology and Andrology, Department of Molecular Reproductive Medicine, Faculty of Veterinary Medicine, Justus-Liebig-University Giessen, Frankfurter Strasse 106, D-35392 Giessen, Germany
| | - Franziska Kotarski
- Clinic for Veterinary Obstetrics, Gynaecology and Andrology, Department of Molecular Reproductive Medicine, Faculty of Veterinary Medicine, Justus-Liebig-University Giessen, Frankfurter Strasse 106, D-35392 Giessen, Germany
| | - Stefan A Wudy
- Steroid Research and Mass Spectrometry Unit, Center of Child and Adolescent Medicine, Faculty of Medicine, Justus-Liebig-University Giessen, Feulgenstrasse 10-12, D-35392 Giessen, Germany
| | - Christine Wrenzycki
- Clinic for Veterinary Obstetrics, Gynaecology and Andrology, Department of Molecular Reproductive Medicine, Faculty of Veterinary Medicine, Justus-Liebig-University Giessen, Frankfurter Strasse 106, D-35392 Giessen, Germany.
| |
Collapse
|
27
|
Xu Y, Lin X, Xu J, Jing H, Qin Y, Li Y. SULT1E1 inhibits cell proliferation and invasion by activating PPARγ in breast cancer. J Cancer 2018; 9:1078-1087. [PMID: 29581787 PMCID: PMC5868175 DOI: 10.7150/jca.23596] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/29/2018] [Indexed: 01/08/2023] Open
Abstract
Sulfotransferase family 1E member 1 (SULT1E1) is known to catalyze sulfoconjugation and play a crucial role in the deactivation of estrogen homeostasis, which is involved in tumorigenesis and the progression of breast and endometrial cancers. Our previous study has shown that the protein levels of SULT1E1 were decreased in breast cancer; however, the underlying mechanism is still poorly understood. In this study, we explored the functional and molecular mechanisms by which SULT1E1 influenced breast cancer. Here, we identified that overexpression of SULT1E1 inhibited breast cancer cell growth through inducing apoptosis and arresting cell cycle progression. Furthermore, enforced expression of SULT1E1 suppressed tumor cell migration and invasion. Moreover, we found that the activation of PPARγ was required for SULT1E1-mediated downregulation of C-myc, Cyclin D1, MMP-2 and MMP-9 as well as for cell apoptosis, migration and invasion. In addition, the overexpression of SULT1E1 significantly inhibited tumor growth in vivo. Taken together, our findings indicated that SULT1E1 performed its tumor suppressor characteristics by activating PPARγ, which provided a novel target for patients with breast cancer.
Collapse
Affiliation(s)
- Yali Xu
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, P.R. China
| | - Xiaoyan Lin
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, P.R. China
| | - Jiawen Xu
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, P.R. China
| | - Haiyan Jing
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, P.R. China
| | - Yejun Qin
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, P.R. China
| | - Yintao Li
- Department of Medical Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, P.R. China
| |
Collapse
|
28
|
Poschner S, Maier-Salamon A, Zehl M, Wackerlig J, Dobusch D, Pachmann B, Sterlini KL, Jäger W. The Impacts of Genistein and Daidzein on Estrogen Conjugations in Human Breast Cancer Cells: A Targeted Metabolomics Approach. Front Pharmacol 2017; 8:699. [PMID: 29051735 PMCID: PMC5633874 DOI: 10.3389/fphar.2017.00699] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 09/19/2017] [Indexed: 11/24/2022] Open
Abstract
The beneficial effect of dietary soy food intake, especially for women diagnosed with breast cancer, is controversial, as in vitro data has shown that the soy isoflavones genistein and daidzein may even stimulate the proliferation of estrogen-receptor alpha positive (ERα+) breast cancer cells at low concentrations. As genistein and daidzein are known to inhibit key enzymes in the steroid metabolism pathway, and thus may influence levels of active estrogens, we investigated the impacts of genistein and daidzein on the formation of estrogen metabolites, namely 17β-estradiol (E2), 17β-estradiol-3-(β-D-glucuronide) (E2-G), 17β-estradiol-3-sulfate (E2-S) and estrone-3-sulfate (E1-S) in estrogen-dependent ERα+ MCF-7 cells. We found that both isoflavones were potent inhibitors of E1 and E2 sulfation (85–95% inhibition at 10 μM), but impeded E2 glucuronidation to a lesser extent (55–60% inhibition at 10 μM). The stronger inhibition of E1 and E2 sulfation compared with E2 glucuronidation was more evident for genistein, as indicated by significantly lower inhibition constants for genistein [Kis: E2-S (0.32 μM) < E1-S (0.76 μM) < E2-G (6.01 μM)] when compared with those for daidzein [Kis: E2-S (0.48 μM) < E1-S (1.64 μM) < E2-G (7.31 μM)]. Concomitant with the suppression of E1 and E2 conjugation, we observed a minor but statistically significant increase in E2 concentration of approximately 20%. As the content of genistein and daidzein in soy food is relatively low, an increased risk of breast cancer development and progression in women may only be observed following consumption of high-dose isoflavone supplements. Further long-term human studies monitoring free estrogens and their conjugates are therefore highly warranted to evaluate the potential side effects of high-dose genistein and daidzein, especially in patients diagnosed with ERα+ breast cancer.
Collapse
Affiliation(s)
- Stefan Poschner
- Division of Clinical Pharmacy and Diagnostics, Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Alexandra Maier-Salamon
- Division of Clinical Pharmacy and Diagnostics, Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Martin Zehl
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Judith Wackerlig
- Division of Drug Design and Medicinal Chemistry, Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Daniel Dobusch
- Division of Drug Design and Medicinal Chemistry, Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Bettina Pachmann
- Division of Clinical Pharmacy and Diagnostics, Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Konstantin L Sterlini
- Division of Clinical Pharmacy and Diagnostics, Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Walter Jäger
- Division of Clinical Pharmacy and Diagnostics, Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria.,Vienna Metabolomics Center (VIME), University of Vienna, Vienna, Austria
| |
Collapse
|
29
|
van Duursen MBM. Modulation of estrogen synthesis and metabolism by phytoestrogens in vitro and the implications for women's health. Toxicol Res (Camb) 2017; 6:772-794. [PMID: 30090542 DOI: 10.1039/c7tx00184c] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 09/07/2017] [Indexed: 12/12/2022] Open
Abstract
Phytoestrogens are increasingly used as dietary supplements due to their suggested health promoting properties, but also by women for breast enhancement and relief of menopausal symptoms. Generally, phytoestrogens are considered to exert estrogenic activity via estrogen receptors (ERs), but they may also affect estrogen synthesis and metabolism locally in breast, endometrial and ovarian tissues. Considering that accurate regulation of local hormone levels is crucial for normal physiology, it is not surprising that interference with hormonal synthesis and metabolism is associated with a wide variety of women's health problems, varying from altered menstrual cycle to hormone-dependent cancers. Yet, studies on phytoestrogens have mainly focused on ER-mediated effects of soy-derived phytoestrogens, with less attention paid to steroid synthesis and metabolism or other phytoestrogens. This review aims to evaluate the potential of phytoestrogens to modulate local estrogen levels and the implications for women's health. For that, an overview is provided of the effects of commonly used phytoestrogens, i.e. 8-prenylnaringenin, biochanin A, daidzein, genistein, naringenin, resveratrol and quercetin, on estrogen synthesizing and metabolizing enzymes in vitro. The potential implications for women's health are assessed by comparing the in vitro effect concentrations with blood concentrations that can be found after intake of these phytoestrogens. Based on this evaluation, it can be concluded that high-dose supplements with phytoestrogens might affect breast and endometrial health or fertility in women via the modulation of steroid hormone levels. However, more data regarding the tissue levels of phytoestrogens and effect data from dedicated, tissue-specific assays are needed for a better understanding of potential risks. At least until more certainty regarding the safety has been established, especially young women would better avoid using supplements containing high doses of phytoestrogens.
Collapse
Affiliation(s)
- Majorie B M van Duursen
- Research group Endocrine Toxicology , Institute for Risk Assessment Sciences , Faculty of Veterinary Medicine , Utrecht University , Yalelaan 104 , 3584 CM , Utrecht , the Netherlands . ; Tel: +31 (0)30 253 5398
| |
Collapse
|
30
|
Geyer J, Bakhaus K, Bernhardt R, Blaschka C, Dezhkam Y, Fietz D, Grosser G, Hartmann K, Hartmann MF, Neunzig J, Papadopoulos D, Sánchez-Guijo A, Scheiner-Bobis G, Schuler G, Shihan M, Wrenzycki C, Wudy SA, Bergmann M. The role of sulfated steroid hormones in reproductive processes. J Steroid Biochem Mol Biol 2017; 172:207-221. [PMID: 27392637 DOI: 10.1016/j.jsbmb.2016.07.002] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 07/04/2016] [Indexed: 10/21/2022]
Abstract
Sulfated steroid hormones, such as dehydroepiandrosterone sulfate or estrone-3-sulfate, have long been regarded as inactive metabolites as they cannot activate classical steroid receptors. Some of them are present in the blood circulation at quite high concentrations, but generally sulfated steroids exhibit low membrane permeation due to their hydrophilic properties. However, sulfated steroid hormones can actively be imported into specific target cells via uptake carriers, such as the sodium-dependent organic anion transporter SOAT, and, after hydrolysis by the steroid sulfatase (so-called sulfatase pathway), contribute to the overall regulation of steroid responsive organs. To investigate the biological significance of sulfated steroid hormones for reproductive processes in humans and animals, the research group "Sulfated Steroids in Reproduction" was established by the German Research Foundation DFG (FOR1369). Projects of this group deal with transport of sulfated steroids, sulfation of free steroids, desulfation by the steroid sulfatase, effects of sulfated steroids on steroid biosynthesis and membrane receptors as well as MS-based profiling of sulfated steroids in biological samples. This review and concept paper presents key findings from all these projects and provides a broad overview over the current research on sulfated steroid hormones in the field of reproduction.
Collapse
Affiliation(s)
- Joachim Geyer
- Institute of Pharmacology and Toxicology, Justus Liebig University, Giessen, Germany.
| | - Katharina Bakhaus
- Institute of Pharmacology and Toxicology, Justus Liebig University, Giessen, Germany
| | - Rita Bernhardt
- Institute of Biochemistry, Saarland University, Saarbrücken, Germany
| | - Carina Blaschka
- Veterinary Clinic for Obstetrics, Gynecology and Andrology, Justus Liebig University, Giessen, Germany
| | - Yaser Dezhkam
- Veterinary Clinic for Obstetrics, Gynecology and Andrology, Justus Liebig University, Giessen, Germany
| | - Daniela Fietz
- Department of Veterinary Anatomy, Histology and Embryology, Justus Liebig University, Giessen, Germany
| | - Gary Grosser
- Institute of Pharmacology and Toxicology, Justus Liebig University, Giessen, Germany
| | - Katja Hartmann
- Department of Veterinary Anatomy, Histology and Embryology, Justus Liebig University, Giessen, Germany
| | - Michaela F Hartmann
- Steroid Research & Mass Spectrometry Unit, Laboratory for Translational Hormone Analytics, Pediatric Endocrinology & Diabetology, Center of Child and Adolescent Medicine, Justus Liebig University, Giessen, Germany
| | - Jens Neunzig
- Institute of Biochemistry, Saarland University, Saarbrücken, Germany
| | - Dimitrios Papadopoulos
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University, Giessen, Germany
| | - Alberto Sánchez-Guijo
- Steroid Research & Mass Spectrometry Unit, Laboratory for Translational Hormone Analytics, Pediatric Endocrinology & Diabetology, Center of Child and Adolescent Medicine, Justus Liebig University, Giessen, Germany
| | - Georgios Scheiner-Bobis
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University, Giessen, Germany
| | - Gerhard Schuler
- Veterinary Clinic for Obstetrics, Gynecology and Andrology, Justus Liebig University, Giessen, Germany
| | - Mazen Shihan
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University, Giessen, Germany
| | - Christine Wrenzycki
- Veterinary Clinic for Obstetrics, Gynecology and Andrology, Justus Liebig University, Giessen, Germany
| | - Stefan A Wudy
- Steroid Research & Mass Spectrometry Unit, Laboratory for Translational Hormone Analytics, Pediatric Endocrinology & Diabetology, Center of Child and Adolescent Medicine, Justus Liebig University, Giessen, Germany
| | - Martin Bergmann
- Department of Veterinary Anatomy, Histology and Embryology, Justus Liebig University, Giessen, Germany
| |
Collapse
|
31
|
Rezvanpour A, Don-Wauchope AC. Clinical implications of estrone sulfate measurement in laboratory medicine. Crit Rev Clin Lab Sci 2016; 54:73-86. [DOI: 10.1080/10408363.2016.1252310] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Atoosa Rezvanpour
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario,Canada and
| | - Andrew C. Don-Wauchope
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario,Canada and
- Hamilton Regional Laboratory Medicine Program, Hamilton, Ontario, Canada
| |
Collapse
|
32
|
Wang LJ, Li J, Hao FR, Yuan Y, Li JY, Lu W, Zhou TY. Dexamethasone suppresses the growth of human non-small cell lung cancer via inducing estrogen sulfotransferase and inactivating estrogen. Acta Pharmacol Sin 2016; 37:845-56. [PMID: 27133297 DOI: 10.1038/aps.2016.39] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 02/17/2016] [Indexed: 12/12/2022] Open
Abstract
AIM Dexamethasone (DEX) is a widely used synthetic glucocorticoid, which has shown anti-cancer efficacy and anti-estrogenic activity. In this study we explored the possibility that DEX might be used as an endocrine therapeutic agent to treat human non-small cell lung cancer (NSCLC). METHODS The viability and proliferation of human NSCLC cell lines A549 and H1299 were assessed in vitro. Anti-tumor action was also evaluated in A549 xenograft nude mice treated with DEX (2 or 4 mg·kg(-1)·d(-1), ig) or the positive control tamoxifen (50 mg·kg(-1)·d(-1), ig) for 32 d. The expression of estrogen sulfotransferase (EST) in tumor cells and tissues was examined. The intratumoral estrogen levels and uterine estrogen responses were measured. RESULTS DEX displayed mild cytotoxicity to the NSCLC cells (IC50 >500 μmol/L) compared to tamoxifen (IC50 <50 μmol/L), but it was able to inhibit the cell proliferation at low micromolar ranges. Furthermore, DEX (0.1-10 μmol/L) dose-dependently up-regulated EST expression in the cells, and inhibited the cell migration in vitro. Triclosan, a sulfation inhibitor, was able to diminish DEX-caused inhibition on the cell viability. In A549 xenograft nude mice, DEX or tamoxifen administration remarkably suppressed the tumor growth. Moreover, DEX administration dose-dependently increased EST expression in tumor tissues, and reduced intratumoral estrogen levels as well as the volumes and weights of uterine. CONCLUSION DEX suppresses the growth of A549 xenograft tumors via inducing EST and decreasing estradiol levels in tumor tissues, suggesting that DEX may be used as anti-estrogenic agent for the treatment of NSCLC.
Collapse
|
33
|
Rižner TL. The Important Roles of Steroid Sulfatase and Sulfotransferases in Gynecological Diseases. Front Pharmacol 2016; 7:30. [PMID: 26924986 PMCID: PMC4757672 DOI: 10.3389/fphar.2016.00030] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 02/03/2016] [Indexed: 01/08/2023] Open
Abstract
Gynecological diseases such as endometriosis, adenomyosis and uterine fibroids, and gynecological cancers including endometrial cancer and ovarian cancer, affect a large proportion of women. These diseases are estrogen dependent, and their progression often depends on local estrogen formation. In peripheral tissues, estrogens can be formed from the inactive precursors dehydroepiandrosterone sulfate and estrone sulfate. Sulfatase and sulfotransferases have pivotal roles in these processes, where sulfatase hydrolyzes estrone sulfate to estrone, and dehydroepiandrosterone sulfate to dehydroepiandrosterone, and sulfotransferases catalyze the reverse reactions. Further activation of estrone to the most potent estrogen, estradiol, is catalyzed by 17-ketosteroid reductases, while estradiol can also be formed from dehydroepiandrosterone by the sequential actions of 3β-hydroxysteroid dehydrogenase-Δ4-isomerase, aromatase, and 17-ketosteroid reductase. This review introduces the sulfatase and sulfotransferase enzymes, in terms of their structures and reaction mechanisms, and the regulation and different transcripts of their genes, together with the importance of their currently known single nucleotide polymorphisms. Data on expression of sulfatase and sulfotransferases in gynecological diseases are also reviewed. There are often unchanged mRNA and protein levels in diseased tissue, with higher sulfatase activities in cancerous endometrium, ovarian cancer cell lines, and adenomyosis. This can be indicative of a disturbed balance between the sulfatase and sulfotransferases enzymes, defining the potential for sulfatase as a drug target for treatment of gynecological diseases. Finally, clinical trials with sulfatase inhibitors are discussed, where two inhibitors have already concluded phase II trials, although so far with no convincing clinical outcomes for patients with endometrial cancer and endometriosis.
Collapse
Affiliation(s)
- Tea Lanišnik Rižner
- Faculty of Medicine, Institute of Biochemistry, University of Ljubljana Ljubljana, Slovenia
| |
Collapse
|
34
|
Rakers C, Schumacher F, Meinl W, Glatt H, Kleuser B, Wolber G. In Silico Prediction of Human Sulfotransferase 1E1 Activity Guided by Pharmacophores from Molecular Dynamics Simulations. J Biol Chem 2015; 291:58-71. [PMID: 26542807 DOI: 10.1074/jbc.m115.685610] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Indexed: 11/06/2022] Open
Abstract
Acting during phase II metabolism, sulfotransferases (SULTs) serve detoxification by transforming a broad spectrum of compounds from pharmaceutical, nutritional, or environmental sources into more easily excretable metabolites. However, SULT activity has also been shown to promote formation of reactive metabolites that may have genotoxic effects. SULT subtype 1E1 (SULT1E1) was identified as a key player in estrogen homeostasis, which is involved in many physiological processes and the pathogenesis of breast and endometrial cancer. The development of an in silico prediction model for SULT1E1 ligands would therefore support the development of metabolically inert drugs and help to assess health risks related to hormonal imbalances. Here, we report on a novel approach to develop a model that enables prediction of substrates and inhibitors of SULT1E1. Molecular dynamics simulations were performed to investigate enzyme flexibility and sample protein conformations. Pharmacophores were developed that served as a cornerstone of the model, and machine learning techniques were applied for prediction refinement. The prediction model was used to screen the DrugBank (a database of experimental and approved drugs): 28% of the predicted hits were reported in literature as ligands of SULT1E1. From the remaining hits, a selection of nine molecules was subjected to biochemical assay validation and experimental results were in accordance with the in silico prediction of SULT1E1 inhibitors and substrates, thus affirming our prediction hypotheses.
Collapse
Affiliation(s)
- Christin Rakers
- From the Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2/4, 14195 Berlin
| | - Fabian Schumacher
- the Department of Toxicology, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, and the Department of Molecular Biology, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Walter Meinl
- the Departments of Molecular Toxicology and Nutritional Toxicology, German Institute of Human Nutrition (DIfE) Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal
| | - Hansruedi Glatt
- Nutritional Toxicology, German Institute of Human Nutrition (DIfE) Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal
| | - Burkhard Kleuser
- the Department of Toxicology, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, and
| | - Gerhard Wolber
- From the Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2/4, 14195 Berlin,
| |
Collapse
|
35
|
Yin Q, Fischer L, Noethling C, Schaefer WR. In vitro-assessment of putative antiprogestin activities of phytochemicals and synthetic UV absorbers in human endometrial Ishikawa cells. Gynecol Endocrinol 2015; 31:578-81. [PMID: 26036805 DOI: 10.3109/09513590.2015.1047448] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Critical steps of embryo implantation are controlled by progesterone. These processes can be interrupted by progesterone receptor (PR) antagonists, e.g. drugs used for abortion. Antiprogestin effects induced by natural compounds and environmental chemicals have been rarely addressed. In our in vitro study, we investigated putative antiprogestin activities of the plant compounds apigenin (API) and trans-ferulic acid (t-FA) as well as the UV absorbers octyl methoxycinnamate (OMC) and 4-methylbenzylidene camphor (4-MBC). They were compared with the selective progesterone receptor modulators (SPRMs) mifepristone (RU486) and ulipristal acetate (UPA) as well as the full PR-antagonist ZK137316. Effects of test compounds in combination with progesterone on the progesterone-sensitive target gene estrogen sulfotransferase (SULT1E1) were characterized by sigmoidal concentration-response curves obtained by RT-qPCR. The agonistic effect of progesterone on SULT1E1 mRNA levels was concentration-dependently antagonized by RU486, UPA and ZK137316 as well as, with lower potency, apigenin. t-FA, OMC and 4-MBC had no effect on SULT1E1 mRNA levels. We demonstrated that apigenin, although at higher concentrations, exerts a similar effect as the well-characterized SPRMs RU486 and UPA or the progesterone antagonist ZK137316 in this model. Our endometrium-specific Ishikawa cell assay is a useful complement to artificial transactivation assays for the identification of environmental substances with antiprogestin activities.
Collapse
Affiliation(s)
- Qinan Yin
- a Department of Obstetrics & Gynecology , University Medical Center , Freiburg , Germany
| | | | | | | |
Collapse
|
36
|
Guo Y, Hu B, Huang H, Tsung A, Gaikwad NW, Xu M, Jiang M, Ren S, Fan J, Billiar TR, Huang M, Xie W. Estrogen Sulfotransferase Is an Oxidative Stress-responsive Gene That Gender-specifically Affects Liver Ischemia/Reperfusion Injury. J Biol Chem 2015; 290:14754-64. [PMID: 25922074 DOI: 10.1074/jbc.m115.642124] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Indexed: 01/08/2023] Open
Abstract
Estrogen sulfotransferase (EST) regulates estrogen homeostasis by sulfonating and deactivating estrogens. Liver ischemia and reperfusion (I/R) involves both hypoxia during the ischemic phase and oxidative damage during the reperfusion phase. In this report, we showed that the expression of EST was markedly induced by I/R. Mechanistically, oxidative stress-induced activation of Nrf2 was responsible for the EST induction, which was abolished in Nrf2(-/-) mice. EST is a direct transcriptional target of Nrf2. In female mice, the I/R-responsive induction of EST compromised estrogen activity. EST ablation attenuated I/R injury as a result of decreased estrogen deprivation, whereas this benefit was abolished upon ovariectomy. The effect of EST ablation was sex-specific because the EST(-/-) males showed heightened I/R injury. Reciprocally, both estrogens and EST regulate the expression and activity of Nrf2. Estrogen deprivation by ovariectomy abolished the I/R-responsive Nrf2 accumulation, whereas the compromised estrogen deprivation in EST(-/-) mice was associated with increased Nrf2 accumulation. Our results suggested a novel I/R-responsive feedback mechanism to limit the activity of Nrf2 in which Nrf2 induces the expression of EST, which subsequently increases estrogen deactivation and limits the estrogen-responsive activation of Nrf2. Inhibition of EST, at least in females, may represent an effective approach to manage hepatic I/R injury.
Collapse
Affiliation(s)
- Yan Guo
- From the Center for Pharmacogenetics and Department of Pharmaceutical Sciences, the Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China 200025
| | - Bingfang Hu
- From the Center for Pharmacogenetics and Department of Pharmaceutical Sciences, the Institute of Clinical Pharmacology, Sun Yat-Sen University, Guangzhou, China 510275
| | | | | | - Nilesh W Gaikwad
- the Department of Nutrition and Department of Environmental Toxicology, University of California, Davis, California 95616, and
| | - Meishu Xu
- From the Center for Pharmacogenetics and Department of Pharmaceutical Sciences
| | - Mengxi Jiang
- From the Center for Pharmacogenetics and Department of Pharmaceutical Sciences
| | - Songrong Ren
- From the Center for Pharmacogenetics and Department of Pharmaceutical Sciences
| | - Jie Fan
- Surgical Research, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania 15240
| | | | - Min Huang
- the Institute of Clinical Pharmacology, Sun Yat-Sen University, Guangzhou, China 510275
| | - Wen Xie
- From the Center for Pharmacogenetics and Department of Pharmaceutical Sciences, Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261,
| |
Collapse
|
37
|
Squirewell EJ, Duffel MW. The effects of endoxifen and other major metabolites of tamoxifen on the sulfation of estradiol catalyzed by human cytosolic sulfotransferases hSULT1E1 and hSULT1A1*1. Drug Metab Dispos 2015; 43:843-50. [PMID: 25819444 DOI: 10.1124/dmd.115.063206] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 03/27/2015] [Indexed: 11/22/2022] Open
Abstract
Tamoxifen is successfully used for both treatment and prevention of estrogen-dependent breast cancer, yet side effects and development of resistance remain problematic. Endoxifen is a major active metabolite of tamoxifen that is being investigated for clinical use. We hypothesized that endoxifen and perhaps other major metabolites of tamoxifen may affect the ability of human estrogen sulfotransferase 1E1 (hSULT1E1) and human phenol sulfotransferase 1A1 isoform 1 (hSULT1A1*1) to catalyze the sulfation of estradiol, an important mechanism in termination of estrogen signaling through loss of activity at estrogen receptors. Our results indicated that endoxifen, N-desmethyltamoxifen (N-desTAM), 4-hydroxytamoxifen (4-OHTAM), and tamoxifen-N-oxide were weak inhibitors of hSULT1E1 with Ki values ranging from 10 μM to 38 μM (i.e., over 1000 times higher than the 8.1 nM Km value for estradiol as substrate for the enzyme). In contrast to the results with hSULT1E1, endoxifen and 4-OHTAM were significant inhibitors of the sulfation of 2.0 µM estradiol catalyzed by hSULT1A1*1, with IC50 values (9.9 μM and 1.6 μM, respectively) that were similar to the Km value (1.5 μM) for estradiol as substrate for this enzyme. Additional investigation of the interaction of these metabolites with the two sulfotransferases revealed that endoxifen, 4-OHTAM, and N-desTAM were substrates for hSULT1E1 and hSULT1A1*1, although the relative catalytic efficiencies varied with both the substrate and the enzyme. These results may assist in future elucidation of cell- and tissue-specific effects of tamoxifen and its metabolites.
Collapse
Affiliation(s)
- Edwin J Squirewell
- Department of Pharmaceutical Sciences and Experimental Therapeutics, Division of Medicinal and Natural Products Chemistry, College of Pharmacy, University of Iowa, Iowa City, Iowa
| | - Michael W Duffel
- Department of Pharmaceutical Sciences and Experimental Therapeutics, Division of Medicinal and Natural Products Chemistry, College of Pharmacy, University of Iowa, Iowa City, Iowa
| |
Collapse
|
38
|
Yao-Borengasser A, Rogers LJ, Edavana VK, Penney RB, Yu X, Dhakal IB, Williams S, Kadlubar SA. Sulfotransferase 1A1 (SULT1A1) gene expression is regulated by members of the NFI transcription factors in human breast cancer cells. BMC Clin Pathol 2014; 14:1. [PMID: 24393253 PMCID: PMC3913331 DOI: 10.1186/1472-6890-14-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 12/18/2013] [Indexed: 12/18/2022] Open
Abstract
Background Sulfotransferase 1A1 (SULT1A1) gene expression is tissue specific, with little to no expression in normal breast epithelia. Expression in breast tumors has been documented, but the transcriptional regulation of SULT1A1 in human breast tissue is poorly understood. We identified Nuclear Factor I (NFI) as a transcription factor family involved in the regulation of SULT1A1 expression. Methods Transcription Factor Activation Profiling Plate Array assay was used to identify the possible transcription factors that regulate the gene expression of SULT1A1in normal breast MCF-10A cells and breast cancer ZR-75-1 cells. Expression levels of NFI-C and SULT1A1 were determined by real-time RT-PCR using total RNA isolated from 84 human liver samples. Expression levels of SULT1A1, NFI-A, NFI-B, NFI-C, and NFI-X were also determined in different human breast cancer cell lines (MCF-7, T-47D, ZR-75-1, and MDA-MB-231), in the transformed human epithelial cell line MCF-10A, and in ZR-75-1 cells that were transfected with siRNAs directed against NFI-A, NFI-B, NFI-C, or NFI-X for 48 h. The copy numbers of SULT1A1 in cell lines ZR-75-1, MCF-7, T-47D, MDA-MB-231, and MCF-10A were determined using a pre-designed Custom Plus TaqMan® Copy Number kit from Life Technologies. Results In normal human liver samples, SULT1A1 mRNA level was positively associated with NFI-C. In different human breast cancer and normal epithelial cell lines, SULT1A1 expression was positively correlated with NFI-B and NFI-C. SULT1A1 expression was decreased 41% and 61% in ZR-75-1 cells treated with siRNAs against NFI-A and NFI-C respectively. SULT1A1 gene expression was higher in cells containing more than one SULT1A1 copy numbers. Conclusions Our data suggests that SULT1A1 expression is regulated by NFI, as well as SULT1A1 copy number variation in human breast cancer cell lines. These data provide a mechanistic basis for the differential expression of SULT1A1 in different tissues and different physiological states of disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Susan A Kadlubar
- Division of Medical Genetics, College of Medicine, University of Arkansas for Medical Sciences, 4301 West Markham St,, Little Rock, AR, 72205, USA.
| |
Collapse
|
39
|
Leukocyte Gene Expression in Patients with Medication Refractory Depression before and after Treatment with ECT or Isoflurane Anesthesia: A Pilot Study. DEPRESSION RESEARCH AND TREATMENT 2014; 2014:582380. [PMID: 24826212 PMCID: PMC4009159 DOI: 10.1155/2014/582380] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 03/22/2014] [Indexed: 12/16/2022]
Abstract
Objective. To evaluate leukocyte gene expression for 9 selected genes (mRNAs) as biological markers in patients with medication refractory depression before and after treatment with ECT or isoflurane anesthesia (ISO). Methods. In a substudy of a nonrandomized open-label trial comparing effects of ECT to ISO therapy, blood samples were obtained before and after treatment from 22 patients with refractory depression, and leukocyte mRNA was assessed by quantitative PCR. Patients' mRNAs were also compared to 17 healthy controls. Results. Relative to controls, patients before treatment showed significantly higher IL10 and DBI and lower ADRA2A and ASIC3 mRNA (P < 0.025). Both ECT and ISO induced significant decreases after treatment in 4 genes: IL10, NR3C1, DRD4, and Sult1A1. After treatment, patients' DBI, ASIC3, and ADRA2A mRNA remained dysregulated. Conclusion. Significant differences from controls and/or significant changes after ECT or ISO treatment were observed for 7 of the 9 mRNAs studied. Decreased expression of 4 genes after effective treatment with either ECT or ISO suggests possible overlap of underlying mechanisms. Three genes showing dysregulation before and after treatment may be trait-like biomarkers of medication refractory depression. Gene expression for these patients has the potential to facilitate diagnosis, clarify pathophysiology, and identify potential biomarkers for treatment effects.
Collapse
|
40
|
Rižner TL. Estrogen biosynthesis, phase I and phase II metabolism, and action in endometrial cancer. Mol Cell Endocrinol 2013; 381:124-39. [PMID: 23911898 DOI: 10.1016/j.mce.2013.07.026] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2013] [Revised: 05/30/2013] [Accepted: 07/22/2013] [Indexed: 12/21/2022]
Abstract
Endometrial cancer is the most common gynecological malignancy in the developed World. Based on their histopathology, clinical manifestation, and epidemiology, the majority of endometrial cancer cases can be divided into two groups: the more prevalent type 1 which is associated with unopposed estrogen exposure; and the less common type 2, which is usually not associated with hyper-estrogenic factors. This manuscript overviews the published data on the expression of genes encoding the estrogen biosynthetic enzymes, the phase I and phase II estrogen metabolic enzymes, and the estrogen receptors in endometrial cancer, at the mRNA, protein and enzyme activity levels. The potential role of altered expression of these enzymes and receptors in cancerous versus control endometrial tissue, and the implication of estrogens in tumor initiation and promotion, are discussed. Finally, based on the published data, a model of estrogen metabolism and actions is proposed for pre-cancerous and cancerous endometrial tissue, and the role of the estrogens in the progression of endometrial cancer from endometrial hyperplasia is suggested.
Collapse
Affiliation(s)
- Tea Lanišnik Rižner
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia.
| |
Collapse
|
41
|
McNamara KM, Nakamura Y, Miki Y, Sasano H. Phase two steroid metabolism and its roles in breast and prostate cancer patients. Front Endocrinol (Lausanne) 2013; 4:116. [PMID: 24027559 PMCID: PMC3761226 DOI: 10.3389/fendo.2013.00116] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 08/19/2013] [Indexed: 12/31/2022] Open
Abstract
Breast and prostate cancer are diseases in which steroids and steroid metabolism could markedly influence clinical outcomes for patients. In both malignancies the modification of ketone and hydroxyl groups attached to the steroid backbone (phase one metabolism) has been examined in detail but the conjugation reactions (phase two metabolism) have not been extensively studied. Therefore, in this review we aim to summarize phase two metabolism in breast and prostate cancers from a number of perspectives, including the impact of variation in serum levels of conjugated steroids, tissue, and pathology specific expression of phase two enzymes, and consequences of genetic variations of these conjugation enzymes. In addition to this biological perspective, we will also address current pharmacological efforts to manipulate phase two metabolism as a potential therapy for hormone dependent cancers, including clinical trials of STS inhibitors and preclinical STS inhibitor development. While this review is not intended to cover any one particular area in great technical depth, it is intended as an introduction to and/or update on the importance of variance in phase two metabolic pathways in breast and prostate cancers.
Collapse
Affiliation(s)
- Keely M. McNamara
- Department of Pathology, Tohoku University School of Medicine, Miyagi, Sendai, Japan
- *Correspondence: Keely M. McNamara, Department of Anatomical Pathology, Tohoku University School of Graduate Medicine, 2-1 Seiryo-Machi Aoba-Ku, Miyagi, Sendai 980-8575, Japan e-mail:
| | - Yasuhiro Nakamura
- Department of Pathology, Tohoku University School of Medicine, Miyagi, Sendai, Japan
| | - Yasuhiro Miki
- Department of Pathology, Tohoku University School of Medicine, Miyagi, Sendai, Japan
| | - Hironobu Sasano
- Department of Pathology, Tohoku University School of Medicine, Miyagi, Sendai, Japan
| |
Collapse
|
42
|
Lim DW, Kim JG, Kim YT. Effects of dietary isoflavones from Puerariae radix on lipid and bone metabolism in ovariectomized rats. Nutrients 2013; 5:2734-46. [PMID: 23867712 PMCID: PMC3738997 DOI: 10.3390/nu5072734] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 06/16/2013] [Accepted: 07/02/2013] [Indexed: 11/16/2022] Open
Abstract
Puerariae radix, the dried root of Pueraria lobata Ohwi, is one of the earliest and most important edible crude herbs used for various medical purposes in oriental medicine. This study evaluated the metabolic effects of total isoflavones from P. lobata (PTIF) in ovariectomized (OVX) rats. The OVX rats were divided into four groups treated with distilled water, 17β-estradiol (E2 10 μg/kg, once daily, i.p.) and PTIF (30 and 100 mg/kg, once daily, p.o.) for eight weeks. The treatments with high-dose PTIF significantly decreased the bone mineral density (BMD) loss in the femur and inhibited the increase in body weight and lipoprotein levels compared to the OVX-control group without elevating the serum levels of the liver enzymes, aspartate aminotransferase (AST) and alanine transaminase (ALT). Furthermore, PTIF exhibits a hepatoprotective effect in OVX-induced hepatic steatosis, indicated with reduced hepatic lipid contents. Taken together, our findings suggest that PTIF may be useful for controlling lipid and bone metabolism, at least in OVX rats. Further research is necessary to determine whether PTIF will have the same effects in humans.
Collapse
Affiliation(s)
- Dong Wook Lim
- Functionality Evaluation Research Group, Korea Food Research Institute, Seongnam 463-746, Korea.
| | | | | |
Collapse
|
43
|
Expression of estrogen receptors (α, β), cyclooxygenase-2 and aromatase in normal endometrium and endometrioid cancer of uterus. Adv Med Sci 2013; 58:96-103. [PMID: 23625281 DOI: 10.2478/v10039-012-0055-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
PURPOSE Endometrial cancer (EC) is one of the most common malignancies of the female genital tract, but the etiology, especially its metabolism is still investigated. The aim of this study was to evaluate the presence and relative expression of Estrogen Receptors (α, β), Cyclooxygenase-2 and Aromatase in both endometrial cancer and normal mucosa. MATERIAL/METHODS Two groups of women were selected for the study: 1) patients with endometrioid endometrial cancer (FIGO I; G1 - G3) (n=35) and 2) subjects with normal endometrial tissue (control group, n=29). The expression of Estrogen Receptors (ERα, β), Cyclooxygenase-2 (COX-2), Aromatase were estimated by Western blot analysis. Furthermore, the associations between FIGO classification (stage: Ia, Ib), tumor grade (G) and expression of ERα, β, COX-2, aromatase proteins were evaluated. Overall and disease-free survival curves were generated according to the Kaplan-Meier method. Median follow-up time of the patients examined in this study was 39 months. RESULTS The relative expression of each examined protein was markedly higher in the endometrial cancer tissue as compared to the healthy endometrium. The trends towards greater expression along with a tumor progression was noticed (FIGO stage: Ia vs. Ib). Analysis of endometrial cancer risk factors and their influence on survival curves showed only an inverse significant correlations between obesity (BMI: 36.2; n=21) and disease-free survival in EC group (p=0.00872), but there was no significant association between obesity and overall survival (p=0.358). CONCLUSIONS Endometrioid endometrial cancer shows relatively higher expression of either ER, COX-2 and aromatase comparing to healthy mucosa, suggesting their involvement in tumor development and progression.
Collapse
|
44
|
Isshiki M, Ohta H, Tamura H. Coffee Reduces SULT1E1 Expression in Human Colon Carcinoma Caco-2 Cells. Biol Pharm Bull 2013; 36:299-304. [DOI: 10.1248/bpb.b12-00902] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Marina Isshiki
- Graduate School of Pharmaceutical Sciences, Keio University
| | - Haruka Ohta
- Graduate School of Pharmaceutical Sciences, Keio University
| | - Hiroomi Tamura
- Graduate School of Pharmaceutical Sciences, Keio University
| |
Collapse
|
45
|
Suzuki-Anekoji M, Suzuki A, Wu SW, Angata K, Murai KK, Sugihara K, Akama TO, Khoo KH, Nakayama J, Fukuda MN, Fukuda M. In vivo regulation of steroid hormones by the Chst10 sulfotransferase in mouse. J Biol Chem 2012; 288:5007-16. [PMID: 23269668 PMCID: PMC3576103 DOI: 10.1074/jbc.m112.433474] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Chst10 adds sulfate to glucuronic acid to form a carbohydrate antigen, HNK-1, in glycoproteins and glycolipids. To determine the role of Chst10 in vivo, we generated systemic Chst10-deficient mutant mice. Although Chst10−/− mice were born and grew to adulthood with no gross defects, they were subfertile. Uteri from Chst10−/− females at the pro-estrus stage were larger than those from wild-type females and exhibited a thick uterine endometrium. Serum estrogen levels in Chst10−/− females were higher than those from wild-type females, suggesting impaired down-regulation of estrogen. Because steroid hormones are often conjugated to glucuronic acid, we hypothesized that Chst10 sulfates glucuronidated steroid hormone to regulate steroid hormone in vivo. Enzymatic activity assays and structural analysis of Chst10 products by HPLC and mass spectrometry revealed that Chst10 indeed sulfates glucuronidated estrogen, testosterone, and other steroid hormones. We also identified an HPLC peak corresponding to sulfated and glucuronidated estradiol in serum from wild-type but not from Chst10 null female mice. Estrogen-response element reporter assays revealed that Chst10-modified estrogen likely did not bind to its receptor. These results suggest that subfertility exhibited by female mice following Chst10 loss results from dysregulation of estrogen. Given that Chst10 transfers sulfates to several steroid hormones, Chst10 likely functions in widespread regulation of steroid hormones in vivo.
Collapse
Affiliation(s)
- Misa Suzuki-Anekoji
- Glycobiology Unit, Tumor Microenvironment Program, Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California 92037, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Huhtinen K, Ståhle M, Perheentupa A, Poutanen M. Estrogen biosynthesis and signaling in endometriosis. Mol Cell Endocrinol 2012; 358:146-54. [PMID: 21875644 DOI: 10.1016/j.mce.2011.08.022] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Revised: 08/03/2011] [Accepted: 08/15/2011] [Indexed: 12/21/2022]
Abstract
Endometriosis is an estrogen-dependent gynecological disease where endometrium-like tissue grows outside uterine cavity. Endometriotic cell proliferation is stimulated by estrogens acting predominantly via their nuclear receptors. Estrogen receptors (ESR1, ESR2) are ligand activated transcription factors whose activation is dependent on the cell-specific dynamic expression of the receptors, on the interacting proteins and on the ligand availability. The different types of endometriotic lesions, peritoneal, deep, and ovarian endometriosis, may respond to estrogens differentially due to differences in the expression of the receptors and interacting proteins, and due to potential differences in the ligand availability regulated by the local estrogen synthesis. This review summarizes the current knowledge of estrogen synthesizing enzymes and estrogen receptors in different types of endometriosis lesions. Further studies are still needed to define the possible differences in steroid metabolism in different types of endometriotic lesions.
Collapse
Affiliation(s)
- Kaisa Huhtinen
- Department of Physiology, Institute of Biomedicine, University of Turku, 20014 Turku, Finland.
| | | | | | | |
Collapse
|
47
|
Fischer L, Deppert W, Pfeifer D, Stanzel S, Weimer M, Hanjalic-Beck A, Stein A, Straßer M, Zahradnik H, Schaefer W. Potential hazards to embryo implantation: A human endometrial in vitro model to identify unwanted antigestagenic actions of chemicals. Toxicol Appl Pharmacol 2012; 260:232-40. [DOI: 10.1016/j.taap.2012.02.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Revised: 02/22/2012] [Accepted: 02/26/2012] [Indexed: 10/28/2022]
|
48
|
Cho HJ, Jun HJ, Lee JH, Jia Y, Hoang MH, Shim JH, Park KH, Lee SJ. Acute Effect of High-dose Isoflavones from Pueraria lobata
(Willd.) Ohwi on Lipid and Bone Metabolism in Ovariectomized Mice. Phytother Res 2012; 26:1864-71. [DOI: 10.1002/ptr.4669] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Revised: 02/13/2012] [Accepted: 02/15/2012] [Indexed: 11/09/2022]
Affiliation(s)
- Hee Joon Cho
- Division of Food Bioscience and Technology, College of Life Sciences and Biotechnology; Korea University; Seoul 136-713 Korea
| | - Hee-jin Jun
- Division of Food Bioscience and Technology, College of Life Sciences and Biotechnology; Korea University; Seoul 136-713 Korea
| | - Ji Hae Lee
- Division of Food Bioscience and Technology, College of Life Sciences and Biotechnology; Korea University; Seoul 136-713 Korea
| | - Yaoyao Jia
- Division of Food Bioscience and Technology, College of Life Sciences and Biotechnology; Korea University; Seoul 136-713 Korea
| | - Minh Hien Hoang
- Division of Food Bioscience and Technology, College of Life Sciences and Biotechnology; Korea University; Seoul 136-713 Korea
| | - Jae-Hoon Shim
- Department of Food and Nutrition; Hallym University, College of Natural Sciences; Chuncheon Gangwon 200-702 Korea
| | - Kwan-Hwa Park
- Department of Foodservice Management and Nutrition; Sangmyuung University; Seoul 110-743 Republic of Korea
| | - Sung-Joon Lee
- Division of Food Bioscience and Technology, College of Life Sciences and Biotechnology; Korea University; Seoul 136-713 Korea
| |
Collapse
|
49
|
Amar D, Berger I, Amara N, Tafa G, Meijler MM, Aharoni A. The transition of human estrogen sulfotransferase from generalist to specialist using directed enzyme evolution. J Mol Biol 2011; 416:21-32. [PMID: 22197379 DOI: 10.1016/j.jmb.2011.12.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2011] [Revised: 12/04/2011] [Accepted: 12/06/2011] [Indexed: 11/29/2022]
Abstract
Broad specificity is believed to be a property of primordial enzymes that diverged during natural protein evolution to produce highly specific and efficient enzymes. Human estrogen sulfotransferase (SULT1E1) is a broad-specificity enzyme that detoxifies a variety of chemicals, including estrogens, by the transfer of sulfate. To study the molecular basis for the broad specificity of this enzyme and to investigate the process of SULT1E1 specialization, we have adopted a directed enzyme evolution approach. Using two iterative rounds of evolution, we generated SULT1E1 mutants with increased thermostability and narrower specificity from the broadly specific wild-type enzyme. To identify mutants with enhanced specificity, we developed an unbiased screening assay to assess sulfate transfer to three different acceptors in parallel. Such an assay enabled the isolation of SULT1E1 mutants with enhanced or wild-type activity toward an estrogen acceptor and significantly reduced activity for phenol or coumarin type of acceptors, leading to up to 3 orders of magnitude increase in specificity. We found that mutations conferring novel specificity are located in the vicinity of the active site and thus may play a direct role in reshaping the acceptor-binding site. Finally, such mutations resulted in reduced SULT1E1 thermostability, revealing a trade-off between SULT1E1 thermostability and acquisition of novel function.
Collapse
Affiliation(s)
- Dotan Amar
- Department of Life Sciences, Ben-Gurion University of the Negev, Be'er Sheva 84105, Israel
| | | | | | | | | | | |
Collapse
|
50
|
Robidoux A, Rich E, Bureau N, Mader S, Laperrière D, Bail M, Tremblay N, Patenaude M, Turgeon J. A prospective pilot study investigating the musculoskeletal pain in postmenopausal breast cancer patients receiving aromatase inhibitor therapy. Curr Oncol 2011; 18:285-94. [PMID: 22184490 PMCID: PMC3224030 DOI: 10.3747/co.v18i6.909] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Although arthralgia is a known adverse effect of aromatase inhibitor (ai) treatment in postmenopausal breast cancer patients, few studies have carried out a comprehensive evaluation of the nature, onset, and incidence of musculoskeletal (msk) pain in these patients. We therefore used a pilot study to identify conditions or markers predictive of pain. METHODS For 24 weeks, we monitored 30 eligible postmenopausal women starting ai therapy. Pre-existing and incident msk conditions and pain were assessed clinically and with ultrasonography of the hands and wrists. In addition, patient questionnaires were used to assess pain before and during ai therapy. Biochemical markers were measured at baseline and at regular intervals after anastrozole therapy began. Gene profiling studies were carried out before and 48 hours after the initial ai administration. RESULTS Over the 24-week study period, 20 participants (67%) showed no pain symptoms; 5 (17%) experienced low or moderate pain at baseline, which did not increase with ai treatment; and during therapy, 5 (17%) showed exacerbation of pain attributable to osteoarthritis of the hand and to finger flexor tenosynovitis. Although all 30 participants had some degree of msk conditions before anastrozole therapy started, the pre-existing conditions did not necessarily predispose the women to increased pain during anastrozole treatment. Higher levels of urinary N-telopeptides of type i collagen were associated with the groups presenting pain, suggesting a higher extent of pre-existing bone resorption, without significant evolution over the 24-week treatment period. Slightly higher levels of 1,25(OH)(2) vitamin D(3) were observed at baseline in patients with pain increase, but did not significantly change during treatment; however, average levels of 25(OH) vitamin D(3) increased, likely because of supplementation. Although biochemical markers did not discriminate efficiently between pain groups, a signature of 166 genes in peripheral blood mononuclear cells was identified that could stratify patients into the various groups observed in this pilot study. The gene signature was enriched in components of inflammatory signalling and chemokine expression, of antitumoural immunity pathways, and of metabolic response to hormones and xenobiotics, although no clinically significant association could be made in the present study, considering the small number of patients. Nevertheless, the observed trend suggests the feasibility of developing surrogate predictive markers of msk pain. Patient compliance was high in this study and was not affected by pain exacerbation. CONCLUSIONS Baseline msk assessment showed pre-existing causes for pain in most of the study patients before initiation of the ai. Exacerbation of existing osteoarthritis pain and tenosynovial symptoms was the primary cause of pain increase. Musculoskeletal pain assessment at baseline and prompt treatment of pain symptoms may help to optimize adherence to ai therapy. The value of routinely assessing inflammatory markers such as C-reactive protein and erythrocyte sedimentation rate was not supported by our pilot study. Gene expression profiles in peripheral blood mononuclear cells may be further explored in larger-scale studies as stratification markers to identify patients at risk of developing arthralgia.
Collapse
Affiliation(s)
- A. Robidoux
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Montreal, QC
| | - E. Rich
- Department of Medicine, Centre hospitalier de l’Université de Montréal, Montreal, QC
| | - N.J. Bureau
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Montreal, QC
- Radiology Department, Université de Montréal, Montreal, QC
| | - S. Mader
- Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, QC
- Biochemistry Department, Université de Montréal, Montreal, QC
| | - D. Laperrière
- Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, QC
| | - M. Bail
- Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, QC
| | - N. Tremblay
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Montreal, QC
| | - M. Patenaude
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Montreal, QC
| | - J. Turgeon
- Faculty of Pharmacy, Université de Montréal, Montreal, QC
| |
Collapse
|