1
|
Zhang Q, Hao X, Sun X, Jia YC, Zhu YY, Yang YX, Zhu BT. 4-Hydroxyestrogen metabolites strongly prevent chemically-induced ferroptotic hepatocyte injury in vitro and in vivo. Eur J Pharmacol 2025; 993:177313. [PMID: 39921062 DOI: 10.1016/j.ejphar.2025.177313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/10/2025]
Abstract
Ferroptosis is a regulated cell death characterized by excessive accumulation of toxic lipid reactive oxygen species (ROS). Ferroptosis is an underlying cause in some human diseases, including the drug-induced liver injury. The present study aims to determine whether 4-hydroxyestrone (4-OH-E1) and 4-hydroxyestradiol (4-OH-E2), two endogenous catechol estrogens, can prevent chemically-induced ferroptotic hepatocyte injury in vitro and in vivo. The induction of ferroptotic cell death by erastin and RSL3 in rat H-4-II-E and human HuH-7 hepatoma cells is used as in vitro models. 4-OH-E1 and 4-OH-E2 each exhibit a strong protection against erastin/RSL3-induced ferroptosis in H-4-II-E hepatoma cells, and they also strongly abrogate erastin/RSL3-induced accumulation of cellular NO, ROS and lipid-ROS. A similar protective effect is observed with 4-OH-E1 and 4-OH-E2 in RSL3-induced ferroptosis in HuH-7 cells. Mechanistically, these two catechol estrogens protect hepatoma cells against chemically-induced ferroptosis mainly through binding to cellular PDI protein with a high affinity, which leads to inhibition of PDI-catalyzed NOS dimerization (activation), thereby preventing the accumulation of cellular NO, ROS and lipid-ROS. In addition, the direct antioxidant activity of these two estrogens may also partially contribute to their cytoprotective effect. In vivo animal studies show that 4-OH-E1 and 4-OH-E2 also have a strong protective effect against acetaminophen-induced liver injury in a mouse model. Together, the results of this study demonstrate that 4-OH-E1 and 4-OH-E2 are endogenous factors with a strong protective activity against chemically-induced hepatocyte injury both in vitro and in vivo.
Collapse
Affiliation(s)
- Qi Zhang
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Xiangyu Hao
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Xi Sun
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Yi-Chen Jia
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Yan-Yin Zhu
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Yong Xiao Yang
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Bao Ting Zhu
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, China; Shenzhen Bay Laboratory, Shenzhen, 518172, China.
| |
Collapse
|
2
|
Sun X, Hao X, Jia YC, Zhang Q, Zhu YY, Yang YX, Zhu BT. Protective effect of 2-hydroxyestrone and 2-hydroxyestradiol against chemically induced hepatotoxicity in vitro and in vivo. J Pharmacol Exp Ther 2025; 392:100050. [PMID: 40023585 DOI: 10.1016/j.jpet.2024.100050] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 10/31/2024] [Indexed: 03/04/2025] Open
Abstract
Ferroptosis is a form of regulated cell death closely associated with glutathione depletion and accumulation of reactive lipid peroxides. In this study, we seek to determine whether 2-hydroxyestrone (2-OH-E1) and 2-hydroxyestradiol (2-OH-E2), 2 major metabolites of endogenous estrone (E1) and 17β-estradiol (E2) formed by cytochrome P450 in the liver, can protect against erastin- and RSL3-induced ferroptosis in hepatoma cells (H-4-II-E and HuH-7) in vitro and acetaminophen-induced mouse liver injury in vivo. We find that 2-OH-E1 and 2-OH-E2 can protect, in a dose-dependent manner, H-4-II-E hepatoma cells against erastin/RSL3-induced ferroptosis. A similar protective effect of 2-OH-E1 and 2-OH-E2 against erastin- and RSL3-induced ferroptosis is also observed in HuH-7 hepatoma cells. These 2 estrogen metabolites can strongly abrogate erastin- and RSL3-induced accumulation of cellular NO, reactive oxygen species (ROS), and lipid-ROS. Mechanistically, 2-OH-E1 and 2-OH-E2 protect cells against chemically induced ferroptosis by binding to cellular protein disulfide isomerase and then inhibiting its catalytic activity and reducing protein disulfide isomerase-mediated activation (dimerization) of inducible nitric oxide synthase, abrogating cellular NO, ROS, and lipid-ROS accumulation. Animal studies show that 2-OH-E1 and 2-OH-E2 also exhibit strong protection against acetaminophen-induced liver injury in mice. Interestingly, although E1 and E2 have a very weak protective effect in cultured hepatoma cells, they exhibit a similarly strong protective effect as 2-OH-E1 and 2-OH-E2 in vivo, suggesting that the metabolic conversion of E1 and E2 to 2-OH-E1 and 2-OH-E2 contributes importantly to their hepatoprotective effect. This study reveals that 2-OH-E1 and 2-OH-E2 are important endogenous factors for protection against chemically induced liver injury in vivo. SIGNIFICANCE STATEMENT: Ferroptosis is an iron-dependent and lipid reactive oxygen species-dependent form of regulated cell death. Recent evidence has shown that protein disulfide isomerase (PDI) is an important mediator of chemically induced ferroptosis and also a new target for ferroptosis protection. This study shows that 2-hydroxyestrone and 2-hydroxyestradiol are 2 inhibitors of PDI that can strongly protect against chemically induced ferroptotic hepatocyte death in vitro and in vivo. This work supports a PDI-mediated, estrogen receptor-independent mechanism of hepatocyte protection by 2-hydroxyestrone and 2-hydroxyestradiol.
Collapse
Affiliation(s)
- Xi Sun
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, China
| | - Xiangyu Hao
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, China
| | - Yi-Chen Jia
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, China
| | - Qi Zhang
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, China
| | - Yan-Yin Zhu
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, China
| | - Yong Xiao Yang
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, China
| | - Bao Ting Zhu
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, China; Shenzhen Bay Laboratory, Shenzhen, China.
| |
Collapse
|
3
|
You Q, Song H, Zhu Z, Wang J, Wang R, Du M, Fu Y, Yuan J, Tan R. Decoding the enigmatic estrogen paradox in pulmonary hypertension: delving into estrogen metabolites and metabolic enzymes. Cell Mol Biol Lett 2024; 29:155. [PMID: 39695964 DOI: 10.1186/s11658-024-00671-w] [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: 06/07/2024] [Accepted: 11/21/2024] [Indexed: 12/20/2024] Open
Abstract
Pulmonary hypertension (PH) presents a puzzling sex bias, being more prevalent in women yet often less severe than in men, and the underlying reasons remain unclear. Studies using animal models, and limited clinical data have revealed a protective influence of exogenous estrogens, known as the estrogen paradox. Research suggests that beyond its receptor-mediated effects, estrogen acts through metabolites such as 2-ME2, 4-OHE2, and 16-OHE2, which are capable of exhibiting protective or detrimental effects in PH, prompting the need to explore their roles in PH to untangle sex differences and the estrogen paradox. Hypoxia disrupts the balance of estrogen metabolites by affecting the enzymes responsible for estrogen metabolism. Delving into the role of these metabolic enzymes not only illuminates the sex difference in PH but also provides a potential rationale for the estrogen paradox. This review delves into the intricate interplay between estrogen metabolites, metabolic enzymes, and PH, offering a deeper understanding of sex-specific differences and the perplexing estrogen paradox in the context of this condition.
Collapse
Affiliation(s)
- Qiang You
- Department of Physiology, Basic Medical School, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - Hequn Song
- First Clinical Medical School, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Ziming Zhu
- College of Second Clinical Medical, Jining Medical University, Jining, 272067, Shandong, China
| | - Jinzheng Wang
- College of Second Clinical Medical, Jining Medical University, Jining, 272067, Shandong, China
| | - Ruixin Wang
- School of Nursing, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Mingjia Du
- School of Nursing, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Yingjie Fu
- School of Pharmacy, Jining Medical University, Rizhao, 276826, Shandong, China.
| | - Jinxiang Yuan
- Lin He's Academician Workstation of New Medicine and Clinical Translation, Jining Medical University, Jining, 272067, Shandong, China.
| | - Rubin Tan
- Department of Physiology, Basic Medical School, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
4
|
Sherwani S, Khan MWA, Khan WA, Rajendrasozhan S, Al-Motair K, Khan H, Ahmad S. Estrogenized HSA induced high-affinity autoantibodies in breast cancer - Novel biomarker for early detection. Front Oncol 2024; 14:1493320. [PMID: 39664179 PMCID: PMC11631743 DOI: 10.3389/fonc.2024.1493320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 10/29/2024] [Indexed: 12/13/2024] Open
Abstract
Objective Breast cancer (BC) is the second most prevalent cancer worldwide. Estrogen has been increasingly recognized as a major contributor to the development of BC, playing a more critical role than previously understood. Estrogen derived nucleic acid and protein adducts have been shown to play significant roles in BC development and progression. However, the alterations in molecular mechanism(s) and immune pathways arising as a result of estrogenization still remain elusive. Patients and methods 4-hydroxyestradiol (4-OHE2) was used for adduct formation with protein human serum albumin (HSA) (4-OHE2-HSA). The affinity of antibodies for 4-OHE2-HSA was evaluated in breast cancer patients. Immunoassays (direct binding ELISA, inhibition ELISA, and quantitative precipitin titration assay) were used to assess autoantibodies against estrogenized HSA in BC patients (n = 85) and healthy controls (n = 45). Results Estrogenization of HSA altered both its structure and function and compromised its interactions with various HSA-binding proteins. BC patients demonstrated high-affinity antibodies against 4-OHE2-HSA as compared to HSA (p < 0.05). Additionally, cytokines Interleukin (IL)-1, IL-6 and tumor necrosis factor-alpha (TNF-α) were significantly elevated in BC patients as compared to the control group. Several factors, such as chemotherapy, estrogen receptors (ERs), and combination of surgery and chemotherapy, influenced the production of antibodies in cancer patients. The affinity constant for estrogenized HSA was 1.31 × 10-7 M, while for HSA and 4-OHE2, it was 1.68 × 10-6 M and 1.36 × 10-6 M, respectively. Conclusions Estrogenized HSA is highly immunogenic, resulting in functional alterations. High affinity antibodies were detected in BC patients against 4-OHE2-HSA. Consequently, 4-OHE2-HSA may serve as a novel molecular target for potential cancer therapeutics. Furthermore, autoantibodies against 4-OHE2-HSA could serve as a potential biomarker for early detection of BC.
Collapse
Affiliation(s)
- Subuhi Sherwani
- Department of Biology, College of Science, University of Hail, Hail, Saudi Arabia
- Medical and Diagnostic Research Center, University of Hail, Hail, Saudi Arabia
| | - Mohd Wajid Ali Khan
- Medical and Diagnostic Research Center, University of Hail, Hail, Saudi Arabia
- Department of Chemistry, College of Science, University of Hail, Hail, Saudi Arabia
| | - Wahid Ali Khan
- Department of Clinical Biochemistry, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Saravanan Rajendrasozhan
- Medical and Diagnostic Research Center, University of Hail, Hail, Saudi Arabia
- Department of Chemistry, College of Science, University of Hail, Hail, Saudi Arabia
| | - Khalid Al-Motair
- Medical and Diagnostic Research Center, University of Hail, Hail, Saudi Arabia
| | - Hamda Khan
- Department of Biochemistry, J.N. Medical College, Aligarh Muslim University, Aligarh, India
- IIRC-1, Department of Biosciences, Integral University, Lucknow, India
| | - Saheem Ahmad
- Medical and Diagnostic Research Center, University of Hail, Hail, Saudi Arabia
- Department of Medical Laboratories, College of Applied Medical Sciences, University of Hail, Hail, Saudi Arabia
| |
Collapse
|
5
|
Huang X, Hou MJ, Zhu BT. Protection of HT22 neuronal cells against chemically-induced ferroptosis by catechol estrogens: protein disulfide isomerase as a mechanistic target. Sci Rep 2024; 14:23988. [PMID: 39402104 PMCID: PMC11473836 DOI: 10.1038/s41598-024-74742-5] [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: 05/15/2024] [Accepted: 09/30/2024] [Indexed: 10/17/2024] Open
Abstract
Ferroptosis is a form of regulated cell death, characterized by excessive iron-dependent lipid peroxidation. Biochemically, ferroptosis can be selectively induced by erastin through glutathione depletion or through inhibition of glutathione peroxidase 4 by RSL3, which leads to accumulation of cytotoxic lipid reactive oxygen species (ROS). Protein disulfide isomerase (PDI) was recently shown to mediate erastin/RSL3-induced ferroptosis and thus also become a new target for protection against chemically-induced ferroptosis. The present study aims to identify endogenous compounds that can protect against erastin/RSL3-induced ferroptotic cell death. We find that 2-hydroxyestrone, 2-hydroxyestradiol, 4-hydroxyestrone and 4-hydroxyestradiol, four major endogenous catechol estrogens, are effective inhibitors of PDI, and can strongly protect against chemically-induced ferroptotic cell death in cultured HT22 mouse hippocampal neuronal cells. The CETSA assay showed that these catechol estrogens can bind to PDI in live cells. PDI knockdown attenuates the protective effect of these catechol estrogens against chemically-induced ferroptosis. Mechanistically, inhibition of PDI's catalytic activity by catechol estrogens abrogates erastin/RSL3-induced dimerization of nitric oxide synthase, thereby preventing the subsequent accumulation of cellular nitric oxide, ROS and lipid-ROS, and ultimately ferroptotic cell death. In addition, joint treatment of cells with catechol estrogens also abrogates erastin/RSL3-induced upregulation of nitric oxide synthase protein levels, which also contributes to the cytoprotective effect of the catechol estrogens. In conclusion, the present study demonstrates that the catechol estrogens are protectors of HT22 neuronal cells against chemically-induced ferroptosis, and inhibition of PDI's catalytic activity by these estrogens contributes to a novel, estrogen receptor-independent mechanism of cytoprotection.
Collapse
Affiliation(s)
- Xuanqi Huang
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Road, Longgang District, Shenzhen, 518172, China
| | - Ming-Jie Hou
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Road, Longgang District, Shenzhen, 518172, China
| | - Bao Ting Zhu
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Road, Longgang District, Shenzhen, 518172, China.
- Shenzhen Bay Laboratory, Shenzhen, 518055, China.
| |
Collapse
|
6
|
Timofeeva АА, Minina VI, Torgunakova AV, Soboleva ОА, Тitov RА, Zakharova YА, Bakanova ML, Glushkov АN. Polymorphic variants of the hOGG1, APEX1, XPD, SOD2, and CAT genes involved in DNA repair processes and antioxidant defense and their association with breast cancer risk. Vavilovskii Zhurnal Genet Selektsii 2024; 28:424-432. [PMID: 39027127 PMCID: PMC11253018 DOI: 10.18699/vjgb-24-48] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 01/12/2024] [Accepted: 02/26/2024] [Indexed: 07/20/2024] Open
Abstract
Breast cancer is one of the leading causes of mortality among women. The most frequently encountered tumors are luminal tumors. Associations of polymorphisms in the hOGG1 (rs1052133), APEX1 (rs1130409), XPD (rs13181), SOD2 (rs4880), and CAT (rs1001179) genes were studied in 313 nonsmoking postmenopausal patients with luminal B subtype breast cancer. The control group consisted of 233 healthy nonsmoking postmenopausal women. Statistically significant associations of the XPD and APEX1 gene polymorphisms with the risk of developing luminal B Her2-negative subtype of breast cancer were observed in a log-additive inheritance model, while the CAT gene polymorphism showed an association in a dominant inheritance model (OR = 1.41; CI 95 %: 1.08-1.85; Padj.= 0.011; OR = 1.39; CI 95 %: 1.07-1.81; Padj = 0.013 и OR = 1.70; CI 95 %: 1.19-2.43; Padj = 0.004, respectively). In the group of elderly women (aged 60-74 years), an association of the CAT gene polymorphism with the risk of developing luminal B subtype of breast cancer was found in a log-additive inheritance model (OR = 1.87; 95 % CI: 1.22-2.85; Padj = 0.0024). Using MDR analysis, the most optimal statistically significant 3-locus model of gene-gene interactions in the development of luminal B Her2-negative subtype breast cancer was found. MDR analysis also showed a close interaction and mutual enhancement of effects between the APEX1 and SOD2 loci and the independence of the effects of these loci from the CAT locus in the formation of luminal B subtype breast cancer.
Collapse
Affiliation(s)
- А А Timofeeva
- Federal Research Center of Coal and Coal Chemistry of the Siberian Branch of the Russian Academy of Sciences, Kemerovo, Russia
| | - V I Minina
- Federal Research Center of Coal and Coal Chemistry of the Siberian Branch of the Russian Academy of Sciences, Kemerovo, Russia Kemerovo State University, Kemerovo, Russia
| | - A V Torgunakova
- Federal Research Center of Coal and Coal Chemistry of the Siberian Branch of the Russian Academy of Sciences, Kemerovo, Russia Kemerovo State University, Kemerovo, Russia
| | - О А Soboleva
- Federal Research Center of Coal and Coal Chemistry of the Siberian Branch of the Russian Academy of Sciences, Kemerovo, Russia
| | - R А Тitov
- Federal Research Center of Coal and Coal Chemistry of the Siberian Branch of the Russian Academy of Sciences, Kemerovo, Russia Kemerovo State University, Kemerovo, Russia
| | - Ya А Zakharova
- Federal Research Center of Coal and Coal Chemistry of the Siberian Branch of the Russian Academy of Sciences, Kemerovo, Russia Kemerovo State University, Kemerovo, Russia
| | - M L Bakanova
- Federal Research Center of Coal and Coal Chemistry of the Siberian Branch of the Russian Academy of Sciences, Kemerovo, Russia
| | - А N Glushkov
- Federal Research Center of Coal and Coal Chemistry of the Siberian Branch of the Russian Academy of Sciences, Kemerovo, Russia
| |
Collapse
|
7
|
Chen L, Xu T, Lou J, Zhang T, Wu S, Xie R, Xu J. The beneficial roles and mechanisms of estrogens in immune health and infection disease. Steroids 2024; 207:109426. [PMID: 38685461 DOI: 10.1016/j.steroids.2024.109426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 02/28/2024] [Accepted: 04/21/2024] [Indexed: 05/02/2024]
Abstract
Multiple epidemiologic studies have revealed that gender is considered one of the important factors in the frequency and severity of certain infectious diseases, in which estrogens may play a vital role. There is growing evidence that estrogens as female sex hormone can modulate multiple biological functions outside of the reproductive system, such as in brain and cardiovascular system. However, it is largely unknown about the roles and mechanisms of estrogens/estrogen receptors in immune health and infection disease. Thence, by reading a lot of literature, we summarized the regulatory mechanisms of estrogens/estrogen receptors in immune cells and their roles in certain infectious diseases with gender differences. Therefore, estrogens may have therapeutic potentials to prevent and treat these infectious diseases, which needs further clinical investigation.
Collapse
Affiliation(s)
- Lan Chen
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Ting Xu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jun Lou
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Ting Zhang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Sheng Wu
- Department of Gastroenterology, Liupanshui People's Hospital, Liupanshui City 553000, Guizhou Province, China
| | - Rui Xie
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| | - Jingyu Xu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| |
Collapse
|
8
|
Azmy Nabeh O, Amr A, Faoosa AM, Esmat E, Osama A, Khedr AS, Amin B, Saud AI, Elmorsy SA. Emerging Perspectives on the Impact of Diabetes Mellitus and Anti-Diabetic Drugs on Premenstrual Syndrome. A Narrative Review. Diabetes Ther 2024; 15:1279-1299. [PMID: 38668996 PMCID: PMC11096298 DOI: 10.1007/s13300-024-01585-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 04/08/2024] [Indexed: 05/16/2024] Open
Abstract
Diabetes mellitus (DM) and premenstrual syndrome (PMS) are global health challenges. Both disorders are often linked to a range of physical and psychological symptoms that significantly impact the quality of life of many women. Yet, the exact relation between DM and PMS is not clear, and the management of both conditions poses a considerable challenge. In this review, we aimed to investigate the interplay between DM, anti-diabetic drugs, and the different theories and symptoms of PMS. Female sex hormones are implicated in the pathophysiology of PMS and can also impair blood glucose control. In addition, patients with diabetes face a higher susceptibility to anxiety and depression disorders, with a significant number of patients experiencing symptoms such as fatigue and difficulty concentrating, which are reported in patients with PMS as well. Complications related to diabetic medications, such as hypoglycemia (with sulfonylurea) and fluid retention (with thiazolidinediones) may also mediate PMS-like symptoms. DM can, in addition, disturb the normal gut microbiota (GM), with a consequent loss of beneficial GM metabolites that guard against PMS, particularly the short-chain fatty acids and serotonin. Among the several available anti-diabetic drugs, those (1) with an anti-inflammatory potential, (2) that can preserve the beneficial GM, and (3) possessing a lower risk for hypoglycemia, might have a favorable outcome in PMS women. Yet, well-designed clinical trials are needed to investigate the anti-diabetic drug(s) of choice for patients with diabetes and PMS.
Collapse
Affiliation(s)
- Omnia Azmy Nabeh
- Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt.
| | - Alaa Amr
- Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | | | - Eshraka Esmat
- Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Alaa Osama
- Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | | | - Basma Amin
- Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Alaa I Saud
- Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | | |
Collapse
|
9
|
Xu J, Tan C. Interaction between CYP1A1 gene polymorphism and environment factors on risk of endometrial cancer. Environ Health Prev Med 2024; 29:54. [PMID: 39384367 PMCID: PMC11473388 DOI: 10.1265/ehpm.24-00007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 07/26/2024] [Indexed: 10/11/2024] Open
Abstract
BACKGROUND The purpose of this study was to investigate the impact of single nucleotide polymorphisms (SNPs) of the CYP1A1 gene and the gene-environment interaction on the susceptibility to endometrial cancer in Chinese women. METHOD Logistic regression was performed to investigate the association between the four SNPs of the CYP1A1 gene and the risk of endometrial cancer. Generalized multifactor dimensionality reduction (GMDR) was employed to analyze the gene-environmental interaction. RESULTS A total of 934 women with a mean age of 61.7 ± 10.5 years were selected, including 310 endometrial cancer patients and 624 normal controls. The frequency of rs4646421- T allele was higher in endometrial cancer patients than normal controls, the T allele of rs4646421 was 28.1% in endometrial cancer patients and 21.0% in normal controls (p < 0.001). Logistic regression analysis showed that the rs4646421 - T allele was associated with increased risk of endometrial cancer, OR (95% CI) were 1.52 (1.11-1.97) and 1.91 (1.35-2.52), respectively. GMDR analysis found a significant two-locus model (p = 0.0107) involving rs4646421 and abdominal obesity (defined by waist circumference), indicating a potential gene-environment interaction between rs4646421 and abdominal obesity. Abdominal obese subjects with rs4646421- CT or TT genotype have the highest risk of endometrial cancer, compared to non-abdominal obese subjects with the rs4646421- CC genotype, the OR (95%CI) was 2.23 (1.62-2.91). CONCLUSIONS Both the rs4646421- T allele and the interaction between rs4646421 and abdominal obesity were associated with increased risk of endometrial cancer.
Collapse
Affiliation(s)
- Jian Xu
- Department of Oncology, the second people's hospital of Nantong, Affiliated Nantong Rehabilitation Hospital of Nantong University, Nantong City, Jiangsu Province, China, 226002
| | - Cheng Tan
- Department of Radiotherapy, Nantong Tumor Hospital, Tumor Hospital Affiliated to Nantong University, Nantong City, Jiangsu Province, China, 226300
| |
Collapse
|
10
|
Kolanczyk RC, Denny JS, Sheedy BR, Olson VV, Serrano JA, Tapper MA. Increased Endocrine Activity of Xenobiotic Chemicals as Mediated by Metabolic Activation. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2023; 42:2747-2757. [PMID: 37712519 DOI: 10.1002/etc.5748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/15/2023] [Accepted: 09/13/2023] [Indexed: 09/16/2023]
Abstract
The US Environmental Protection Agency (USEPA) is faced with long lists of chemicals that require hazard assessment. The present study is part of a larger effort to develop in vitro assays and quantitative structure-activity relationships applicable to untested chemicals on USEPA inventories through study of estrogen receptor (ER) binding and estrogen-mediated gene expression in fish. The present effort investigates metabolic activation of chemicals resulting in increased estrogenicity. Phenolphthalin (PLIN) was shown not to bind rainbow trout (Oncorhynchus mykiss) ER (rtER) in a competitive binding assay, but vitellogenin (Vtg) expression was induced in trout liver slices exposed to 10-4 and 10-3.7 M PLIN. Phenolphthalein (PLEIN), a metabolite of PLIN, was subsequently determined to be formed when slices were exposed to PLIN. It binds rtER with a relative binding affinity to 17β-estradiol of 0.020%. Slices exposed to PLEIN expressed Vtg messenger RNA (mRNA) at 10-4.3 , 10-4 , and 10-3.7 M, with no detectable PLIN present. Thus, Vtg expression noted in PLIN slice exposures was explained by metabolism to PLEIN in trout liver slices. A second model chemical, 4,4'-methylenedianiline (MDA), was not shown to bind rtER but did induce Vtg mRNA production in tissue slices at 10-4.3 , 10-4 , and 10-3.7 M in amounts nearly equal to reference estradiol induction, thus indicating metabolic activation of MDA. A series of experiments were performed to identify a potential metabolite responsible for the observed increase in activity. Potential metabolites hydroxylamine-MDA, nitroso-MDA, azo-MDA, and azoxy-MDA were not observed. However, acetylated MDA was observed and tested in both ER-binding and tissue slice Vtg induction assays. Environ Toxicol Chem 2023;42:2747-2757. © 2023 SETAC. This article has been contributed to by U.S. Government employees and their work is in the public domain in the USA.
Collapse
Affiliation(s)
- Richard C Kolanczyk
- Great Lakes Toxicology and Ecology Division, Office of Research and Development, Center for Computational Toxicology and Exposure, US Environmental Protection Agency, Duluth, Minnesota
| | - Jeffrey S Denny
- Great Lakes Toxicology and Ecology Division, Office of Research and Development, Center for Computational Toxicology and Exposure, US Environmental Protection Agency, Duluth, Minnesota
| | - Barbara R Sheedy
- Great Lakes Toxicology and Ecology Division, Office of Research and Development, Center for Computational Toxicology and Exposure, US Environmental Protection Agency, Duluth, Minnesota
| | - Victoria V Olson
- Department of Biology, The College of Saint Scholastica, Duluth, Minnesota, USA
| | - Jose A Serrano
- Great Lakes Toxicology and Ecology Division, Office of Research and Development, Center for Computational Toxicology and Exposure, US Environmental Protection Agency, Duluth, Minnesota
| | - Mark A Tapper
- Great Lakes Toxicology and Ecology Division, Office of Research and Development, Center for Computational Toxicology and Exposure, US Environmental Protection Agency, Duluth, Minnesota
| |
Collapse
|
11
|
Lin YC, Cheung G, Zhang Z, Papadopoulos V. Mitochondrial cytochrome P450 1B1 is involved in pregnenolone synthesis in human brain cells. J Biol Chem 2023; 299:105035. [PMID: 37442234 PMCID: PMC10413356 DOI: 10.1016/j.jbc.2023.105035] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/02/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Neurosteroids, which are steroids synthesized by the nervous system, can exert neuromodulatory and neuroprotective effects via genomic and nongenomic pathways. The neurosteroid and major steroid precursor pregnenolone has therapeutical potential in various diseases, such as psychiatric and pain disorders, and may play important roles in myelination, neuroinflammation, neurotransmission, and neuroplasticity. Although pregnenolone is synthesized by CYP11A1 in peripheral steroidogenic organs, our recent study showed that pregnenolone must be synthesized by another mitochondrial cytochrome P450 (CYP450) enzyme other than CYP11A1 in human glial cells. Therefore, we sought to identify the CYP450 responsible for pregnenolone production in the human brain. Upon screening for CYP450s expressed in the human brain that have mitochondrial localization, we identified three enzyme candidates: CYP27A1, CYP1A1, and CYP1B1. We found that inhibition of CYP27A1 through inhibitors and siRNA knockdown did not negatively affect pregnenolone synthesis in human glial cells. Meanwhile, treatment of human glial cells with CYP1A1/CYP1B1 inhibitors significantly reduced pregnenolone production in the presence of 22(R)-hydroxycholesterol. We performed siRNA knockdown of CYP1A1 or CYP1B1 in human glial cells and found that only CYP1B1 knockdown significantly decreased pregnenolone production. Furthermore, overexpression of mitochondria-targeted CYP1B1 significantly increased pregnenolone production under basal conditions and in the presence of hydroxycholesterols and low-density lipoprotein. Inhibition of CYP1A1 and/or CYP1B1 via inhibitors or siRNA knockdown did not significantly reduce pregnenolone synthesis in human adrenal cortical cells, implying that CYP1B1 is not a major pregnenolone-producing enzyme in the periphery. These data suggest that mitochondrial CYP1B1 is involved in pregnenolone synthesis in human glial cells.
Collapse
Affiliation(s)
- Yiqi Christina Lin
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California, USA
| | - Garett Cheung
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California, USA
| | - Zeyu Zhang
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California, USA
| | - Vassilios Papadopoulos
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California, USA.
| |
Collapse
|
12
|
Gigli GL, de Biase S, Pellitteri G, Pez S, Garbo R, Tereshko Y, Valente M. Restless legs syndrome in internal medicine. ENCYCLOPEDIA OF SLEEP AND CIRCADIAN RHYTHMS 2023:743-756. [DOI: 10.1016/b978-0-12-822963-7.00146-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
13
|
Steiner BM, Berry DC. The Regulation of Adipose Tissue Health by Estrogens. Front Endocrinol (Lausanne) 2022; 13:889923. [PMID: 35721736 PMCID: PMC9204494 DOI: 10.3389/fendo.2022.889923] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/25/2022] [Indexed: 12/14/2022] Open
Abstract
Obesity and its' associated metabolic diseases such as type 2 diabetes and cardiometabolic disorders are significant health problems confronting many countries. A major driver for developing obesity and metabolic dysfunction is the uncontrolled expansion of white adipose tissue (WAT). Specifically, the pathophysiological expansion of visceral WAT is often associated with metabolic dysfunction due to changes in adipokine secretion profiles, reduced vascularization, increased fibrosis, and enrichment of pro-inflammatory immune cells. A critical determinate of body fat distribution and WAT health is the sex steroid estrogen. The bioavailability of estrogen appears to favor metabolically healthy subcutaneous fat over visceral fat growth while protecting against changes in metabolic dysfunction. Our review will focus on the role of estrogen on body fat partitioning, WAT homeostasis, adipogenesis, adipocyte progenitor cell (APC) function, and thermogenesis to control WAT health and systemic metabolism.
Collapse
Affiliation(s)
| | - Daniel C. Berry
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| |
Collapse
|
14
|
Corona R, Ordaz B, Robles-Osorio L, Sabath E, Morales T. Neuroimmunoendocrine Link Between Chronic Kidney Disease and Olfactory Deficits. Front Integr Neurosci 2022; 16:763986. [PMID: 35173591 PMCID: PMC8841736 DOI: 10.3389/fnint.2022.763986] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 01/07/2022] [Indexed: 11/18/2022] Open
Abstract
Chronic kidney disease (CKD) is a multifactorial pathology that progressively leads to the deterioration of metabolic functions and results from deficient glomerular filtration and electrolyte imbalance. Its economic impact on public health is challenging. Mexico has a high prevalence of CKD that is strongly associated with some of the most common metabolic disorders like diabetes and hypertension. The gradual loss of kidney functions provokes an inflammatory state and endocrine alterations affecting several systems. High serum levels of prolactin have been associated with CKD progression, inflammation, and olfactory function. Also, the nutritional status is altered due to impaired renal function. The decrease in calorie and protein intake is often accompanied by malnutrition, which can be severe at advanced stages of the disease. Nutrition and olfactory functioning are closely interconnected, and CKD patients often complain of olfactory deficits, which ultimately can lead to deficient food intake. CKD patients present a wide range of deficits in olfaction like odor discrimination, identification, and detection threshold. The chronic inflammatory status in CKD damages the olfactory epithelium leading to deficiencies in the chemical detection of odor molecules. Additionally, the decline in cognitive functioning impairs the capacity of odor differentiation. It is not clear whether peritoneal dialysis and hemodialysis improve the olfactory deficits, but renal transplants have a strong positive effect. In the present review, we discuss whether the olfactory deficiencies caused by CKD are the result of the induced inflammatory state, the hyperprolactinemia, or a combination of both.
Collapse
Affiliation(s)
- Rebeca Corona
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | - Benito Ordaz
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | | | - Ernesto Sabath
- Facultad de Nutrición, Universidad Autónoma de Querétaro, Querétaro, Mexico
| | - Teresa Morales
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
| |
Collapse
|
15
|
Lin C, Chen DR, Kuo SJ, Feng CY, Chen DR, Hsieh WC, Lin PH. Profiling of Protein Adducts of Estrogen Quinones in 5-Year Survivors of Breast Cancer Without Recurrence. Cancer Control 2022; 29:10732748221084196. [PMID: 35303784 PMCID: PMC8935573 DOI: 10.1177/10732748221084196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aims The aim of this study was to simultaneously analyze estrogen quinone-derived adducts, including 17β-estradiol-2,3-quinone (E2-2,3-Q) and 17β-estradiol-3,4-quinone (E2-3,4-Q), in human albumin (Alb) and hemoglobin (Hb) derived from breast cancer patients with five-year postoperative treatment without recurrence in Taiwan and to evaluate the treatment-related effects on the production of these adducts. Settings and Design Cohort Methods and Material: Blood samples derived from breast cancer 5-year survivors without recurrence were collected. Albumin and hemoglobin adducts of E2-3,4-Q and E2-2,3-Q were analyzed to evaluate the degree of disposition of estrogen to quinones and to compare these adduct levels with those in patients before treatment. Statistical Analysis All data are expressed as mean ± standard deviation of three determinations. We used Student’s t-test to examine subgroups. Data were transformed to the natural logarithm and tested for normal distribution for parametric analyses. Linear correlations were investigated between individual adduct levels by simple regression. Statistical analysis was performed using the SPSS Statistics 20.0. Results Result confirmed that logged levels of E2-2,3-Q-derived adducts correlated significantly with those of E2-3,4-Q-derived adducts (correlation coefficient r=.336-.624). Mean levels of E2-2,3-Q-4-S-Alb and E2-3,4-Q-2-S-Alb in 5-year survivors were reduced by 60-70% when compared to those in the breast cancer patients with less than one year of diagnosis/preoperative treatment (P<.001). Conclusions Our findings add support to the theme that hormonal therapy including aromatase inhibitors and Tamoxifen may dramatically reduce burden of estrogen quinones. We hypothesize that combination of treatment-related effects and environmental factors may modulate estrogen homeostasis and diminish the production of estrogen quinones in breast cancer patients.
Collapse
Affiliation(s)
- Che Lin
- Comprehensive Breast Cancer Center, 36596Changhua Christian Hospital, Changhua, Taiwan.,Department of Optometry, 89578Chung Hwa University of Medical Technology, Tainan, Taiwan
| | - Ding-Ru Chen
- Comprehensive Breast Cancer Center, 36596Changhua Christian Hospital, Changhua, Taiwan
| | - Shou-Jen Kuo
- Comprehensive Breast Cancer Center, 36596Changhua Christian Hospital, Changhua, Taiwan
| | - Chi-Yen Feng
- Department of Surgery, 89578Da-Chien Health Medical System, Taiwan
| | - Dar-Ren Chen
- Comprehensive Breast Cancer Center, 36596Changhua Christian Hospital, Changhua, Taiwan
| | - Wei-Chung Hsieh
- Department of Laboratory Medicine, 384207Da-Chien General Hospital, Miaoli, Taiwan
| | - Po-Hsiung Lin
- Department of Environmental Engineering, 34916National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
16
|
Less Carcinogenic Chlorinated Estrogens Applicable to Hormone Replacement Therapy. Int J Mol Sci 2021; 22:ijms22137222. [PMID: 34281275 PMCID: PMC8268473 DOI: 10.3390/ijms22137222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/03/2021] [Accepted: 07/04/2021] [Indexed: 11/21/2022] Open
Abstract
Human estrogens prescribed for hormone replacement therapy (HRT) are known to be potent carcinogens. To find safer estrogens, several chlorinated estrogens were synthesized and their carcinogenic potential were determined. A pellet containing either 2-chloro-17β-estradiol (2-ClE2) or 4-chloro-17β-estradiol (4-ClE2) was implanted subcutaneously for 52 weeks into August Copenhagen Irish (ACI) rats, a preferred animal model for human breast cancer. 17β-Estradiol (E2) frequently induced mammary tumors while both 2-ClE2 and 4-ClE2 did not. Their 17α-ethinyl forms, thought to be orally active estrogens, were also synthesized. Neither 2-chloro-17α-ethinylestradiol (2-ClEE2) nor 4-chloro-17α-ethinylestradiol (4-ClEE2) induced tumors. The less carcinogenic effects were supported by histological examination of mammary glands of ACI rats treated with the chlorinated estrogens. A chlorine atom positioned at the 2- or 4-position of E2 may prevent the metabolic activation, resulting in reducing the carcinogenicity. 2-ClE2 and 4-ClE2 administered subcutaneously and 2-ClEE2 and 4-ClEE2 given orally to ovariectomized rats all showed uterotrophic potency, albeit slightly weaker than that of E2. Our results indicate that less carcinogenic chlorinated estrogens retaining estrogenic potential could be safer alternatives to the carcinogenic estrogens now in use for HRT.
Collapse
|
17
|
Cortés-Benítez F, Roy J, Perreault M, Maltais R, Poirier D. 16-Picolyl-androsterone derivative exhibits potent 17β-HSD3 inhibitory activity, improved metabolic stability and cytotoxic effect on various cancer cells: Synthesis, homology modeling and docking studies. J Steroid Biochem Mol Biol 2021; 210:105846. [PMID: 33609690 DOI: 10.1016/j.jsbmb.2021.105846] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 02/11/2021] [Accepted: 02/13/2021] [Indexed: 11/18/2022]
Abstract
A new androsterone derivative bearing a 16β-picolyl group (compound 5; FCO-586-119) was synthetized in four steps from the lead compound 1 (RM-532-105). We measured its inhibitory activity on 17β-HSD3 using microsomal fraction of rat testes as well as transfected LNCaP[17β-HSD3] cells. We then assessed its metabolic stability as well as its cytotoxic effect against a panel of cancer cell lines. The addition of a picolyl moiety at C-16 of RM-532-105 steroid core improves the 17β-HSD3 inhibitory activity in the microsomal fraction of rat testes, but not in whole LNCaP[17β-HSD3] cells. Interestingly, this structural modification enhances 3-fold the metabolic stability in conjunction with a significant cytotoxic effect against pancreatic, ovarian, breast, lung, and prostate cancer cells. Because the inhibitory activity data against 17β-HSD3 suggested that both steroid derivatives are non-competitive inhibitors, we performed docking and molecular dynamics simulations using a homology model of this membrane-associated enzyme. The results of these simulations revealed that both RM-532-105 (1) and FCO-586-119 (5) can compete for the cofactor-binding site displaying better binding energy than NADP+.
Collapse
Affiliation(s)
- Francisco Cortés-Benítez
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU De Québec - Research Center, Québec City, Québec, G1V 4G2, Canada; Laboratory of Synthesis and Isolation of Bioactive Substances, Department of Biological Systems, Biological and Health Sciences Division, Metropolitan Autonomous University- Xochimilco (UAM-X), Mexico City 04960, Mexico
| | - Jenny Roy
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU De Québec - Research Center, Québec City, Québec, G1V 4G2, Canada
| | - Martin Perreault
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU De Québec - Research Center, Québec City, Québec, G1V 4G2, Canada
| | - René Maltais
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU De Québec - Research Center, Québec City, Québec, G1V 4G2, Canada
| | - Donald Poirier
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU De Québec - Research Center, Québec City, Québec, G1V 4G2, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec City, Québec, G1V 0A6, Canada.
| |
Collapse
|
18
|
Gautam M, Thapa G. Cytochrome P450-mediated estrogen catabolism therapeutic avenues in epilepsy. Acta Neurol Belg 2021; 121:603-612. [PMID: 32743748 DOI: 10.1007/s13760-020-01454-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 07/23/2020] [Indexed: 01/11/2023]
Abstract
Epilepsy is a neuropsychiatric disorder, which does not have any identifiable cause. However, experimental and clinical results have asserted that the sex hormone estrogen level and endocrine system function influence the seizure and epileptic episodes. There are available drugs to control epilepsy, which passes through the metabolism process. Cytochrome P-450 family 1 (CYP1A1) is a heme-containing mono-oxygenase that are induced several folds in most of the tissues and cells contributing to their differential expression, which regulates various metabolic processes upon administration of therapeutics. CYP1A1 gene family has been found to metabolize estrogen, a female sex hormone, which plays a central role in maintaining the health of brain altering the level of estrogen active neuropsychiatric disorder like epilepsy. Hence, in this article, we endeavor to provide an opinion of estrogen, its effects on epilepsy and catamenial epilepsy, their metabolism by CYP1A1 and new way forward to differential diagnosis and clinical management of epilepsy in future.
Collapse
Affiliation(s)
- Megha Gautam
- Department of Biological Science, Faculty of Science and Engineering, Health Research Institute, University of Limerick, Limerick, V94 T9PX, Ireland
| | - Ganesh Thapa
- Department of Biological Science, Faculty of Science and Engineering, Health Research Institute, University of Limerick, Limerick, V94 T9PX, Ireland.
- Biohazards and Biosafety, Estates and Facilities, Trinity College of Dublin, The University of Dublin, College Green, Dublin 2, D02 PN40, Ireland.
| |
Collapse
|
19
|
Sprague R, Kim JW, Kirimlioglu E, Guo X, Günay N, Guzeloglu-Kayisli O, Ozmen A, Schatz F, Imudia AN, Lockwood CJ, Magness RR, Kayisli UA. Catecholestradiol Activation of Adrenergic Receptors Induces Endometrial Cell Survival via p38 MAPK Signaling. J Clin Endocrinol Metab 2021; 106:337-350. [PMID: 33247592 DOI: 10.1210/clinem/dgaa866] [Citation(s) in RCA: 7] [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: 01/28/2020] [Indexed: 02/03/2023]
Abstract
CONTEXT Enhanced levels of catecholestradiols, 2-hydroxyestradiol (2-OHE2) or 4-hydroxyestradiol (4-OHE2), are reported in endometriosis. During gestation, catecholestradiol activation of adrenergic receptors (AR) elevates estrogen receptor (ER)-independent proliferation of uterine arterial endothelial cells. OBJECTIVE To investigate β-AR-mediated catecholestradiol effects on human endometrial stromal cell (HESC) and epithelial cell survival in endometriosis. DESIGN β-AR immunostaining of eutopic and ectopic endometria (n = 9). Assays for cell viability, 5-bromo-2'-deoxyuridine proliferation, apoptosis, quantitative PCR, and estrogenicity (alkaline phosphatase activity), as well as siRNA β-AR silencing and immunoblot analyses of cultured HESCs or Ishikawa cells treated with control or 2-OHE2 or 4-OHE2 ±β-AR antagonist or ±p38 MAPK inhibitor. SETTING University research institution. PATIENTS Women with or without endometriosis. INTERVENTIONS None. MAIN OUTCOME MEASURES β-AR expression in eutopic vs ectopic endometria and regulation of HESC survival by 2-OHE2 and 4-OHE2. RESULTS Eutopic and ectopic endometrial stromal and epithelial cells displayed β2-AR immunoreactivity with increased staining in the functionalis vs basalis layer (P < 0.05). Both 2-OHE2 and 4-OHE2 enhanced HESC and Ishikawa cell survival (P < 0.05), an effect abrogated by β-AR antagonist propranolol, but not ER antagonist ICI182,780. 2-OHE2 or 4-OHE2 failed to induce cell survival and estrogenic activity in ADRB2-silenced HESCs and in Ishikawa cells, respectively. Although 2-OHE2 inhibited apoptosis and BAX mRNA expression, 4-OHE2 induced proliferation and decreased apoptosis (P < 0.05). Both catecholestradiols elevated phospho-p38 MAPK levels (P < 0.05), which was blocked by propranolol, and p38 MAPK inhibitor reversed catecholestradiol-enhanced HESC survival. CONCLUSIONS Catecholestradiols increase endometrial cell survival by an ER-independent β-AR-mediated p38 MAPK activation, suggesting that agents blocking β-AR (e.g., propranolol) or inhibiting 2-OHE2- or 4-OHE2-generating enzymes (i.e., CYP1A1/B1) could treat endometriosis.
Collapse
Affiliation(s)
- Rachel Sprague
- Department of Obstetrics & Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Joung W Kim
- Department of Obstetrics & Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Esma Kirimlioglu
- Department of Obstetrics & Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Xiaofang Guo
- Department of Obstetrics & Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Nihan Günay
- Department of Obstetrics & Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Ozlem Guzeloglu-Kayisli
- Department of Obstetrics & Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Asli Ozmen
- Department of Obstetrics & Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Frederick Schatz
- Department of Obstetrics & Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Anthony N Imudia
- Department of Obstetrics & Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Charles J Lockwood
- Department of Obstetrics & Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Ronald R Magness
- Department of Obstetrics & Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Umit A Kayisli
- Department of Obstetrics & Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| |
Collapse
|
20
|
Merico V, Zanoni M, Parada-Bustamante A, Garagna S, Zuccotti M. In Vitro Maturation of Fully Grown Mouse Antral Follicles in the Presence of 1 nM 2-Hydroxyestradiol Improves Oocytes' Developmental Competence. Reprod Sci 2020; 28:121-133. [PMID: 32757137 PMCID: PMC7782423 DOI: 10.1007/s43032-020-00276-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 07/24/2020] [Indexed: 11/06/2022]
Abstract
Cathecolestrogens are estradiol metabolites produced during folliculogenesis in the mammalian ovary. 2-Hydroxyestradiol (2-OHE2) is one of the most abundant although its role remains unknown. The aim of this study is to investigate whether the presence of 2-OHE2 during the germinal vesicle-to-metaphase II transition affects oocyte meiotic and preimplantation developmental competence. Mouse cumulus-oocyte complexes (COCs), isolated from fully grown antral follicles, were in vitro–matured (IVM) in the presence of 2-OHE2 (0.1, 1, 10 or 100 nM) for 6 or 15 h; then, their meiotic and developmental competence was evaluated using a number of cytological quality markers. With the exception of the highest dose (100 nM), the addition of 2-OHE2 to the IVM medium, did not alter, compared with untreated control, the frequency of oocytes that reached the MII stage. Instead, IVM in the presence of 1 nM 2-OHE2 highly increased the rate of preimplantation development and blastocyst quality. To understand whether this positive effect could be attributed to the events occurring during meiosis resumption, we analysed a number of specific cytological quality markers of the asymmetric division, such as PB-I volume and position, presence and extension of the cortical F-actin cap, meiotic spindle shape and area, and microtubule organisation centre localisation. The results highlighted how the presence of 1 nM 2-OHE2 significantly improved the overall cytological organisation required for a correct asymmetric division. Our results contribute a first step to acknowledge a potential role of this estradiol metabolite during the GV-to-MII transition, contributing to the acquisition of oocytes developmental competence.
Collapse
Affiliation(s)
- Valeria Merico
- Laboratorio di Biologia dello Sviluppo, Dipartimento di Biologia e Biotecnologie "Lazzaro Spallanzani", University of Pavia, Via Ferrata, 9, 27100, Pavia, Italy. .,Centre for Health Technologies (C.H.T.), University of Pavia, Via Ferrata, 9, 27100, Pavia, Italy.
| | - Mario Zanoni
- Laboratorio di Biologia dello Sviluppo, Dipartimento di Biologia e Biotecnologie "Lazzaro Spallanzani", University of Pavia, Via Ferrata, 9, 27100, Pavia, Italy.,Centre for Health Technologies (C.H.T.), University of Pavia, Via Ferrata, 9, 27100, Pavia, Italy
| | - Alexis Parada-Bustamante
- Institute of Maternal and Child Research, School of Medicine, University of Chile, Santiago, Chile
| | - Silvia Garagna
- Laboratorio di Biologia dello Sviluppo, Dipartimento di Biologia e Biotecnologie "Lazzaro Spallanzani", University of Pavia, Via Ferrata, 9, 27100, Pavia, Italy. .,Centre for Health Technologies (C.H.T.), University of Pavia, Via Ferrata, 9, 27100, Pavia, Italy.
| | - Maurizio Zuccotti
- Laboratorio di Biologia dello Sviluppo, Dipartimento di Biologia e Biotecnologie "Lazzaro Spallanzani", University of Pavia, Via Ferrata, 9, 27100, Pavia, Italy. .,Centre for Health Technologies (C.H.T.), University of Pavia, Via Ferrata, 9, 27100, Pavia, Italy.
| |
Collapse
|
21
|
Amare DE. Anti-Cancer and Other Biological Effects of a Dietary Compound 3,3ʹ-Diindolylmethane Supplementation: A Systematic Review of Human Clinical Trials . NUTRITION AND DIETARY SUPPLEMENTS 2020. [DOI: 10.2147/nds.s261577] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
|
22
|
Imbalances in the disposition of estrogen and naphthalene in breast cancer patients: a potential biomarker of breast cancer risk. Sci Rep 2020; 10:11773. [PMID: 32678225 PMCID: PMC7366907 DOI: 10.1038/s41598-020-68814-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 06/29/2020] [Indexed: 12/21/2022] Open
Abstract
Elevation of naphthoquinones and estrogen quinones, which are reactive metabolites of naphthalene and estrogen, is thought to be an important indicator of naphthalene- and estrogen-induced carcinogenesis. We compared background levels of naphthalene and estrogen quinone-derived adducts in serum albumin (Alb) from 143 women with breast cancer and 119 healthy controls. Cysteinyl adducts of naphthoquinones, including 1,2-naphthoquinone (1,2-NPQ) and 1,4-naphthoquinone (1,4-NPQ), and estrogen quinones, including estrogen-2,3-quinones (E2-2,3-Q) and estrogen-3,4-quinones (E2-3,4-Q), were characterized after adduct cleavage. Levels of estrogen quinones and naphthoquinones were positively correlated in healthy controls, but not in breast cancer patients (p < 0.05). Compared with controls, levels of 1,2-NPQ and E2-3,4-Q were elevated by two- to ten-fold in cancer patients (p < 0.001). To explore the correlation between estrogen- and naphthalene-derived quinone adducts and disease status, we performed linear discriminant analysis of the ratio of 1,2-NPQ-Alb to (1,2-NPQ-Alb plus 1,4-NPQ-Alb) versus the ratio of E2-3,4-Q-2-S-Alb to (E2-2,3-Q-4-S-Alb plus E2-3,4-Q-2-S-Alb) in patients and controls. These two groups were separable using albumin adducts of estrogen quinones and naphthoquinones, with 99.6% overall correct classification rate (overall accuracy). The findings of this study suggest that differences in the disposition of estrogen and naphthalene, and the subsequent elevation of cumulative E2-3,4-Q and 1,2-NPQ may serve as biomarkers of breast cancer risk.
Collapse
|
23
|
Okamoto Y, Jinno H, Itoh S, Shibutani S. Carcinogenic potential of fluorinated estrogens in mammary tumorigenesis. Toxicol Lett 2019; 318:99-103. [PMID: 31669098 DOI: 10.1016/j.toxlet.2019.10.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 10/18/2019] [Accepted: 10/18/2019] [Indexed: 11/17/2022]
Abstract
Fluorination preventing metabolic hydroxylation of 17β-estradiol (E2) was applied to investigate the mechanisms underlying estrogen-induced carcinogenesis. Either 2-fluoro-17β-estradiol (2-FE2) or 4-fluoro-17β-estradiol (4-FE2) was administered subcutaneously for 52 weeks to August Copenhagen Irish (ACI) rats, the preferred animal model for human breast cancer. 4-FE2 induced frequent mammary tumors whereas 2-FE2 did not. The cumulative incidence of mammary tumors in rats treated with 4-FE2 was comparable to that observed with E2. The carcinogenic results were supported by histological examination of mammary glands of fluorinated estrogen-treated ACI rats. To evaluate the estrogenic potential of the fluorinated estrogens, 2-FE2 or 4-FE2 was administrated subcutaneously to ovariectomized rats. Both 4-FE2 and 2-FE2 showed high uterotrophic potency. Our results indicate that estrogenic potential may not be the sole factor driving mammary tumorigenesis. Since fluorination inhibits metabolic hydroxylation of E2 at the substituted position, the carcinogenic effect may occur through the metabolic activation of 2-hydroxylated E2, in combination with the compound's estrogenic potency.
Collapse
Affiliation(s)
- Yoshinori Okamoto
- Faculty of Pharmacy, Meijo University, Yagotoyama, Nagoya 468-8503, Japan
| | - Hideto Jinno
- Faculty of Pharmacy, Meijo University, Yagotoyama, Nagoya 468-8503, Japan
| | - Shinji Itoh
- Faculty of Pharmaceutical Sciences, Hokkaido University of Science, Maeda, Sapporo, Hokkaido 006-8585, Japan
| | - Shinya Shibutani
- Department of Pharmacological Sciences, State University of New York at Stony Brook, Stony Brook, New York 11794-8651, USA.
| |
Collapse
|
24
|
Ervin SM, Li H, Lim L, Roberts LR, Liang X, Mani S, Redinbo MR. Gut microbial β-glucuronidases reactivate estrogens as components of the estrobolome that reactivate estrogens. J Biol Chem 2019; 294:18586-18599. [PMID: 31636122 DOI: 10.1074/jbc.ra119.010950] [Citation(s) in RCA: 188] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 10/17/2019] [Indexed: 02/06/2023] Open
Abstract
Gut microbial β-glucuronidase (GUS) enzymes have been suggested to be involved in the estrobolome, the collection of microbial reactions involving estrogens. Furthermore, bacterial GUS enzymes within the gastrointestinal tract have been postulated to be a contributing factor in hormone-driven cancers. However, to date, there has been no experimental evidence to support these hypotheses. Here we provide the first in vitro analysis of the ability of 35 human gut microbial GUS enzymes to reactivate two distinct estrogen glucuronides, estrone-3-glucuronide and estradiol-17-glucuronide, to estrone and estradiol, respectively. We show that certain members within the Loop 1, mini-Loop 1, and FMN-binding classes of gut microbial GUS enzymes can reactivate estrogens from their inactive glucuronides. We provide molecular details of key interactions that facilitate these catalytic processes and present the structures of two novel human gut microbial GUS enzymes related to the estrobolome. Further, we demonstrate that estrogen reactivation by Loop 1 bacterial GUS enzymes can be inhibited both in purified enzymes and in fecal preparations of mixed murine fecal microbiota. Finally, however, despite these in vitro and ex vivo data, we show that a Loop 1 GUS-specific inhibitor is not capable of reducing the development of tumors in the PyMT mouse model of breast cancer. These findings validate that gut microbial GUS enzymes participate in the estrobolome but also suggest that the estrobolome is a multidimensional set of processes on-going within the mammalian gastrointestinal tract that likely involves many enzymes, including several distinct types of GUS proteins.
Collapse
Affiliation(s)
- Samantha M Ervin
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Hao Li
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Lauren Lim
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Lee R Roberts
- Exploratory Science Center, Merck & Co., Inc., Cambridge, Massachusetts 02141
| | - Xue Liang
- Exploratory Science Center, Merck & Co., Inc., Cambridge, Massachusetts 02141
| | - Sridhar Mani
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Matthew R Redinbo
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599; Integrated Program for Biological and Genome Sciences and Departments of Biochemistry and Microbiology, University of North Carolina, Chapel Hill, North Carolina 27599.
| |
Collapse
|
25
|
Cortés-Benítez F, Roy J, Perreault M, Maltais R, Poirier D. A- and D-Ring Structural Modifications of an Androsterone Derivative Inhibiting 17β-Hydroxysteroid Dehydrogenase Type 3: Chemical Synthesis and Structure–Activity Relationships. J Med Chem 2019; 62:7070-7088. [DOI: 10.1021/acs.jmedchem.9b00624] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Francisco Cortés-Benítez
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Quebéc—Research Center, Québec, Québec G1V 4G2, Canada
- Department of Biological Systems, Biological and Health Sciences Division, Metropolitan Autonomous University—Campus Xochimilco (UAM-X), Mexico City 04960, Mexico
- Department of Pharmacy, Faculty of Chemistry, National Autonomous University of Mexico, Mexico City 04510, Mexico
| | - Jenny Roy
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Quebéc—Research Center, Québec, Québec G1V 4G2, Canada
| | - Martin Perreault
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Quebéc—Research Center, Québec, Québec G1V 4G2, Canada
| | - René Maltais
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Quebéc—Research Center, Québec, Québec G1V 4G2, Canada
| | - Donald Poirier
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Quebéc—Research Center, Québec, Québec G1V 4G2, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, Québec G1V 0A6, Canada
| |
Collapse
|
26
|
Grossmann M, Wierman ME, Angus P, Handelsman DJ. Reproductive Endocrinology of Nonalcoholic Fatty Liver Disease. Endocr Rev 2019; 40:417-446. [PMID: 30500887 DOI: 10.1210/er.2018-00158] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 11/19/2018] [Indexed: 02/07/2023]
Abstract
The liver and the reproductive system interact in a multifaceted bidirectional fashion. Sex steroid signaling influences hepatic endobiotic and xenobiotic metabolism and contributes to the pathogenesis of functional and structural disorders of the liver. In turn, liver function affects the reproductive axis via modulating sex steroid metabolism and transport to tissues via sex hormone-binding globulin (SHBG). The liver senses the body's metabolic status and adapts its energy homeostasis in a sex-dependent fashion, a dimorphism signaled by the sex steroid milieu and possibly related to the metabolic costs of reproduction. Sex steroids impact the pathogenesis of nonalcoholic fatty liver disease, including development of hepatic steatosis, fibrosis, and carcinogenesis. Preclinical studies in male rodents demonstrate that androgens protect against hepatic steatosis and insulin resistance both via androgen receptor signaling and, following aromatization to estradiol, estrogen receptor signaling, through regulating genes involved in hepatic lipogenesis and glucose metabolism. In female rodents in contrast to males, androgens promote hepatic steatosis and dysglycemia, whereas estradiol is similarly protective against liver disease. In men, hepatic steatosis is associated with modest reductions in circulating testosterone, in part consequent to a reduction in circulating SHBG. Testosterone treatment has not been demonstrated to improve hepatic steatosis in randomized controlled clinical trials. Consistent with sex-dimorphic preclinical findings, androgens promote hepatic steatosis and dysglycemia in women, whereas endogenous estradiol appears protective in both men and women. In both sexes, androgens promote hepatic fibrosis and the development of hepatocellular carcinoma, whereas estradiol is protective.
Collapse
Affiliation(s)
- Mathis Grossmann
- Department of Medicine Austin Health, University of Melbourne, Heidelberg, Victoria, Australia.,Department of Endocrinology, Austin Health, Heidelberg, Victoria, Australia
| | - Margaret E Wierman
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado.,Research Service, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, Colorado
| | - Peter Angus
- Department of Medicine Austin Health, University of Melbourne, Heidelberg, Victoria, Australia.,Departments of Gastroenterology and Hepatology, Heidelberg, Victoria, Australia
| | - David J Handelsman
- ANZAC Research Institute, University of Sydney, Concord Hospital, Sydney, New South Wales, Australia
| |
Collapse
|
27
|
QSAR modelling of a large imbalanced aryl hydrocarbon activation dataset by rational and random sampling and screening of 80,086 REACH pre-registered and/or registered substances. PLoS One 2019; 14:e0213848. [PMID: 30870500 PMCID: PMC6417725 DOI: 10.1371/journal.pone.0213848] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 03/01/2019] [Indexed: 12/02/2022] Open
Abstract
The Aryl hydrocarbon receptor (AhR) plays important roles in many normal and pathological physiological processes, including endocrine homeostasis, foetal development, cell cycle regulation, cellular oxidation/antioxidation, immune regulation, metabolism of endogenous and exogenous substances, and carcinogenesis. An experimental data set for human in vitro AhR activation comprising 324,858 substances, of which 1,982 were confirmed actives, was used to test an in-house-developed approach to rationally select Quantitative Structure-Activity Relationship (QSAR) training set substances from an unbalanced data set. In the first iteration, active and inactive substances were selected by random to make QSAR models. Then, more inactive substances were added to the training set in two further iterations based on incorrect or out-of-domain predictions to produce larger models. The resulting ‘rational’ model, comprising 832 actives and four times as many inactives, i.e. 3,328, was compared to a model with a training set of same size and proportion of inactives chosen entirely by random. Both models underwent robust cross-validation and external validation showing good statistical performance, with the rational model having external validation sensitivity of 85.1% and specificity of 97.1%, compared to the random model with sensitivity 89.1% and specificity 91.3%. Furthermore, we integrated the training sets for both models with the 93 external validation test set actives and 372 randomly selected inactives to make two final models. They also underwent external validations for specificity and cross-validations, which confirmed that good predictivity was maintained. All developed models were applied to predict 80,086 EU REACH substances. The rational and random final models had 63.1% and 56.9% coverage of the REACH set, respectively, and predicted 1,256 and 3,214 substances as actives. The final models as well as predictions for AhR activation for 650,000 substances will be published in the Danish (Q)SAR Database and can, for example, be used for priority setting, in read-across predictions and in weight-of-evidence assessments of chemicals.
Collapse
|
28
|
Arnst KE, Banerjee S, Chen H, Deng S, Hwang DJ, Li W, Miller DD. Current advances of tubulin inhibitors as dual acting small molecules for cancer therapy. Med Res Rev 2019; 39:1398-1426. [PMID: 30746734 DOI: 10.1002/med.21568] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 01/16/2019] [Accepted: 01/19/2019] [Indexed: 12/25/2022]
Abstract
Microtubule (MT)-targeting agents are highly successful drugs as chemotherapeutic agents, and this is attributed to their ability to target MT dynamics and interfere with critical cellular functions, including, mitosis, cell signaling, intracellular trafficking, and angiogenesis. Because MT dynamics vary in the different stages of the cell cycle, these drugs tend to be the most effective against mitotic cells. While this class of drug has proven to be effective against many cancer types, significant hurdles still exist and include overcoming aspects such as dose limited toxicities and the development of resistance. Newer generations of developed drugs attack these problems and alternative approaches such as the development of dual tubulin and kinase inhibitors are being investigated. This approach offers the potential to show increased efficacy and lower toxicities. This review covers different categories of MT-targeting agents, recent advances in dual inhibitors, and current challenges for this drug target.
Collapse
Affiliation(s)
- Kinsie E Arnst
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Souvik Banerjee
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Hao Chen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Shanshan Deng
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Dong-Jin Hwang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Wei Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Duane D Miller
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
29
|
Knutsen HK, Alexander J, Barregård L, Bignami M, Brüschweiler B, Ceccatelli S, Cottrill B, Dinovi M, Edler L, Grasl-Kraupp B, Hogstrand C, Nebbia CS, Oswald IP, Petersen A, Rose M, Roudot AC, Schwerdtle T, Vleminckx C, Vollmer G, Wallace H, Fürst P, Håkansson H, Halldorsson T, Lundebye AK, Pohjanvirta R, Rylander L, Smith A, van Loveren H, Waalkens-Berendsen I, Zeilmaker M, Binaglia M, Gómez Ruiz JÁ, Horváth Z, Christoph E, Ciccolallo L, Ramos Bordajandi L, Steinkellner H, Hoogenboom LR. Risk for animal and human health related to the presence of dioxins and dioxin-like PCBs in feed and food. EFSA J 2018; 16:e05333. [PMID: 32625737 PMCID: PMC7009407 DOI: 10.2903/j.efsa.2018.5333] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The European Commission asked EFSA for a scientific opinion on the risks for animal and human health related to the presence of dioxins (PCDD/Fs) and DL-PCBs in feed and food. The data from experimental animal and epidemiological studies were reviewed and it was decided to base the human risk assessment on effects observed in humans and to use animal data as supportive evidence. The critical effect was on semen quality, following pre- and postnatal exposure. The critical study showed a NOAEL of 7.0 pg WHO2005-TEQ/g fat in blood sampled at age 9 years based on PCDD/F-TEQs. No association was observed when including DL-PCB-TEQs. Using toxicokinetic modelling and taking into account the exposure from breastfeeding and a twofold higher intake during childhood, it was estimated that daily exposure in adolescents and adults should be below 0.25 pg TEQ/kg bw/day. The CONTAM Panel established a TWI of 2 pg TEQ/kg bw/week. With occurrence and consumption data from European countries, the mean and P95 intake of total TEQ by Adolescents, Adults, Elderly and Very Elderly varied between, respectively, 2.1 to 10.5, and 5.3 to 30.4 pg TEQ/kg bw/week, implying a considerable exceedance of the TWI. Toddlers and Other Children showed a higher exposure than older age groups, but this was accounted for when deriving the TWI. Exposure to PCDD/F-TEQ only was on average 2.4- and 2.7-fold lower for mean and P95 exposure than for total TEQ. PCDD/Fs and DL-PCBs are transferred to milk and eggs, and accumulate in fatty tissues and liver. Transfer rates and bioconcentration factors were identified for various species. The CONTAM Panel was not able to identify reference values in most farm and companion animals with the exception of NOAELs for mink, chicken and some fish species. The estimated exposure from feed for these species does not imply a risk.
Collapse
|
30
|
Lin PH, Yang HJ, Hsieh WC, Lin C, Chan YC, Wang YF, Yang YT, Lin KJ, Lin LS, Chen DR. Albumin and hemoglobin adducts of estrogen quinone as biomarkers for early detection of breast cancer. PLoS One 2018; 13:e0201241. [PMID: 30222738 PMCID: PMC6141067 DOI: 10.1371/journal.pone.0201241] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 07/11/2018] [Indexed: 12/21/2022] Open
Abstract
Cumulative estrogen concentration is an important determinant of the risk of developing breast cancer. Estrogen carcinogenesis is attributed to the combination of receptor-driven mitogenesis and DNA damage induced by quinonoid metabolites of estrogen. The present study was focused on developing an improved breast cancer prediction model using estrogen quinone-protein adduct concentrations. Blood samples from 152 breast cancer patients and 71 healthy women were collected, and albumin (Alb) and hemoglobin (Hb) adducts of estrogen-3,4-quinone and estrogen-2,3-quinone were extracted and evaluated as potential biomarkers of breast cancer. A multilayer perceptron (MLP) was used as the predictor model and the resultant prediction of breast cancer was more accurate than other existing detection methods. A MLP using the logarithm of the concentrations of the estrogen quinone-derived adducts (four input nodes, 10 hidden nodes, and one output node) was used to predict breast cancer risk with accuracy close to 100% and area under curve (AUC) close to one. The AUC value of one showed that both data sets were separable. We conclude that Alb and Hb adducts of estrogen quinones are promising biomarkers for the early detection of breast cancer.
Collapse
Affiliation(s)
- Po-Hsiung Lin
- Department of Environmental Engineering, National Chung Hsing University, South Dist., Taichung, Taiwan, R.O.C
| | - Hui-Ju Yang
- Department of Dermatology, Changhua Christian Hospital, Changhua, Taiwan, R.O.C
| | - Wei-Chung Hsieh
- Department of Internal Medicine, Da-Chien General Hospital, Miaoli, Taiwan, R.O.C
| | - Che Lin
- Comprehensive Breast Cancer Center, Changhua Christian Hospital, Changhua, Taiwan, R.O.C
| | - Ya-Chi Chan
- Cancer Research Center, Department of Research, Changhua Christian Hospital, Changhua, Taiwan, R.O.C
| | - Yu-Fen Wang
- Cancer Research Center, Department of Research, Changhua Christian Hospital, Changhua, Taiwan, R.O.C
| | - Yuan-Ting Yang
- Department of Pharmacy, Changhua Christian Hospital, Changhua, Taiwan, R.O.C
| | - Kuo-Juei Lin
- Department of Surgery, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan, R.O.C
| | - Li-Sheng Lin
- Department of Breast Surgery, the Affiliated Hospital (Group) of Putian University, Putian, Fujian, China
| | - Dar-Ren Chen
- Comprehensive Breast Cancer Center, Changhua Christian Hospital, Changhua, Taiwan, R.O.C
- Cancer Research Center, Department of Research, Changhua Christian Hospital, Changhua, Taiwan, R.O.C
- School of Medicine, Chung Shan Medical University, South Dist., Taichung, Taiwan, R.O.C
- * E-mail:
| |
Collapse
|
31
|
Thomas P. Reprint of "Role of G protein-coupled estrogen receptor (GPER/GPR30) in maintenance of meiotic arrest in fish oocytes". J Steroid Biochem Mol Biol 2018; 176:23-30. [PMID: 29102625 DOI: 10.1016/j.jsbmb.2017.10.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 11/23/2016] [Accepted: 12/16/2016] [Indexed: 01/07/2023]
Abstract
An essential role for GPER (formerly known as GPR30) in regulating mammalian reproduction has not been identified to date, although it has shown to be involved in the regulation a broad range of other estrogen-dependent functions. In contrast, an important reproductive role for GPER in the maintenance of oocyte meiotic arrest has been identified in teleost fishes, which is briefly reviewed here. Recent studies have clearly shown that ovarian follicle production of estradiol-17β (E2) maintains meiotic arrest in several teleost species through activation of GPER coupled to a stimulatory G protein (Gs) on oocyte plasma membranes, resulting in stimulation of cAMP production and maintenance of elevated cAMP levels. Studies with denuded zebrafish oocytes and with microinjection of GPER antisense oligonucleotides into oocytes have demonstrated the requirement for both ovarian follicle production of estrogens and expression of GPER on the oocyte surface for maintenance of meiotic arrest. This inhibitory action of E2 on the resumption of meiosis is mimicked by the GPER-selective agonist G-1, by the GPER agonists and nuclear ER antagonists, ICI 182,780 and tamoxifen, and also by the xenoestrogen bisphenol-A (BPA) and related alkylphenols. GPER also maintains meiotic arrest of zebrafish oocytes through estrogen- and BPA-dependent GPER activation of epidermal growth factor receptor (EGFR) and mitogen-activated protein kinase (MAPK) signaling. Interestingly, progesterone receptor component 1 (PGRMC1) is also involved in estrogen maintenance of meiotic arrest through regulation of EGFR expression on the oocyte plasma membrane. The preovulatory surge in LH secretion induces the ovarian synthesis of progestin hormones that activate a membrane progestin receptor alpha (mPRα)/inhibitory G protein (Gi) pathway. It also increases ovarian synthesis of the catecholestrogen, 2-hydroxy-estradiol-17β (2-OHE2) which inhibits the GPER/Gs/adenylyl cyclase pathway. Both of these LH actions cause declines in oocyte cAMP levels resulting in the resumption of meiosis. GPER is also present on murine oocytes but there are no reports of studies investigating its possible involvement in maintaining meiotic arrest in mammals.
Collapse
Affiliation(s)
- Peter Thomas
- Marine Science Institute, University of Texas at Austin, 750 Channel View Drive, Port Aransas, TX 78373, United States.
| |
Collapse
|
32
|
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: 49] [Impact Index Per Article: 6.1] [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
|
33
|
Thomas P. Role of G-protein-coupled estrogen receptor (GPER/GPR30) in maintenance of meiotic arrest in fish oocytes. J Steroid Biochem Mol Biol 2017; 167:153-161. [PMID: 28007532 DOI: 10.1016/j.jsbmb.2016.12.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 11/23/2016] [Accepted: 12/16/2016] [Indexed: 02/08/2023]
Abstract
An essential role for GPER (formerly known as GPR30) in regulating mammalian reproduction has not been identified to date, although it has shown to be involved in the regulation a broad range of other estrogen-dependent functions. In contrast, an important reproductive role for GPER in the maintenance of oocyte meiotic arrest has been identified in teleost fishes, which is briefly reviewed here. Recent studies have clearly shown that ovarian follicle production of estradiol-17β (E2) maintains meiotic arrest in several teleost species through activation of GPER coupled to a stimulatory G protein (Gs) on oocyte plasma membranes resulting in stimulation of cAMP production and maintenance of elevated cAMP levels. Studies with denuded zebrafish oocytes and with microinjection of GPER antisense oligonucleotides into oocytes have demonstrated the requirement for both ovarian follicle production of estrogens and expression of GPER on the oocyte surface for maintenance of meiotic arrest. This inhibitory action of E2 on the resumption of meiosis is mimicked by the GPER-selective agonist G-1, by the GPER agonists and nuclear ER antagonists, ICI 182,780 and tamoxifen, and also by the xenoestrogen bisphenol-A (BPA) and related alkylphenols. GPER also maintains meiotic arrest of zebrafish oocytes through estrogen- and BPA-dependent GPER activation of epidermal growth factor receptor (EGFR) and mitogen-activated protein kinase (MAPK) signaling. Interestingly, progesterone receptor component 1 (PGRMC1) is also involved in estrogen maintenance of meiotic arrest through regulation of EGFR expression on the oocyte plasma membrane. The preovulatory surge in LH secretion induces the ovarian synthesis of progestin hormones that activate a membrane progestin receptor alpha (mPRα)/inhibitory G protein (Gi) pathway. It also increases ovarian synthesis of the catecholestrogen, 2-hydroxy-estradiol-17β (2-OHE2) which inhibits the GPER/Gs/adenylyl cyclase pathway. Both of these LH actions cause declines in oocyte cAMP levels resulting in the resumption of meiosis. GPER is also present on murine oocytes but there are no reports of studies investigating its possible involvement in maintaining meiotic arrest in mammals.
Collapse
Affiliation(s)
- Peter Thomas
- Marine Science Institute, University of Texas at Austin, 750 Channel View Drive, Port Aransas, TX 78373, United States.
| |
Collapse
|
34
|
Wang P, Zhu BT. Unique effect of 4-hydroxyestradiol and its methylation metabolites on lipid and cholesterol profiles in ovariectomized female rats. Eur J Pharmacol 2017; 800:107-117. [PMID: 28219710 DOI: 10.1016/j.ejphar.2017.02.032] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 02/14/2017] [Accepted: 02/17/2017] [Indexed: 10/20/2022]
Abstract
Animal studies have shown that endogenous estrogens such as 17β-estradiol (E2) can modulate lipid profiles in vivo, and this effect is generally thought to be mediated by the estrogen receptors (ERs). The present study sought to test a hypothesis that some of the endogenous estrogen metabolites that have very weak estrogenic activity may exert some of their modulating effects on lipid metabolism in an ER-independent manner. Using ovariectomized female rats as an in vivo model, we found that 4-hydroxyestradiol (4-OH-E2) has a markedly stronger effect in reducing the adipocyte size and serum cholesterol level in rats compared to E2, despite the weaker estrogenic activity of 4-OH-E2. Moreover, when E2 or 4-OH-E2 is used in combination with ICI-182,780 (an ER antagonist), some of their lipid-modulating effects are not blocked by this antiestrogen. Interestingly, two of the O-methylation metabolites of 4-OH-E2, namely, 4-methoxyestradiol and 4-methoxyestrone, which have much weaker estrogenic activity, were also found to have similar lipid-modulating effects compared to 4-OH-E2. Mechanistically, up-regulation of the expression of leptin, cytochrome P450 7A1 and LXRα genes is observed in the liver of animals treated with E2 or 4-OH-E2, and the up-regulation is essentially not inhibited by co-treatment with ICI-182,780. These results demonstrate that some of the endogenous E2 metabolites are functionally important modulators of lipid metabolic profiles in vivo. In addition, our findings indicate that an ER-independent pathway likely mediates some of the lipid-modulating effects of endogenous estrogens and their metabolic derivatives.
Collapse
Affiliation(s)
- Pan Wang
- Department of Pharmacology, Toxicology and Therapeutics, School of Medicine, The University of Kansas Medical Center, Kansas City, KS 66160, USA; Laboratory of Molecular Toxicology, State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Bao-Ting Zhu
- Department of Pharmacology, Toxicology and Therapeutics, School of Medicine, The University of Kansas Medical Center, Kansas City, KS 66160, USA; School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen, Shenzhen, Guangdong 518172, China.
| |
Collapse
|
35
|
Cortés-Benítez F, Roy J, Maltais R, Poirier D. Impact of androstane A- and D-ring inversion on 17β-hydroxysteroid dehydrogenase type 3 inhibitory activity, androgenic effect and metabolic stability. Bioorg Med Chem 2017; 25:2065-2073. [PMID: 28254377 DOI: 10.1016/j.bmc.2017.02.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 01/31/2017] [Accepted: 02/06/2017] [Indexed: 10/20/2022]
Abstract
17β-Hydroxysteroid dehydrogenase type 3 (17β-HSD3) is a major player in human endocrinology, being one of the most important enzymes involved in testosterone production. To capitalize on the discovery of RM-532-105, a steroidal 17β-HSD3 inhibitor, we explored the effect of its backbone configuration on inhibitory activity, androgenic profile, and metabolic stability. Two modifications that greatly alter the natural shape of steroids, i.e. inversion of the methyl on carbon 13 (13α-CH3 instead of 13β-CH3) and inversion of the hydrogen on carbon 5 (5β-H instead of 5α-H), were tested after the syntheses in 6 steps of 2 isomeric forms (5α/13α-RM-532-105 (6a) and 5β/13β-RM-532-105 (6b), respectively) of the 17β-HSD3 inhibitor RM-532-105 (5α/13β-configurations). For compound 6b, a cis/trans junction of the A/B rings did not significantly alter the inhibitory activity on 17β-HSD3 (IC50=0.15μM) as well as the liver microsomal stability (16.6% of 6b remaining after 1h incubation) compared to RM-532-105 (IC50=0.11μM and 14.1% remaining). In contrast, a trans/cis junction of C/D rings reduced the inhibitory activity on 17β-HSD3 (IC50=1.09μM) but increased the metabolic stability with 29.4% of compound 6a remaining after incubation. The structural modifications represented by compounds 6a and 6b did not change the non-androgenicity profile of an androsterone derivative such as RM-532-105, but slightly increased its cytotoxic activity.
Collapse
Affiliation(s)
- Francisco Cortés-Benítez
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec - Research Center, Québec, Québec, Canada; Department of Pharmacy, Faculty of Chemistry, National Autonomous University of Mexico, Mexico City 04510, Mexico
| | - Jenny Roy
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec - Research Center, Québec, Québec, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, Québec, Canada
| | - René Maltais
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec - Research Center, Québec, Québec, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, Québec, Canada
| | - Donald Poirier
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec - Research Center, Québec, Québec, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, Québec, Canada.
| |
Collapse
|
36
|
Genistein attenuates D-GalN induced liver fibrosis/chronic liver damage in rats by blocking the TGF-β/Smad signaling pathways. Chem Biol Interact 2017; 261:80-85. [DOI: 10.1016/j.cbi.2016.11.022] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 10/29/2016] [Accepted: 11/18/2016] [Indexed: 01/07/2023]
|
37
|
Lee SR, Lee SY, Kim SY, Ryu SY, Park BK, Hong EJ. Hydroxylation and sulfation of sex steroid hormones in inflammatory liver. J Biomed Res 2017; 31:437-444. [PMID: 28866654 PMCID: PMC5706436 DOI: 10.7555/jbr.31.20170031] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Sex steroids, also known as gonadal steroids, are oxidized with hydroxylation by cytochrome P450, glucuronidation by UDP-glucuronosyltransferase, sulfation by sulfotransferase, andO-methylation by catechol O-methyltransferase. Thus, it is important to determine the process by which inflammation influences metabolism of gonadal hormones. Therefore, we investigated the mechanism of metabolic enzymes against high physiologic inflammatory responsein vivo to study their biochemical properties in liver diseases. In this study, C57BL/6N mice were induced with hepatic inflammation by diethylnitrosamine (DEN) exposure. We observed upregulation of Cyp19a1, Hsd17b1, Cyp1a1, Sult1e1 in the DEN-treated livers compared to the control-treated livers using real time PCR. Moreover, the increased Cyp19a1 and Hsd17b1 levels support the possibility that estrogen biosynthesis from androgens are accumulated during inflammatory liver diseases. Furthermore, the increased levels of Cyp1a1 and Cyp1b1 in the hydroxylation of estrogen facilitated the conversion of estrogen to 2- or 4-hydroxyestrogen, respectively. In addition, the substantial increase in the Sult1e1 enzyme levels could lead to sulfate conjugation of hydroxyestrogen. The present information supports the concept that inflammatory response can sequester sulfate conjugates from the endogenous steroid hormones and may suppress binding of sex steroid hormones to their receptors in the whole body.
Collapse
Affiliation(s)
- Sang R Lee
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Seung-Yeon Lee
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Sang-Yun Kim
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Si-Yun Ryu
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Bae-Kuen Park
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Eui-Ju Hong
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
38
|
Eriksson AL, Wilhelmson AS, Fagman JB, Ryberg H, Koskela A, Tuukkanen J, Tivesten Å, Ohlsson C. The Bone Sparing Effects of 2-Methoxyestradiol Are Mediated via Estrogen Receptor-α in Male Mice. Endocrinology 2016; 157:4200-4205. [PMID: 27631553 PMCID: PMC5086527 DOI: 10.1210/en.2016-1402] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
2-Methoxyestradiol (2ME2), a metabolite of 17β-estradiol (E2), exerts bone sparing effects in animal models. We hypothesized that the underlying mechanism is back conversion of 2ME2 to E2, which subsequently acts via estrogen receptor (ER)α. We measured serum E2 levels in orchidectomized wild-type (WT) mice treated with 2ME2 66.6 μg/d or placebo. In placebo-treated animals, E2 was below the detection limit. In 2ME2-treated mice, the serum E2 level was 4.97 ± 0.68 pg/mL. This corresponds to the level found in diesterus in cycling female mice. Next, we investigated bone parameters in orchidectomized WT and ERα knockout mice treated with 2ME2 or placebo for 35 days. 2ME2 (6.66 μg/d) preserved trabecular and cortical bone in WT mice. Trabecular volumetric-bone mineral density was 64 ± 20%, and trabecular bone volume/total volume was 60 ± 20% higher in the metaphyseal region of the femur in the 2ME2 group, compared with placebo (P < .01). Both trabecular number and trabecular thickness were increased (P < .01). Cortical bone mineral content in the diaphyseal region of the femur was 31 ± 3% higher in the 2ME2 group, compared with placebo (P < .001). This was due to larger cortical area (P < .001). Three-point bending showed an increased bone strength in WT 2ME2-treated animals compared with placebo (maximum load [Fmax] +19±5% in the 2ME2 group, P < .05). Importantly, no bone parameter was affected by 2ME2 treatment in ERα knockout mice. In conclusion, 2ME2 treatment of orchidectomized mice results in increased serum E2. ERα mediates the bone sparing effects of 2ME2. The likely mediator of this effect is E2 resulting from back conversion of 2ME2.
Collapse
Affiliation(s)
- Anna L Eriksson
- Center for Bone and Arthritis Research (A.L.E., H.R., C.O.), Department of Internal Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden; Wallenberg Laboratory for Cardiovascular and Metabolic Research (A.S.W., A.T.), Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden; Sahlgrenska Cancer Center (J.F.), Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden; Clinical Chemistry (H.R.), Sahlgrenska University Hospital, SE-413 45 Gothenburg, Sweden; and Unit of Cancer Research and Translational Medicine (A.K., J.T.), Medical Research Center, Oulu and Department of Anatomy and Cell Biology, University of Oulu, FI-900 14 Oulu, Finland
| | - Anna S Wilhelmson
- Center for Bone and Arthritis Research (A.L.E., H.R., C.O.), Department of Internal Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden; Wallenberg Laboratory for Cardiovascular and Metabolic Research (A.S.W., A.T.), Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden; Sahlgrenska Cancer Center (J.F.), Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden; Clinical Chemistry (H.R.), Sahlgrenska University Hospital, SE-413 45 Gothenburg, Sweden; and Unit of Cancer Research and Translational Medicine (A.K., J.T.), Medical Research Center, Oulu and Department of Anatomy and Cell Biology, University of Oulu, FI-900 14 Oulu, Finland
| | - Johan B Fagman
- Center for Bone and Arthritis Research (A.L.E., H.R., C.O.), Department of Internal Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden; Wallenberg Laboratory for Cardiovascular and Metabolic Research (A.S.W., A.T.), Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden; Sahlgrenska Cancer Center (J.F.), Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden; Clinical Chemistry (H.R.), Sahlgrenska University Hospital, SE-413 45 Gothenburg, Sweden; and Unit of Cancer Research and Translational Medicine (A.K., J.T.), Medical Research Center, Oulu and Department of Anatomy and Cell Biology, University of Oulu, FI-900 14 Oulu, Finland
| | - Henrik Ryberg
- Center for Bone and Arthritis Research (A.L.E., H.R., C.O.), Department of Internal Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden; Wallenberg Laboratory for Cardiovascular and Metabolic Research (A.S.W., A.T.), Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden; Sahlgrenska Cancer Center (J.F.), Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden; Clinical Chemistry (H.R.), Sahlgrenska University Hospital, SE-413 45 Gothenburg, Sweden; and Unit of Cancer Research and Translational Medicine (A.K., J.T.), Medical Research Center, Oulu and Department of Anatomy and Cell Biology, University of Oulu, FI-900 14 Oulu, Finland
| | - Antti Koskela
- Center for Bone and Arthritis Research (A.L.E., H.R., C.O.), Department of Internal Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden; Wallenberg Laboratory for Cardiovascular and Metabolic Research (A.S.W., A.T.), Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden; Sahlgrenska Cancer Center (J.F.), Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden; Clinical Chemistry (H.R.), Sahlgrenska University Hospital, SE-413 45 Gothenburg, Sweden; and Unit of Cancer Research and Translational Medicine (A.K., J.T.), Medical Research Center, Oulu and Department of Anatomy and Cell Biology, University of Oulu, FI-900 14 Oulu, Finland
| | - Juha Tuukkanen
- Center for Bone and Arthritis Research (A.L.E., H.R., C.O.), Department of Internal Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden; Wallenberg Laboratory for Cardiovascular and Metabolic Research (A.S.W., A.T.), Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden; Sahlgrenska Cancer Center (J.F.), Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden; Clinical Chemistry (H.R.), Sahlgrenska University Hospital, SE-413 45 Gothenburg, Sweden; and Unit of Cancer Research and Translational Medicine (A.K., J.T.), Medical Research Center, Oulu and Department of Anatomy and Cell Biology, University of Oulu, FI-900 14 Oulu, Finland
| | - Åsa Tivesten
- Center for Bone and Arthritis Research (A.L.E., H.R., C.O.), Department of Internal Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden; Wallenberg Laboratory for Cardiovascular and Metabolic Research (A.S.W., A.T.), Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden; Sahlgrenska Cancer Center (J.F.), Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden; Clinical Chemistry (H.R.), Sahlgrenska University Hospital, SE-413 45 Gothenburg, Sweden; and Unit of Cancer Research and Translational Medicine (A.K., J.T.), Medical Research Center, Oulu and Department of Anatomy and Cell Biology, University of Oulu, FI-900 14 Oulu, Finland
| | - Claes Ohlsson
- Center for Bone and Arthritis Research (A.L.E., H.R., C.O.), Department of Internal Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden; Wallenberg Laboratory for Cardiovascular and Metabolic Research (A.S.W., A.T.), Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden; Sahlgrenska Cancer Center (J.F.), Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden; Clinical Chemistry (H.R.), Sahlgrenska University Hospital, SE-413 45 Gothenburg, Sweden; and Unit of Cancer Research and Translational Medicine (A.K., J.T.), Medical Research Center, Oulu and Department of Anatomy and Cell Biology, University of Oulu, FI-900 14 Oulu, Finland
| |
Collapse
|
39
|
Tripathi K, Mani C, Somasagara RR, Clark DW, Ananthapur V, Vinaya K, Palle K. Detection and evaluation of estrogen DNA-adducts and their carcinogenic effects in cultured human cells using biotinylated estradiol. Mol Carcinog 2016; 56:1010-1020. [DOI: 10.1002/mc.22566] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Revised: 08/31/2016] [Accepted: 09/04/2016] [Indexed: 12/19/2022]
Affiliation(s)
- Kaushlendra Tripathi
- Department of Oncologic Sciences, Mitchell Cancer Institute; University of South Alabama; Mobile Alabama
| | - Chinnadurai Mani
- Department of Oncologic Sciences, Mitchell Cancer Institute; University of South Alabama; Mobile Alabama
| | - Ranganatha R. Somasagara
- Department of Oncologic Sciences, Mitchell Cancer Institute; University of South Alabama; Mobile Alabama
| | - David W. Clark
- Department of Oncologic Sciences, Mitchell Cancer Institute; University of South Alabama; Mobile Alabama
| | - Venkateshwari Ananthapur
- Department of Oncologic Sciences, Mitchell Cancer Institute; University of South Alabama; Mobile Alabama
- Institute of Genetics and Hospital for Genetic Diseases; Osmania University; Begumpet, Hyderabad Telangana India
| | - Kambappa Vinaya
- Department of Oncologic Sciences, Mitchell Cancer Institute; University of South Alabama; Mobile Alabama
- Department of Chemistry; Government First Grade College; Kadur Karnataka India
| | - Komaraiah Palle
- Department of Oncologic Sciences, Mitchell Cancer Institute; University of South Alabama; Mobile Alabama
| |
Collapse
|
40
|
Banu SK, Stanley JA, Sivakumar KK, Arosh JA, Burghardt RC. Resveratrol protects the ovary against chromium-toxicity by enhancing endogenous antioxidant enzymes and inhibiting metabolic clearance of estradiol. Toxicol Appl Pharmacol 2016; 303:65-78. [PMID: 27129868 PMCID: PMC5830085 DOI: 10.1016/j.taap.2016.04.016] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 03/29/2016] [Accepted: 04/26/2016] [Indexed: 12/31/2022]
Abstract
Resveratrol (RVT), a polyphenolic component in grapes and red wine, has been known for its cytoprotective actions against several diseases. However, beneficial effects of RVT against early exposure to endocrine disrupting chemicals (EDCs) have not been understood. EDCs are linked to several ovarian diseases such as premature ovarian failure, polycystic ovary syndrome, early menopause and infertility in women. Hexavalent chromium (CrVI) is a heavy metal EDC, and widely used in >50 industries. Environmental contamination with CrVI in the US is rapidly increasing, predisposing the human to several illnesses including cancers and still birth. Our lab has been involved in determining the molecular mechanism of CrVI-induced female infertility and intervention strategies to mitigate CrVI effects. Lactating mother rats were exposed to CrVI (50ppm potassium dichromate) from postpartum days 1-21 through drinking water with or without RVT (10mg/kg body wt., through oral gavage daily). During this time, F1 females received respective treatments through mother's milk. On postnatal day (PND) 25, blood and the ovary, kidney and liver were collected from the F1 females for analyses. CrVI increased atresia of follicles by increasing cytochrome C and cleaved caspase-3; decreasing antiapoptotic proteins; decreasing estradiol (E2) biosynthesis and enhancing metabolic clearance of E2, increasing oxidative stress and decreasing endogenous antioxidants. RVT mitigated the effects of CrVI by upregulating cell survival proteins and AOXs; and restored E2 levels by inhibiting hydroxylation, glucuronidation and sulphation of E2. This is the first study to report the protective effects of RVT against any toxicant in the ovary.
Collapse
Affiliation(s)
- Sakhila K Banu
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA.
| | - Jone A Stanley
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Kirthiram K Sivakumar
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Joe A Arosh
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Robert C Burghardt
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
41
|
Piccinato CA, Neme RM, Torres N, Sanches LR, Cruz Derogis PBM, Brudniewski HF, E Silva JCR, Ferriani RA. Increased expression of CYP1A1 and CYP1B1 in ovarian/peritoneal endometriotic lesions. Reproduction 2016; 151:683-92. [PMID: 27012269 DOI: 10.1530/rep-15-0581] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 03/24/2016] [Indexed: 12/22/2022]
Abstract
Endometriosis is an estrogen-dependent disease affecting up to 10% of all premenopausal women. There is evidence that different endometriosis sites show distinct local estrogen concentration, which, in turn, might be due to a unique local estrogen metabolism. We aimed to investigate whether there was a site-specific regulation of selected enzymes responsible for the oxidative metabolism of estrogens in biopsy samples and endometrial and endometriotic stromal cells. Cytochrome P450 (CYP) 1A1 and CYP1B1 mRNA and protein expressions in deep-infiltrating (rectal, retossigmoidal, and uterossacral) lesions, superficial (ovarian and peritoneal) lesions, and eutopic and healthy (control) endometrium were evaluated by real-time PCR and western blot. Using a cross-sectional study design with 58 premenopausal women who were not under hormonal treatment, we were able to identify an overall increased CYP1A1 and CYP1B1 mRNA expression in superficial lesions compared with the healthy endometrium. CYP1A1 mRNA expression in superficial lesions was also greater than in the eutopic endometrium. Interestingly, we found a similar pattern of CYP1A1 and CYP1B1 expression in in vitro stromal cells isolated from ovarian lesions (n=3) when compared with stromal cells isolated from either rectum lesions or eutopic endometrium. In contradiction, there was an increased half-life of estradiol (measured by HPLC-MS-MS) in ovarian endometriotic stromal cells compared with paired eutopic stromal endometrial cells. Our results indicate that there is a site-dependent regulation of CYP1A1 and CYP1B1 in ovarian/peritoneal lesions and ovarian endometriotic stromal cells, whereas a slower metabolism is taking place in these cells.
Collapse
Affiliation(s)
- Carla A Piccinato
- Hospital Israelita Albert EinsteinSão Paulo, Brazil Department of Gynaecology and ObstetricsSchool of Medicine of Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Rosa M Neme
- Hospital Israelita Albert EinsteinSão Paulo, Brazil Centro de Endometriose São PauloAv. República do Líbano, São Paulo, Brazil
| | | | - Lívia Renta Sanches
- Department of Clinical PathologyHospital Israelita Albert Einstein, São Paulo, Brazil
| | | | - Heloísa F Brudniewski
- Hospital Israelita Albert EinsteinSão Paulo, Brazil Centro de Endometriose São PauloAv. República do Líbano, São Paulo, Brazil
| | - Júlio C Rosa E Silva
- Department of Gynaecology and ObstetricsSchool of Medicine of Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Rui A Ferriani
- Department of Gynaecology and ObstetricsSchool of Medicine of Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
42
|
Michalska MM, Samulak D, Jabłoński F, Romanowicz H, Smolarz B. The R156R ERCC2 polymorphism as a risk factor of endometrial cancer. Tumour Biol 2015; 37:2171-6. [PMID: 26349749 DOI: 10.1007/s13277-015-4040-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 09/02/2015] [Indexed: 01/05/2023] Open
Abstract
Endometrial carcinoma (EC) is the most frequent malignant neoplasm of female genitals and the fourth most frequent malignant neoplasm in Polish women, after breast, colorectal and lung cancer. Despite intensive research, EC aetiology remains unknown. The variability, perceived in DNA repair genes, may be of clinical importance for evaluation of the risk of occurrence of a given type of cancer, its prophylactics and therapy. The aim of the study was to determine the relationship between gene polymorphism R156R (C to A, rs238406) of ERCC2 gene and modulation of the risk of endometrial cancer in Poland. Our research included 1360 patients with EC and 1320 healthy controls. The genotype analysis of ERCC2 gene polymorphism was performed using the PCR-based restriction fragment length polymorphism (PCR-RFLP). In the presented study, a relationship was identified between R156R polymorphism of the ERCC2 gene and the incidence of endometrial cancer. An association was observed between EC occurrence and the presence of A/A genotype (odds ratio (OR) 9.71, 95 % Cl 7.53-12.50, p < .0001). A tendency for an increased risk of endometrial cancer was detected with the occurrence of A allele of ERCC2 polymorphism (OR = 5.95, 95 % Cl 5.23-6.78, p < .0001). A relationship was confirmed between R156R polymorphism and endometrial cancer progression, assessed by histological grades. On the basis of these results, we conclude that ERCC2 gene polymorphism R156R may be associated with an increased risk of endometrial cancer.
Collapse
Affiliation(s)
- Magdalena M Michalska
- Department of Obstetrics and Gynaecology, Regional Hospital in Kalisz, Kalisz, Poland
| | - Dariusz Samulak
- Department of Obstetrics and Gynaecology, Regional Hospital in Kalisz, Kalisz, Poland.,Cathedral of Mother's and Child's Health, Poznań University of Medical Sciences, Poznań, Poland
| | - Filip Jabłoński
- Laboratory of Cancer Genetics, Department of Pathology, Institute of Polish Mother's Memorial Hospital, Rzgowska 281/289, 93-338, Lodz, Poland
| | - Hanna Romanowicz
- Laboratory of Cancer Genetics, Department of Pathology, Institute of Polish Mother's Memorial Hospital, Rzgowska 281/289, 93-338, Lodz, Poland
| | - Beata Smolarz
- Laboratory of Cancer Genetics, Department of Pathology, Institute of Polish Mother's Memorial Hospital, Rzgowska 281/289, 93-338, Lodz, Poland.
| |
Collapse
|
43
|
Michalska MM, Samulak D, Romanowicz H, Bieńkiewicz J, Sobkowski M, Ciesielski K, Smolarz B. Single nucleotide polymorphisms (SNPs) of hOGG1 and XRCC1 DNA repair genes and the risk of ovarian cancer in Polish women. Tumour Biol 2015; 36:9457-63. [PMID: 26124010 DOI: 10.1007/s13277-015-3707-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 06/22/2015] [Indexed: 11/26/2022] Open
Abstract
The aim of this study was to determine single nucleotide polymorphisms in hOGG1 (Ser326Cys (rs13181)) and XRCC1 (Arg194Trp (rs1799782)) genes, respectively, and to identify the correlation between them and the overall risk, grading and staging of ovarian cancer in Polish women. Our study comprised 720 patients diagnosed with ovarian cancer and 720 healthy controls. The genotype analysis of hOGG1 and XRCC1 polymorphisms was performed using polymerase chain reaction (PCR)-based restriction fragment length polymorphism (PCR-RFLP). Odds ratios (OR) and 95 % confidence intervals (CI) for each genotype and allele were calculated. Results revealed an association between hOGG1 Ser326Cys polymorphism and the incidence of ovarian cancer. Variant Cys allele of hOGG1 increased the overall cancer risk (OR 2.89; 95 % CI 2.47-3.38; p < .0001). Moreover, ovarian cancer grading remained in a relationship with both analysed polymorphisms; G1 tumours presented increased frequencies of hOGG1 Cys/Cys homozygotes (OR 18.33; 95 % CI 9.38-35.81; p < .0001) and XRCC1 Trp/Trp homozygotes (OR 20.50; 95 % CI 10.17-41.32; p < .0001). Furthermore, G1 ovarian cancers displayed an overrepresentation of Cys and Trp allele. In conclusion, hOGG1 Ser326Cys and XRCC1 Arg194Trp polymorphisms may be regarded as risk factors of ovarian cancer.
Collapse
Affiliation(s)
- Magdalena M Michalska
- Department of Obstetrics and Gynaecology, Regional Hospital in Kalisz, Kalisz, Poland
| | - Dariusz Samulak
- Department of Obstetrics and Gynaecology, Regional Hospital in Kalisz, Kalisz, Poland
- Cathedral of Mother's and Child's Health, Poznan University of Medical Sciences, Poznań, Poland
| | - Hanna Romanowicz
- Laboratory of Cancer Genetics, Department of Pathology, Institute of Polish Mother's Memorial Hospital, Rzgowska 281/289, 93-338, Lodz, Poland
| | - Jan Bieńkiewicz
- Department of Surgical, Endoscopic and Oncologic Gynaecology, Institute of Polish Mother's Memorial Hospital, Lodz, Poland
| | - Maciej Sobkowski
- Department of Obstetrics and Gynaecology, University Hospital, Polna 33, Poznań, Poland
| | | | - Beata Smolarz
- Laboratory of Cancer Genetics, Department of Pathology, Institute of Polish Mother's Memorial Hospital, Rzgowska 281/289, 93-338, Lodz, Poland.
| |
Collapse
|
44
|
Cigarettes, genetic background, and menopausal timing: the presence of single nucleotide polymorphisms in cytochrome P450 genes is associated with increased risk of natural menopause in European-American smokers. Menopause 2015; 21:694-701. [PMID: 24448104 DOI: 10.1097/gme.0000000000000140] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE This study aims to evaluate associations between variations in genes involved in the metabolism of environmental chemicals and steroid hormones and risk of menopause in smokers. METHODS Survival analysis was performed on 410 eligible participants from the Penn Ovarian Aging study (ongoing for 14 years), a cohort study of late-reproductive-age women. Single nucleotide polymorphisms at the following loci were studied: COMT Val158Met, CYP1B1*4 Asn452Ser, CYP1B1*3 Leu432Val, and CYP3A4*1B. RESULTS Significant interactions between smoking and single nucleotide polymorphisms were observed in European-American carriers of CYP3A4*1B and CYP1B1*3, supporting a greater risk of menopause entry compared with those not carrying these alleles. Among CYP1B1*3 carriers, smokers had a greater risk of menopause entry than nonsmokers (adjusted hazard ratio [HR], 2.26; 95% CI, 1.4-3.67; median time to menopause, 10.42 and 11.07 y, respectively). No association between smoking and menopause was identified in CYP1B1 wild types. Among CYP3A4*1B carriers, smokers were at greater risk for menopause entry than nonsmokers (adjusted HR, 15.1; 95% CI, 3.31-69.2; median time to menopause, 11.36 and 13.91 y, respectively). Risk of menopause entry in CYP3A4 wild types who smoked was far lower (adjusted HR, 1.59; 95% CI, 1.03-2.44). Heavily smoking CYP1B1*3 carriers (adjusted HR, 3.0; 95% CI, 1.54-5.84; median time to menopause, 10.41 y) and heavily smoking CYP3A4*1B carriers (adjusted HR, 17.79; 95% CI, 3.21-98.65; median time to menopause, 5.09 y) had the greatest risk of menopause entry. CONCLUSIONS Our finding that the risk of menopause entry in European-American smokers varies depending on genetic background represents a novel gene-environment interaction in reproductive aging.
Collapse
|
45
|
Chourasia TK, Pang Y, Thomas P. The catecholestrogen, 2-hydroxyestradiol-17beta, acts as a G protein-coupled estrogen receptor 1 (GPER/GPR30) antagonist to promote the resumption of meiosis in zebrafish oocytes. Biol Reprod 2015; 92:69. [PMID: 25609836 DOI: 10.1095/biolreprod.114.125674] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Estradiol-17beta (E2) maintains high cAMP levels and meiotic arrest in zebrafish oocytes through activation of G protein-coupled estrogen receptor (GPER). The catecholestrogen 2-hydroxyestradiol-17beta (2-OHE2) has an opposite effect to that of E2 on oocyte maturation (OM) and cAMP levels in Indian catfish oocytes. We tested the hypothesis that 2-OHE2 is produced in zebrafish ovaries and promotes the resumption of oocyte meiosis through its action as a GPER antagonist. Ovarian 2-OHE2 production by estrogen-2-hydroxylase (EH) was up-regulated by gonadotropin treatment at the onset of OM, consistent with a physiological role for 2-OHE2 in regulating OM. The increases in EH activity and OM were blocked by treatment with CYP1A1 and CYP1B1 inhibitors. Expression of cyp1a, cyp1b1, and cyp1c mRNAs was increased by gonadotropin treatment, further implicating these Cyp1s in 2-OHE2 synthesis prior to OM. Conversely, aromatase activity and cyp19a1 mRNA expression declined during gonadotropin induction of OM. 2-OHE2 treatment significantly increased spontaneous OM in defolliculated zebrafish oocytes and reversed the inhibition of OM by E2 and the GPER agonist G-1. 2-OHE2 was an effective competitor of [(3)H]-E2 binding to recombinant zebrafish GPER expressed in HEK-293 cells. 2-OHE2 also antagonized estrogen actions through GPER on cAMP production in zebrafish oocytes, resulting in a reduction in cAMP levels. Stimulation of OM by 2-OHE2 was blocked by pretreatment of defolliculated oocytes with the GPER antibody. Collectively, the results suggest that 2-OHE2 functions as a GPER antagonist and promotes OM in zebrafish through blocking GPER-dependent E2 inhibition of the resumption of OM.
Collapse
Affiliation(s)
- Tapan K Chourasia
- University of Texas at Austin, Marine Science Institute, Port Aransas, Texas
| | - Yefei Pang
- University of Texas at Austin, Marine Science Institute, Port Aransas, Texas
| | - Peter Thomas
- University of Texas at Austin, Marine Science Institute, Port Aransas, Texas
| |
Collapse
|
46
|
Zhang J, Huang Y, Wang X, Lin K, Wu K. Environmental Polychlorinated Biphenyl Exposure and Breast Cancer Risk: A Meta-Analysis of Observational Studies. PLoS One 2015; 10:e0142513. [PMID: 26555153 PMCID: PMC4640539 DOI: 10.1371/journal.pone.0142513] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 10/22/2015] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Association between polychlorinated biphenyl (PCB) exposure and breast cancer risk has been widely studied, but the results remain controversial. We performed a meta-analysis to evaluate the evidences from observational studies on PCB exposure and breast cancer risk. METHODS Relevant studies with data on internal PCB dose were identified from PubMed, EMBASE, CBM and CNKI databases through November 2014. Multivariable-adjusted odds ratio (OR) with 95% confidence intervals (CIs) were applied to assess the association between PCB exposure and breast cancer risk. Heterogeneity test, sensitivity analysis, subgroup analysis and publication bias test were also performed. To further explore the association between specific groups of PCB congeners and breast cancer, we examined the PCB congeners classified, according to their structural, biological and pharmacokinetics properties, as group I (potentially estrogenic), group II (potentially anti-estrogenic and immunotoxic, dioxin-like), and group III (phenobarbital, CYP1A and CYP2B inducers, biologically persistent). RESULTS Of 660 studies screened, 25 studies which met criteria were selected, involving a total of 12866 participants (6088 cases and 6778 controls) from eight countries. The results showed that the risk of breast cancer was associated with group II (OR = 1.23, 95% CI: 1.08-1.40) and group III (OR = 1.25, 95% CI: 1.09-1.43) PCBs, but not with group I (OR = 1.10, 95%CI: 0.97-1.24) PCBs or total PCB exposure (OR = 1.09, 95%CI: 0.97-1.22). CONCLUSIONS Our meta-analysis based on the selected studies found group II and group III PCB exposure might contribute to the risk of breast cancer. More studies in developing countries with higher PCB levels are needed, as well as studies to explore the relationships between mixtures of organochlorine compounds and breast cancer risk.
Collapse
Affiliation(s)
- Jingwen Zhang
- Department of Preventive Medicine, Shantou University Medical College, Shantou, Guangdong, China
| | - Yue Huang
- Department of Preventive Medicine, Shantou University Medical College, Shantou, Guangdong, China
| | - Xiaoling Wang
- Department of Preventive Medicine, Shantou University Medical College, Shantou, Guangdong, China
| | - Kun Lin
- Department of Preventive Medicine, Shantou University Medical College, Shantou, Guangdong, China
| | - Kusheng Wu
- Department of Preventive Medicine, Shantou University Medical College, Shantou, Guangdong, China
- * E-mail:
| |
Collapse
|
47
|
Stack DE, Ritonya J, Jakopovic S, Maloley-Lewis B. Regioselective deuterium labeling of estrone and catechol estrogen metabolites. Steroids 2014; 92:32-8. [PMID: 25174785 DOI: 10.1016/j.steroids.2014.08.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 08/07/2014] [Accepted: 08/13/2014] [Indexed: 12/21/2022]
Abstract
Increased exposure to estrogens and estrogen metabolites is linked with increased rates of breast, ovarian and other human cancers. Metabolism of estrogen can led to formation of electrophilic o-quinones capable of binding to DNA. In order to gain insight into the mechanism of estrogen-induced DNA damage, estrone and catechol estrogens derived from estrone, have been regioselectively labeled with deuterium at the 1-position. Estrone-1-d, estrone-1,2,4-d3, 4-hydroxyestrone-1-d and 2-hydroxyestrone-1-d have been synthesized with or without deuteriums at the 16-position. The key labeling step involves deuterated trifluoroacetic acid exchange catalyzed by t-butyl alcohol. This economical, straightforward labeling technique makes available a range of estrone compounds containing deuterium at the 1-position.
Collapse
Affiliation(s)
- Douglas E Stack
- Department of Chemistry, University of Nebraska at Omaha, 6001 Dodge Street, Omaha, NE 68182-0109, United States.
| | - Justin Ritonya
- Department of Chemistry, University of Nebraska at Omaha, 6001 Dodge Street, Omaha, NE 68182-0109, United States
| | - Scott Jakopovic
- Department of Chemistry, University of Nebraska at Omaha, 6001 Dodge Street, Omaha, NE 68182-0109, United States
| | - Brittney Maloley-Lewis
- Department of Chemistry, University of Nebraska at Omaha, 6001 Dodge Street, Omaha, NE 68182-0109, United States
| |
Collapse
|
48
|
L'Héritier F, Marques M, Fauteux M, Gaudreau L. Defining molecular sensors to assess long-term effects of pesticides on carcinogenesis. Int J Mol Sci 2014; 15:17148-61. [PMID: 25257533 PMCID: PMC4200861 DOI: 10.3390/ijms150917148] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 09/11/2014] [Accepted: 09/22/2014] [Indexed: 12/04/2022] Open
Abstract
The abundance of dioxins and dioxin-like pollutants has massively increased in the environment due to human activity. These chemicals are particularly persistent and accumulate in the food chain, which raises major concerns regarding long-term exposure to human health. Most dioxin-like pollutants activate the aryl hydrocarbon receptor (AhR) transcription factor, which regulates xenobiotic metabolism enzymes that belong to the cytochrome P450 1A family (that includes CYP1A1 and CYP1B1). Importantly, a crosstalk exists between estrogen receptor α (ERα) and AhR. More specifically, ERα represses the expression of the CYP1A1 gene, which encodes an enzyme that converts 17β-estradiol into 2-hydroxyestradiol. However, (ERα) does not repress the CYP1B1 gene, which encodes an enzyme that converts 17β-estradiol into 4-hydroxyestradiol, one of the most genotoxic estrogen metabolites. In this review, we discuss how chronic exposure to xenobiotic chemicals, such as pesticides, might affect the expression of genes regulated by the AhR–ERα crosstalk. Here, we focus on recent advances in the understanding of molecular mechanisms that mediate this crosstalk repression, and particularly on how ERα represses the AhR target gene CYP1A1, and could subsequently promote breast cancer. Finally, we propose that genes implicated in this crosstalk could constitute important biomarkers to assess long-term effects of pesticides on human health.
Collapse
Affiliation(s)
- Fanny L'Héritier
- Département de Biologie, Université de Sherbrooke, 2500 boul. de l'Université, Sherbrooke, QC J1K 2R1, Canada.
| | - Maud Marques
- Département de Biologie, Université de Sherbrooke, 2500 boul. de l'Université, Sherbrooke, QC J1K 2R1, Canada.
| | - Myriam Fauteux
- Département de Biologie, Université de Sherbrooke, 2500 boul. de l'Université, Sherbrooke, QC J1K 2R1, Canada.
| | - Luc Gaudreau
- Département de Biologie, Université de Sherbrooke, 2500 boul. de l'Université, Sherbrooke, QC J1K 2R1, Canada.
| |
Collapse
|
49
|
Jung SY, Vitolins MZ, Paskett ED, Chang S. Exogenous estrogen as mediator of racial differences in bioactive insulin-like growth factor-I levels among postmenopausal women. J Gerontol A Biol Sci Med Sci 2014; 70:495-502. [PMID: 25238773 DOI: 10.1093/gerona/glu164] [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: 11/14/2022] Open
Abstract
BACKGROUND The role of exogenous estrogen use in racial differences in insulin-like growth factor-I (IGF-I) levels which affect cancer risk is unclear. We investigated whether the relationship between race and circulating bioactive IGF-I proteins was mediated by exogenous estrogen and the extent to which exogenous estrogen influenced the race-IGF-I relationship in postmenopausal women. METHODS This cross-sectional study included 636 white and 133 African American postmenopausal women enrolled in an ancillary study of the Women's Health Initiative Observational Study. To assess exogenous estrogen use (nonusers [n = 262] vs users [n = 507]) as a mediator of the race-IGF-I relationship, we used the Baron-Kenny method and an estimation of the proportional change in the odd ratios for IGF-I levels on race plus a bootstrapping test for the significance of the mediation effect. RESULTS Compared with white women, African American women were more likely to have high IGF-I levels and less likely to use exogenous estrogen. After accounting for race, estrogen nonusers had higher IGF-I levels than estrogen users did. Among oral contraceptive ever users, exogenous estrogen had a strong mediation effect (67%; p = .018) in the race-IGF-I relationship. In the women with a history of hypertension, exogenous estrogen explained racial differences in IGF-I levels to a modest degree (23%; p = .029). CONCLUSIONS Exogenous estrogen use has a potentially important role in disparities in IGF-I bioactivity between postmenopausal African American and white women. A history of oral contraceptive use and hypertension may be part of the interconnected hormonal pathways related to racial differences in IGF-I levels.
Collapse
Affiliation(s)
- Su Yon Jung
- Department of Epidemiology, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston.
| | - Mara Z Vitolins
- Department of Epidemiology and Prevention, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Electra D Paskett
- Division of Cancer Prevention and Control, College of Medicine, Ohio State University, Columbus
| | - Shine Chang
- Department of Epidemiology, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston
| |
Collapse
|
50
|
Quesnot N, Bucher S, Fromenty B, Robin MA. Modulation of metabolizing enzymes by bisphenol a in human and animal models. Chem Res Toxicol 2014; 27:1463-73. [PMID: 25142872 DOI: 10.1021/tx500087p] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Xenobiotics, such as contaminants and drugs, can be converted to potentially toxic reactive metabolites by phase 1 oxidizing enzymes. These metabolites are further detoxified by phase 2 conjugating enzymes and eliminated from cells by phase 3 transporters. Moreover, many of these xenobiotics are also able to induce or inhibit these enzymes, potentially modulating their own toxicity or that of other chemicals. The present review is focused on bisphenol A, a synthetic monomer used for many industrial applications and exhibiting xenoestrogen properties. The impact of this contaminant on all major classes of metabolizing enzymes (i.e., cytochromes P450, glutathione-S-transferases, sulfotransferases, UDP-glucuronyltransferases, and transporters) was reviewed, with a highlight on the modulation of cytochromes P450 involved in steroid metabolism. Interestingly, most of the studies reported in this review show that BPA is able to induce or inhibit metabolizing enzymes at high doses but also at doses compatible with human exposure.
Collapse
|