1
|
Michel R, Hazimeh D, Saad EE, Olson SL, Musselman K, Elgindy E, Borahay MA. Common Beverage Consumption and Benign Gynecological Conditions. BEVERAGES (BASEL, SWITZERLAND) 2024; 10:33. [PMID: 38948304 PMCID: PMC11211953 DOI: 10.3390/beverages10020033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
The purpose of this article is to review the effects of four commonly consumed beverage types-sugar-sweetened beverages (SSBs), caffeinated beverages, green tea, and alcohol-on five common benign gynecological conditions: uterine fibroids, endometriosis, polycystic ovary syndrome (PCOS), anovulatory infertility, and primary dysmenorrhea (PD). Here we outline a plethora of research, highlighting studies that demonstrate possible associations between beverage intake and increased risk of certain gynecological conditions-such as SSBs and dysmenorrhea-as well as studies that demonstrate a possible protective effect of beverage against risk of gynecological condition-such as green tea and uterine fibroids. This review aims to help inform the diet choices of those with the aforementioned conditions and give those with uteruses autonomy over their lifestyle decisions.
Collapse
Affiliation(s)
- Rachel Michel
- Department of Population, Family, and Reproductive Health, Bloomberg School of Public Health, Baltimore, MD 21205 USA
| | - Dana Hazimeh
- Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, MD 21205 USA
| | - Eslam E. Saad
- Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, MD 21205 USA
| | - Sydney L. Olson
- Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, MD 21205 USA
| | - Kelsey Musselman
- Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, MD 21205 USA
| | - Eman Elgindy
- Department of Gynecology and Obstetrics, Zagazig University School of Medicine, Zagazig, 44519, Egypt
| | - Mostafa A. Borahay
- Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, MD 21205 USA
| |
Collapse
|
2
|
Davis EHS, Jones C, Coward K. Rethinking the application of nanoparticles in women's reproductive health and assisted reproduction. Nanomedicine (Lond) 2024; 19:1231-1251. [PMID: 38686941 PMCID: PMC11285225 DOI: 10.2217/nnm-2023-0346] [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: 11/29/2023] [Accepted: 03/25/2024] [Indexed: 05/02/2024] Open
Abstract
Nanoparticles and nanotechnology may present opportunities to revolutionize the prevention, treatment and diagnosis of a range of reproductive health conditions in women. These technologies are also used to improve outcomes of assisted reproductive technology. We highlight a range of these potential clinical uses of nanoparticles for polycystic ovary syndrome, endometriosis, uterine fibroids and sexually transmitted infections, considering in vitro and in vivo studies along with clinical trials. In addition, we discuss applications of nanoparticles in assisted reproductive technology, including sperm loading, gamete and embryo preservation and preventing preterm birth. Finally, we present some of the concerns associated with the medical use of nanoparticles, identifying routes for further exploration before nanoparticles can be applied to women's reproductive health in the clinic.
Collapse
Affiliation(s)
- Emily HS Davis
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Women’s Centre, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
| | - Celine Jones
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Women’s Centre, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
| | - Kevin Coward
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Women’s Centre, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
| |
Collapse
|
3
|
Enazy SA, Kirschen GW, Vincent K, Yang J, Saada J, Shah M, Oberhauser AF, Bujalowski PJ, Motamedi M, Salama SA, Kilic G, Rytting E, Borahay MA. PEGylated Polymeric Nanoparticles Loaded with 2-Methoxyestradiol for the Treatment of Uterine Leiomyoma in a Patient-Derived Xenograft Mouse Model. J Pharm Sci 2023; 112:2552-2560. [PMID: 37482124 PMCID: PMC10529399 DOI: 10.1016/j.xphs.2023.07.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 07/17/2023] [Accepted: 07/17/2023] [Indexed: 07/25/2023]
Abstract
Leiomyomas, the most common benign neoplasms of the female reproductive tract, currently have limited medical treatment options. Drugs targeting estrogen/progesterone signaling are used, but side effects and limited efficacy in many cases are major limitation of their clinical use. Previous studies from our laboratory and others demonstrated that 2-methoxyestradiol (2-ME) is promising treatment for uterine fibroids. However, its poor bioavailability and rapid degradation hinder its development for clinical use. The objective of this study is to evaluate the in vivo effect of biodegradable and biocompatible 2-ME-loaded polymeric nanoparticles in a patient-derived leiomyoma xenograft mouse model. PEGylated poly(lactide-co-glycolide) (PEG-PLGA) nanoparticles loaded with 2-ME were prepared by nanoprecipitation. Female 6-week age immunodeficient NOG (NOD/Shi-scid/IL-2Rγnull) mice were used. Estrogen-progesterone pellets were implanted subcutaneously. Five days later, patient-derived human fibroid tumors were xenografted bilaterally subcutaneously. Engrafted mice were treated with 2-ME-loaded or blank (control) PEGylated nanoparticles. Nanoparticles were injected intraperitoneally and after 28 days of treatment, tumor volume was measured by caliper following hair removal, and tumors were removed and weighed. Up to 99.1% encapsulation efficiency was achieved, and the in vitro release profile showed minimal burst release, thus confirming the high encapsulation efficiency. In vivo administration of the 2-ME-loaded nanoparticles led to 51% growth inhibition of xenografted tumors compared to controls (P < 0.01). Thus, 2-ME-loaded nanoparticles may represent a novel approach for the treatment of uterine fibroids.
Collapse
Affiliation(s)
- Sanaalarab Al Enazy
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, USA; Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gregory W Kirschen
- Department of Gynecology & Obstetrics, Johns Hopkins University, Baltimore, MD, USA
| | - Kathleen Vincent
- Center for Biomedical Engineering, University of Texas Medical Branch, Galveston, TX, USA; Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, USA
| | - Jinping Yang
- Center for Biomedical Engineering, University of Texas Medical Branch, Galveston, TX, USA
| | - Jamal Saada
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, USA
| | - Mansi Shah
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, USA
| | - Andres F Oberhauser
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Paul J Bujalowski
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Massoud Motamedi
- Center for Biomedical Engineering, University of Texas Medical Branch, Galveston, TX, USA
| | - Salama A Salama
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Gokhan Kilic
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, USA
| | - Erik Rytting
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, USA; Center for Biomedical Engineering, University of Texas Medical Branch, Galveston, TX, USA; Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, USA
| | - Mostafa A Borahay
- Department of Gynecology & Obstetrics, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
4
|
Hazimeh D, Massoud G, Parish M, Singh B, Segars J, Islam MS. Green Tea and Benign Gynecologic Disorders: A New Trick for An Old Beverage? Nutrients 2023; 15:1439. [PMID: 36986169 PMCID: PMC10054707 DOI: 10.3390/nu15061439] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/08/2023] [Accepted: 03/14/2023] [Indexed: 03/19/2023] Open
Abstract
Green tea is harvested from the tea plant Camellia sinensis and is one of the most widely consumed beverages worldwide. It is richer in antioxidants than other forms of tea and has a uniquely high content of polyphenolic compounds known as catechins. Epigallocatechin-3-gallate (EGCG), the major green tea catechin, has been studied for its potential therapeutic role in many disease contexts, including pathologies of the female reproductive system. As both a prooxidant and antioxidant, EGCG can modulate many cellular pathways important to disease pathogenesis and thus has clinical benefits. This review provides a synopsis of the current knowledge on the beneficial effects of green tea in benign gynecological disorders. Green tea alleviates symptom severity in uterine fibroids and improves endometriosis through anti-fibrotic, anti-angiogenic, and pro-apoptotic mechanisms. Additionally, it can reduce uterine contractility and improve the generalized hyperalgesia associated with dysmenorrhea and adenomyosis. Although its role in infertility is controversial, EGCG can be used as a symptomatic treatment for menopause, where it decreases weight gain and osteoporosis, as well as for polycystic ovary syndrome (PCOS).
Collapse
Affiliation(s)
| | | | | | | | - James Segars
- Department of Gynecology and Obstetrics, Division of Reproductive Sciences & Women’s Health Research, Johns Hopkins Medicine, Baltimore, MD 21205, USA
| | - Md Soriful Islam
- Department of Gynecology and Obstetrics, Division of Reproductive Sciences & Women’s Health Research, Johns Hopkins Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
5
|
Al-Qahtani SD, Bin-Melaih HH, Atiya EM, Fahmy UA, Binmahfouz LS, Neamatallah T, Al-Abbasi FA, Abdel-Naim AB. Self-Nanoemulsifying Drug Delivery System of 2-Methoxyestradiol Exhibits Enhanced Anti-Proliferative and Pro-Apoptotic Activities in MCF-7 Breast Cancer Cells. Life (Basel) 2022; 12:life12091369. [PMID: 36143405 PMCID: PMC9503162 DOI: 10.3390/life12091369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 08/25/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
(1) Background: 2-Methoxyestradiol (2ME) is a metabolite of estrogens and possesses promising anti-proliferative and cytotoxic activities. However, it suffers unfavorable pharmacokinetic characteristics such as absorption after oral administration. The aim of this study was to prepare an optimized 2ME self-nanoemulsifying drug delivery system (2ME-SNEDDS) and evaluate its cytotoxicity and pro-apoptotic activities in MCF-7 breast cancer cells. (2) Methods: For optimization of the 2ME-SNEDDS, a three-component system was used in the D-optimal mixture experimental study. MCF-7 cells were incubated with the 2ME-SNEDDS and subjected to an assessment of growth inhibition, cell cycle progression, annexin V staining, caspase-3 concentration, Bax, Bcl-2, and cyclin D1 mRNA expression, and reactive oxygen species (ROS) generation. (3) Results: The optimized formula had a globule size of 94.97 ± 4.35 nm. Zeta potential was found to be −3.4 ± 1.2 mV with a polydispersity index (PDI) of 0.34. In addition, 96.3 ± 4.3% of 2ME was released from the 2ME-SNEDDS within 24 h using the activated analysis bag technique. Moreover, the prepared 2ME-SNEDDS exhibited a significant enhancement of the anti-proliferative activity against MCF-7 cells in comparison to raw 2ME. This was associated with cyclin D1 expression down-regulation and the accumulation of cells in the G0/G1 and G2/M phases. The pro-apoptotic activities of the 2ME-SNEDDS were confirmed by annexin V staining, which indicated enhanced early and late cell death. This accompanied modulation of the mRNA expression of Bax and Bcl-2 in favor of apoptosis. The 2ME-SNEDDS significantly enhanced cleaved caspase-3 concentration in comparison to raw 2ME. In addition, the 2ME-SNEDDS significantly increased the generation of ROS in MCF-7 cells. (4) Conclusions: The 2ME-SNEDDS exhibits enhanced cytotoxicity and pro-apoptotic activity in MCF-7 cells. This is mediated by, at least partially, ROS generation.
Collapse
Affiliation(s)
- Salwa D. Al-Qahtani
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Science, Majmaah University, Majmaah 11952, Saudi Arabia
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Hawazen H. Bin-Melaih
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Eman M. Atiya
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Usama A. Fahmy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Lenah S. Binmahfouz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Thikryat Neamatallah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Fahad A. Al-Abbasi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ashraf B. Abdel-Naim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Correspondence:
| |
Collapse
|
6
|
Tunau KA, Garba JA, Panti AA, Shehu CE, Adamu AN, AbdulRahman MB, Ahmad MK. Low plasma vitamin D as a predictor of uterine fibroids in a nigerian population. Niger Postgrad Med J 2021; 28:181-186. [PMID: 34708704 DOI: 10.4103/npmj.npmj_495_21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Background Uterine fibroid is the most common benign tumour of the uterus and the exact cause is unknown. Vitamin D has been found to inhibit cell proliferation and enhance apoptosis in vitro. Aim The aim of this study is to compare the plasma level of Vitamin D among women with uterine fibroids and those without uterine fibroids. Materials and Methods This was an analytical cross-sectional study conducted among women with uterine fibroids and those without uterine fibroids. An interviewer-administered questionnaire was used to obtain relevant information. Five millilitres of venous blood was obtained for plasma Vitamin D assay. Data analysis was performed using the SPSS version 22. Level of significance was set at P < 0.05. Results There were 100 participants recruited for the study. The mean age of the participants with uterine fibroid was 35.06 ± 6.6 years and that of controls was 33.28 ± 7 years. The mean plasma level of Vitamin D for all the participants was 12.47 ± 7.53 ng/ml. The mean plasma level of Vitamin D in those with uterine fibroids was significantly lower than in those without uterine fibroid (10.16 ± 7.78 vs. 14.35 ± 6.8; t = 2.517, P = 0.014). It also shows that 1 unit increase in Vitamin D level will reduce the risk of developing uterine fibroids by 8% (odds ratio = 0.92, 95% confidence interval = 0.86-0.99). Conclusion The predictors of uterine fibroids were fewer hours spent outdoors, middle social class and low Vitamin D levels.
Collapse
Affiliation(s)
- Karima Abubakar Tunau
- Department of Obstetrics and Gynaecology, Usmanu Danfodiyo University/Usmanu Danfodiyo University Teaching Hospital, Sokoto, Sokoto State, Nigeria
| | - Jamila Abubakar Garba
- Department of Obstetrics and Gynaecology, Usmanu Danfodiyo University Teaching Hospital, Sokoto, Sokoto State,, Nigeria
| | - Abubakar Abubakar Panti
- Department of Obstetrics and Gynaecology, Usmanu Danfodiyo University/Usmanu Danfodiyo University Teaching Hospital, Sokoto, Sokoto State, Nigeria
| | - Constance Egondu Shehu
- Department of Obstetrics and Gynaecology, Usmanu Danfodiyo University/Usmanu Danfodiyo University Teaching Hospital, Sokoto, Sokoto State, Nigeria
| | - Aisha Nana Adamu
- Department of Obstetrics and Gynaecology, Federal Medical Centre, Birnin Kebbi, Kebbi State, Nigeria
| | - Muhammad Bashir AbdulRahman
- Department of Chemical Pathology, Usmanu Danfodiyo University/Usmanu Danfodiyo University Teaching Hospital, Sokoto, Sokoto State, Nigeria
| | - Mohammad Kaoje Ahmad
- Department of Chemical Pathology, Usmanu Danfodiyo University Teaching Hospital, Sokoto, Sokoto State, Nigeria
| |
Collapse
|
7
|
Oscilowska I, Huynh TYL, Baszanowska W, Prokop I, Surazynski A, Galli M, Zabielski P, Palka J. Proline oxidase silencing inhibits p53-dependent apoptosis in MCF-7 breast cancer cells. Amino Acids 2021; 53:1943-1956. [PMID: 34085157 PMCID: PMC8651586 DOI: 10.1007/s00726-021-03013-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 05/28/2021] [Indexed: 02/02/2023]
Abstract
Proline oxidase (POX) is mitochondrial proline-degrading enzyme of dual apoptosis/survival function. POX expression and proline availability are considered an underlying mechanism for differential POX functions. The mechanism for POX-dependent regulation of cell death/survival was studied in wild-type (MCF-7WT) and shRNA POX-silenced breast cancer cells (MCF-7iPOX). Proline concentration and proteomic analyses were determined by LC/MS/QTOF and LC/MS/ORBITRA, respectively. Inhibition of collagen biosynthesis (proline utilizing process) by 2-methoxyestradiol (2ME) contributed to induction of apoptosis in MCF-7WT cells, as detected by increase in the expression of active caspase-3, -9 and p53. The process was not shown in MCF-7iPOX. In MCF-7iPOX cells prolidase activity and expression as well as proline concentration were drastically increased, compared to MCF-7WT cells. Down-regulation of p53 in MCF-7iPOX cells was corroborated by proteomic analysis showing decrease in the expression of p53-related proteins. The mechanism for down-regulation of p53 expression in MCF-7iPOX cells was found at the level of p53-PEPD complex formation that was counteracted by hydrogen peroxide treatment. In this study, we found that silencing POX modulate pro-survival phenotype of MCF-7 cells and suggest that the mechanism of this process undergoes through down-regulation of p53-dependent signaling.
Collapse
Affiliation(s)
- Ilona Oscilowska
- Department of Medicinal Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222, Bialystok, Poland
| | - Thi Y L Huynh
- Department of Medicinal Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222, Bialystok, Poland
| | - Weronika Baszanowska
- Department of Medicinal Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222, Bialystok, Poland
| | - Izabela Prokop
- Department of Medicinal Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222, Bialystok, Poland
| | - Arkadiusz Surazynski
- Department of Medicinal Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222, Bialystok, Poland
| | - Mauro Galli
- Department of Medical Biology, Medical University of Bialystok, Mickiewicza 2C, 15-222, Bialystok, Poland
| | - Piotr Zabielski
- Department of Medical Biology, Medical University of Bialystok, Mickiewicza 2C, 15-222, Bialystok, Poland
| | - Jerzy Palka
- Department of Medicinal Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222, Bialystok, Poland.
| |
Collapse
|
8
|
Palka J, Oscilowska I, Szoka L. Collagen metabolism as a regulator of proline dehydrogenase/proline oxidase-dependent apoptosis/autophagy. Amino Acids 2021; 53:1917-1925. [PMID: 33818628 PMCID: PMC8651534 DOI: 10.1007/s00726-021-02968-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/19/2021] [Indexed: 12/21/2022]
Abstract
Recent studies on the regulatory role of amino acids in cell metabolism have focused on the functional significance of proline degradation. The process is catalysed by proline dehydrogenase/proline oxidase (PRODH/POX), a mitochondrial flavin-dependent enzyme converting proline into ∆1-pyrroline-5-carboxylate (P5C). During this process, electrons are transferred to electron transport chain producing ATP for survival or they directly reduce oxygen, producing reactive oxygen species (ROS) inducing apoptosis/autophagy. However, the mechanism for switching survival/apoptosis mode is unknown. Although PRODH/POX activity and energetic metabolism were suggested as an underlying mechanism for the survival/apoptosis switch, proline availability for this enzyme is also important. Proline availability is regulated by prolidase (proline supporting enzyme), collagen biosynthesis (proline utilizing process) and proline synthesis from glutamine, glutamate, α-ketoglutarate (α-KG) and ornithine. Proline availability is dependent on the rate of glycolysis, TCA and urea cycles, proline metabolism, collagen biosynthesis and its degradation. It is well established that proline synthesis enzymes, P5C synthetase and P5C reductase as well as collagen prolyl hydroxylases are up-regulated in most of cancer types and control rates of collagen biosynthesis. Up-regulation of collagen prolyl hydroxylase and its exhaustion of ascorbate and α-KG may compete with DNA and histone demethylases (that require the same cofactors) to influence metabolic epigenetics. This knowledge led us to hypothesize that up-regulation of prolidase and PRODH/POX with inhibition of collagen biosynthesis may represent potential pharmacotherapeutic approach to induce apoptosis or autophagic death in cancer cells. These aspects of proline metabolism are discussed in the review as an approach to understand complex regulatory mechanisms driving PRODH/POX-dependent apoptosis/survival.
Collapse
Affiliation(s)
- Jerzy Palka
- Department of Medicinal Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222 Bialystok, Poland
| | - Ilona Oscilowska
- Department of Medicinal Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222 Bialystok, Poland
| | - Lukasz Szoka
- Department of Medicinal Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222 Bialystok, Poland
| |
Collapse
|
9
|
Pritchard N, Kaitu’u-Lino T, Harris L, Tong S, Hannan N. Nanoparticles in pregnancy: the next frontier in reproductive therapeutics. Hum Reprod Update 2021; 27:280-304. [PMID: 33279994 PMCID: PMC9034208 DOI: 10.1093/humupd/dmaa049] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 09/26/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Nanotechnology involves the engineering of structures on a molecular level. Nanomedicine and nano-delivery systems have been designed to deliver therapeutic agents to a target site or organ in a controlled manner, maximizing efficacy while minimizing off-target effects of the therapeutic agent administered. In both reproductive medicine and obstetrics, developing innovative therapeutics is often tempered by fears of damage to the gamete, embryo or developing foetus or of negatively impacting a woman's reproductive potential. Thus, nanomedicine delivery systems may provide alternative targeted intervention strategies, treating the source of the disease and minimizing long-term consequences for the mother and/or her foetus. OBJECTIVE AND RATIONALE This review summarizes the current state of nanomedicine technology in reproductive medicine and obstetrics, including safety, potential applications, future directions and the hurdles for translation. SEARCH METHODS A comprehensive electronic literature search of PubMed and Web of Science databases was performed to identify studies published in English up until February 2020. Relevant keywords were used to obtain information regarding use of nanoparticle technology in fertility and gene therapy, early pregnancy complications (ectopic pregnancy and gestational trophoblastic disease) and obstetric complications (preeclampsia, foetal growth restriction, preterm birth and gestational diabetes) and for selective treatment of the mother or foetus. Safety of specific nanoparticles to the gamete, embryo and foetus was also investigated. OUTCOMES Pre-clinical research in the development of nanoparticle therapeutic delivery is being undertaken in many fields of reproductive medicine. Non-hormonal-targeted nanoparticle therapy for fibroids and endometriosis may provide fertility-sparing medical management. Delivery of interventions via nanotechnology provides opportunities for gene manipulation and delivery in mammalian gametes. Targeting cytotoxic treatments to early pregnancy tissue provides an alternative approach to manage ectopic pregnancies and gestational trophoblastic disease. In pregnancy, nanotherapeutic delivery offers options to stably deliver silencing RNA and microRNA inhibitors to the placenta to regulate gene expression, opening doors to novel genetic treatments for preeclampsia and foetal growth restriction. Restricting delivery of teratogenic drugs to the maternal compartment (such as warfarin) may reduce risks to the foetus. Alternatively, targeted delivery of drugs to the foetus (such as those to treat foetal arrythmias) may minimize side effects for the mother. WIDER IMPLICATIONS We expect that further development of targeted therapies using nanoparticles in a reproductive setting has promise to eventually allow safe and directed treatments for conditions impacting the health and reproductive capacity of women and for the management of pregnancy and serious pregnancy complications.
Collapse
Affiliation(s)
- Natasha Pritchard
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Tu’uhevaha Kaitu’u-Lino
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
- Department of Obstetrics and Gynaecology, Diagnostics Discovery and Reverse Translation, University of Melbourne, Heidelberg, Victoria, Australia
| | - Lynda Harris
- Division of Pharmacy and Optometry, University of Manchester, Manchester, UK
- Maternal and Fetal Health Research Centre, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Maternal and Fetal Health Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Sciences Centre, St Mary’s Hospital, Manchester, UK
| | - Stephen Tong
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Natalie Hannan
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
- Therapeutics Discovery and Vascular Function Group, Department of Obstetrics and Gynaecology, University of Melbourne, Heidelberg, Victoria, Australia
| |
Collapse
|
10
|
Borahay MA, Vincent KL, Motamedi M, Tekedereli I, Salama SA, Ozpolat B, Kilic GS. Liposomal 2-Methoxyestradiol Nanoparticles for Treatment of Uterine Leiomyoma in a Patient-Derived Xenograft Mouse Model. Reprod Sci 2021; 28:271-277. [PMID: 32632769 PMCID: PMC7785630 DOI: 10.1007/s43032-020-00248-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 06/23/2020] [Accepted: 06/30/2020] [Indexed: 10/23/2022]
Abstract
Uterine leiomyomas represent a challenging problem with limited medical treatment options. The anti-tumor agent 2-methoxyestradiol (2-ME) shows promising results but its efficacy is limited by inadequate pharmacokinetics. We previously demonstrated that 2-ME nanoparticles can be successfully formulated and that they show improved in vitro anti-leiomyoma cell activity. Here, we examined the effects of the in vivo delivery of 2-ME nanoparticles in a patient-derived xenograft (PDX) leiomyoma mouse model. Patient-derived leiomyoma tumor tissues were xenografted subcutaneously in estrogen/progesterone pretreated immunodeficient NOG mice. Animals (n = 12) were treated with liposomal 2-ME nanoparticles by intra-peritoneal (IP) injection (50 mg/kg/dose, three times weekly) or control for 28 days. Tumor volume was measured weekly by calipers and prior to sacrifice by ultrasound. In addition, the expression of the cell proliferation marker Ki67 and the apoptosis marker cleaved caspase-3 in tumor tissues after treatment were measured by immunohistochemistry. Liposomal 2-ME treatment was associated with a significant tumor growth inhibition (30.5% less than controls as early as 2 weeks, p = 0.025). In addition, injections of liposomal 2-ME inhibited the expression of the proliferation marker Ki67 (55.8% reduction, p < 0.001). Furthermore, liposomal 2-ME treatment was associated with a 67.5% increase of cleaved caspase-3 expression of increase (p = 0.048). Our findings suggest that liposomal nanoparticle formulation can successfully deliver 2-ME and can be a promising therapeutic strategy for uterine leiomyoma. Further characterization of the liposomal-2ME, including pharmacokinetics, maximal tolerated dose, and safety, is needed in preclinical models prior to clinical trials.
Collapse
Affiliation(s)
- Mostafa A Borahay
- Department of Gynecology & Obstetrics, Johns Hopkins University, 4940 Eastern Ave, Baltimore, MD, 21224-2780, USA.
| | - Kathleen L Vincent
- Department of Obstetrics and Gynecology, and Biomedical Engineering Center, University of Texas Medical Branch, Galveston, TX, USA
| | - Massoud Motamedi
- Biomedical Engineering Center, University of Texas Medical Branch, Galveston, TX, USA
| | - Ibrahim Tekedereli
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Salama A Salama
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Bulent Ozpolat
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Gokhan S Kilic
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
11
|
Kim M, Kim YS, Choi JI, Kim JM, Lee HH, Kim TH. G protein-coupled estrogen receptor 1 expression in normal myometrium, leiomyoma, and adenomyosis tissues of premenopausal women. Gynecol Endocrinol 2020; 36:599-604. [PMID: 32321334 DOI: 10.1080/09513590.2020.1751108] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
To verify the different expression of G protein-coupled estrogen receptor 1 (GPER1) among normal uterine, leiomyoma, and adenomyosis tissues. Normal uterine, leiomyoma, and adenomyosis tissue samples were obtained from women aged 35-52 years from a tertiary university hospital. The tissue samples were subjected to immunohistochemical, Western blot, and reverse-transcription polymerase chain reaction (RT-PCR) analyses of GPER1. GPER1 protein expression was confirmed in the tissues by immunohistochemical and Western blot analyses and compared with GPER1 mRNA levels using RT-PCR. GPER1 was detected in the tissue samples of leiomyoma and adenomyosis, which are estrogen-dependent diseases. GPER1 expression was similar between normal uterine and leiomyoma tissues but was reduced in adenomyosis tissue. The level of phosphorylated extracellular signal-regulated kinases 1/2 was lower and higher in leiomyoma and adenomyosis tissues, respectively, than in normal tissue, but the differences among the groups were not statistically significant. Our immunohistochemical, Western blot, and RT-PCR results suggest that GPER1 expression is involved in cell proliferation in leiomyoma and in cell invasion and migration in adenomyosis. Functional studies of GPER1 involving larger sample sizes should be performed to confirm the adenomyosis and leiomyoma disease mechanisms and eventually to develop new therapeutic interventions for these diseases.
Collapse
Affiliation(s)
- Mijin Kim
- Department of Interdisciplinary Program in Biomedical Science, Soonchunhyang University Graduate School, Asan, Republic of Korea
| | - Yeon-Suk Kim
- Department of Interdisciplinary Program in Biomedical Science, Soonchunhyang University Graduate School, Asan, Republic of Korea
| | - Jeong In Choi
- Department of Obstetrics and Gynecology, Soonchunhyang University College of Medicine, Bucheon, Republic of Korea
| | - Jun-Mo Kim
- Department of Urology, Soonchunhyang University College of Medicine, Bucheon, Republic of Korea
| | - Hae-Hyeog Lee
- Department of Obstetrics and Gynecology, Soonchunhyang University College of Medicine, Bucheon, Republic of Korea
| | - Tae-Hee Kim
- Department of Obstetrics and Gynecology, Soonchunhyang University College of Medicine, Bucheon, Republic of Korea
| |
Collapse
|
12
|
Tofovic SP, Jackson EK. Estradiol Metabolism: Crossroads in Pulmonary Arterial Hypertension. Int J Mol Sci 2019; 21:ijms21010116. [PMID: 31877978 PMCID: PMC6982327 DOI: 10.3390/ijms21010116] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 12/17/2019] [Indexed: 12/17/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a debilitating and progressive disease that predominantly develops in women. Over the past 15 years, cumulating evidence has pointed toward dysregulated metabolism of sex hormones in animal models and patients with PAH. 17β-estradiol (E2) is metabolized at positions C2, C4, and C16, which leads to the formation of metabolites with different biological/estrogenic activity. Since the first report that 2-methoxyestradiol, a major non-estrogenic metabolite of E2, attenuates the development and progression of experimental pulmonary hypertension (PH), it has become increasingly clear that E2, E2 precursors, and E2 metabolites exhibit both protective and detrimental effects in PH. Furthermore, both experimental and clinical data suggest that E2 has divergent effects in the pulmonary vasculature versus right ventricle (estrogen paradox in PAH). The estrogen paradox is of significant clinical relevance for understanding the development, progression, and prognosis of PAH. This review updates experimental and clinical findings and provides insights into: (1) the potential impacts that pathways of estradiol metabolism (EMet) may have in PAH; (2) the beneficial and adverse effects of estrogens and their precursors/metabolites in experimental PH and human PAH; (3) the co-morbidities and pathological conditions that may alter EMet and influence the development/progression of PAH; (4) the relevance of the intracrinology of sex hormones to vascular remodeling in PAH; and (5) the advantages/disadvantages of different approaches to modulate EMet in PAH. Finally, we propose the three-tier-estrogen effects in PAH concept, which may offer reconciliation of the opposing effects of E2 in PAH and may provide a better understanding of the complex mechanisms by which EMet affects the pulmonary circulation–right ventricular interaction in PAH.
Collapse
Affiliation(s)
- Stevan P. Tofovic
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, BST E1240, 200 Lothrop Street, Pittsburgh, PA 15261, USA
- Department of Pharmacology and Chemical Biology University of Pittsburgh School of Medicine, 100 Technology Drive, PA 15219, USA;
- Correspondence: ; Tel.: +1-412-648-3363
| | - Edwin K. Jackson
- Department of Pharmacology and Chemical Biology University of Pittsburgh School of Medicine, 100 Technology Drive, PA 15219, USA;
| |
Collapse
|
13
|
2-Methoxyestradiol Attenuates Testosterone-Induced Benign Prostate Hyperplasia in Rats through Inhibition of HIF-1 α/TGF- β/Smad2 Axis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:4389484. [PMID: 30154949 PMCID: PMC6093036 DOI: 10.1155/2018/4389484] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 06/24/2018] [Accepted: 07/09/2018] [Indexed: 12/18/2022]
Abstract
Benign prostatic hyperplasia (BPH) is a common disorder in the male population. 2-Methoxyestradiol (2ME) is an end metabolite of estrogens with pleiotropic pharmacological properties. This study aimed to explore the potential ameliorative effects of 2ME against testosterone-induced BPH in rats. 2-Methoxyestradiol (50 and 100 mg/kg, dissolved in DMSO) prevented the rise in prostatic index and weight in comparison to testosterone-alone-treated animals for 2 weeks. Histological examination indicated that 2ME ameliorated pathological changes in prostate architecture. This was confirmed by the ability of 2ME to decrease the glandular epithelial height when compared to the testosterone group. Also, 2ME improved testosterone-induced oxidative stress as it inhibited the rise in lipid peroxide content and the exhaustion of superoxide dismutase (SOD) activity. The beneficial effects of 2ME against the development of BPH were substantiated by assessing proliferation markers, preventing the rise in cyclin D1 protein expression and enhancing Bax/Bcl2 mRNA ratio. It significantly reduced prostate content of tumor necrosis factor α (TNF-α), interleukin-1β (IL-1β), nuclear factor κB (NF-κB), and transforming growth factor β (TGF-β). In addition, 2ME reduced hypoxia-inducible factor 1-α (HIF-1α) and phospho-Smad2 (p-Smad2) protein expression compared to the testosterone group. In conclusion, 2ME attenuates experimentally induced BPH by testosterone in rats through, at least partly, inhibition of HIF-1α/TGF-β/Smad2 axis.
Collapse
|
14
|
Friend DR. Drug delivery for the treatment of endometriosis and uterine fibroids. Drug Deliv Transl Res 2017; 7:829-839. [DOI: 10.1007/s13346-017-0423-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
15
|
Lee JS, Ahn C, Kang HY, Jeung EB. Effect of 2-methoxyestradiol on SK-LMS-1 uterine leiomyosarcoma cells. Oncol Lett 2017; 14:103-110. [PMID: 28693141 PMCID: PMC5494911 DOI: 10.3892/ol.2017.6165] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 03/07/2017] [Indexed: 12/22/2022] Open
Abstract
An endogenous metabolite of 17β-estradiol, 2-methoxyestradiol (2-ME), has affinity for estrogen receptors. This compound was reported to be a promising antitumor drug due to its anti-proliferative effects on a wide range of tumor cell types. Numerous previous studies have been performed to evaluate the cytotoxic effects of 2-ME on tumor cell lines in following the induction of G2/M cell cycle arrest and subsequent apoptosis. Uterine leiomyosarcoma (ULMS) is a relatively rare malignant smooth muscle cell tumor that develops in the uterus muscle layer. The aim of the present study was to examine the in vitro anti-proliferative effects of 2-ME on SK-LMS-1 human leiomyosarcoma cells. An MTT assay, terminal deoxynucleotidyltransferase-mediated dUTP nick-end labeling assay, immunocytochemistry and western blotting were performed. A high concentration (10−5 M) of 2-ME was identified to have an anti-proliferative effect on SK-LMS-1 cells. Additionally, expression of the apoptosis markers was upregulated in the presence of 10−5 M 2-ME, according to western blot analysis. Furthermore, the expression level of an autophagic marker, light chain 3, was increased by 2-ME treatment in a dose-dependent manner. This was associated with cell death induced by the upregulation of phosphorylated extracellular-signal-regulated kinase 1/2 signaling pathway. The results of the present study demonstrated that 2-ME, which is used as a therapeutic agent for treating solid tumors, exhibits apoptotic and anti-proliferative effects depending on the dose. Therefore, 2-ME may be a potential therapeutic reagent for human ULMS, but the appropriate dose of this compound should be carefully selected.
Collapse
Affiliation(s)
- Ji-Sun Lee
- Laboratory of Veterinary Biochemistry and Molecular Biology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Changhwan Ahn
- Laboratory of Veterinary Biochemistry and Molecular Biology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Hee Young Kang
- Laboratory of Veterinary Biochemistry and Molecular Biology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Eui-Bae Jeung
- Laboratory of Veterinary Biochemistry and Molecular Biology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| |
Collapse
|
16
|
Bastos P, Gomes T, Ribeiro L. Catechol-O-Methyltransferase (COMT): An Update on Its Role in Cancer, Neurological and Cardiovascular Diseases. Rev Physiol Biochem Pharmacol 2017; 173:1-39. [DOI: 10.1007/112_2017_2] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
17
|
Borahay MA, Asoglu MR, Mas A, Adam S, Kilic GS, Al-Hendy A. Estrogen Receptors and Signaling in Fibroids: Role in Pathobiology and Therapeutic Implications. Reprod Sci 2016; 24:1235-1244. [PMID: 27872195 DOI: 10.1177/1933719116678686] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Uterine fibroids are the most common gynecologic tumors with a significant medical and financial burden. Several genetic, hormonal, and biological factors have been shown to contribute to the development and growth of fibroid tumors. Of these factors, estrogen is particularly critical since fibroids are considered estrogen dependent because no prepubertal cases have been described in the literature and tumors tend to regress after menopause. Understanding the role of estrogen in fibroids is not only important for understanding the pathobiology of fibroids but also for the development of successful therapeutics. In this review, we discuss the types and structure of estrogen receptors (nuclear and membrane bound, including α and β receptors and G protein-coupled estrogen receptor 1 GPER1). Estrogen-signaling pathways in fibroids include genomic (direct and indirect) and nongenomic including Ras-Raf-MEK (MAPK/Erk Kinase)-mitogen-activated protein kinase (MAPK) and phosphatidylinositide 3-kinase (PI3K)-phosphatidylinositol-3,4,5-trisphosphate (PIP3)-Akt (Protein kinase B)-mammalian target of rapamycin (mTOR) pathways; shortly Ras-Raf-MEK-MAPK and PI3K-PIP3-Akt-mTOR pathways. Several aberrations in estrogen receptors and signaling pathways are implicated in fibroid pathobiology. Current therapeutic and research agents targeting ERs/signaling include gonadotropin-releasing hormone (GnRH) agonists, GnRH antagonists, aromatase inhibitors, selective ER modulators, gene therapy, and others. Future research can identify potential targets for the development of novel treatments. In particular, epigenomics of estrogen activity and individualized (precision) medicine appear to be attractive areas for future research.
Collapse
Affiliation(s)
- Mostafa A Borahay
- 1 Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,2 Department of Obstetrics & Gynecology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Mehmet R Asoglu
- 2 Department of Obstetrics & Gynecology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Aymara Mas
- 3 Department of Obstetrics & Gynecology, Georgia Regents University, Augusta, GA, USA
| | - Sarah Adam
- 4 Department of Obstetrics & Gynecology, Mercer University, Macon, GA, USA
| | - Gokhan S Kilic
- 2 Department of Obstetrics & Gynecology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Ayman Al-Hendy
- 3 Department of Obstetrics & Gynecology, Georgia Regents University, Augusta, GA, USA
| |
Collapse
|
18
|
Borahay MA, Al-Hendy A, Kilic GS, Boehning D. Signaling Pathways in Leiomyoma: Understanding Pathobiology and Implications for Therapy. Mol Med 2015; 21:242-56. [PMID: 25879625 DOI: 10.2119/molmed.2014.00053] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 04/13/2015] [Indexed: 12/15/2022] Open
Abstract
Uterine leiomyomas are the most common tumors of the female genital tract, affecting 50% to 70% of females by the age of 50. Despite their prevalence and enormous medical and economic impact, no effective medical treatment is currently available. This is, in part, due to the poor understanding of their underlying pathobiology. Although they are thought to start as a clonal proliferation of a single myometrial smooth muscle cell, these early cytogenetic alterations are considered insufficient for tumor development and additional complex signaling pathway alterations are crucial. These include steroids, growth factors, transforming growth factor-beta (TGF-β)/Smad; wingless-type (Wnt)/β-catenin, retinoic acid, vitamin D, and peroxisome proliferator-activated receptor γ (PPARγ). An important finding is that several of these pathways converge in a summative way. For example, mitogen-activated protein kinase (MAPK) and Akt pathways seem to act as signal integrators, incorporating input from several signaling pathways, including growth factors, estrogen and vitamin D. This underlines the multifactorial origin and complex nature of these tumors. In this review, we aim to dissect these pathways and discuss their interconnections, aberrations and role in leiomyoma pathobiology. We also aim to identify potential targets for development of novel therapeutics.
Collapse
Affiliation(s)
- Mostafa A Borahay
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, Texas, United States of America.,Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, Texas, United States of America
| | - Ayman Al-Hendy
- Department of Obstetrics and Gynecology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, United States of America
| | - Gokhan S Kilic
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Darren Boehning
- Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, Texas, United States of America
| |
Collapse
|
19
|
Leptin influences estrogen metabolism and accelerates prostate cell proliferation. Life Sci 2014; 121:10-5. [PMID: 25433128 DOI: 10.1016/j.lfs.2014.11.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Revised: 11/12/2014] [Accepted: 11/13/2014] [Indexed: 01/17/2023]
Abstract
AIM The present study was designed to investigate the effect of leptin on estrogen metabolism in prostatic cells. MAIN METHODS Malignant (PC-3) and benign (BPH-1) human prostate cells were treated with 17-β-hydroxyestradiol (1 μM) alone or in combination with leptin (0.4, 4, 40 ng/ml) for 72 h. Cell proliferation assay, immunocytochemical staining of estrogen receptor (ER), liquid chromatography-tandem mass spectrometry method (LC-MS) and semi-quantitative reverse transcriptase polymerase chain reaction (RT-PCR) were used. KEY FINDINGS Cell proliferation assay demonstrated that leptin caused significant growth potentiation in both cells. Immunocytochemical staining showed that leptin significantly increased the expression of ER-α and decreased that of ER-β in PC-3 cells. LC-MS method revealed that leptin increased the concentration 4-hydroxyestrone and/or decreased that of 2-methoxyestradiol, 4-methoxyestradiol and 2-methoxyestrone. Interestingly, RT-PCR showed that leptin significantly up-regulated the expression of aromatase and cytochrome P450 1B1 (CYP1B1) enzymes; however down-regulated the expression of catechol-o-methyltransferase (COMT) enzyme. SIGNIFICANCE These data indicate that leptin-induced proliferative effect in prostate cells might be partly attributed to estrogen metabolism. Thus, leptin might be a novel target for therapeutic intervention in prostatic disorders.
Collapse
|
20
|
Zhu L, Song Y, Li M. 2-Methoxyestradiol inhibits bleomycin-induced systemic sclerosis through suppression of fibroblast activation. J Dermatol Sci 2014; 77:63-70. [PMID: 25465161 DOI: 10.1016/j.jdermsci.2014.10.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2014] [Revised: 09/30/2014] [Accepted: 10/20/2014] [Indexed: 02/03/2023]
Abstract
BACKGROUND The most dominant feature of systemic sclerosis (SSc) is fibrosis, which is caused by overproduction of collagen by fibroblasts. 2-Methoxyestradiol (2-ME) has exhibited disease-modifying activity in animal models of rheumatoid arthritis and autoimmune encephalomyelitis and inhibitory effect in cell proliferation and collagen synthesis. Therefore, we hypothesized that 2-ME may exhibit antifibrotic effect in SSc. OBJECTIVE To investigate the antifibrotic effect of 2-ME in SSc. METHODS We established a bleomycin-induced SSc mice model by injection with bleomycin daily for 21 days. 2-ME (100mg/kg/d) was simultaneously administered for 14 days. On the end of Week1 (W1), W2, W3 and W4, skins and lungs were collected for histological examination and analysis of hydroxyproline content and mRNA level of α1(I) procollagen (COL1A1) and COL1A2. In skin fibroblasts derived from SSc patients and healthy subjects treated with 2-ME (1, 5, or 25 μM), we examined cell proliferation, expression of α-smooth muscle actin (SMA) and mRNA level of COL1A1, COL1A2, COL3A1, matrix metalloproteinase(MMP)-1 and tissue inhibitors of MMP (TIMP)-1. RESULTS We found reduced dermal thickness and lung fibrosis and decreased hydroxyproline content and mRNA level of COL1A1 and COL1A2 in skin and lung in SSc mice treated with 2-ME. In cell study, we observed a dose- and time-dependent inhibitory effect on proliferation of SSc fibroblasts by 2-ME. We also detected reduced α-SMA expression, decreased mRNA level of COL1A1, COL1A2, COL3A1 and TIMP-1, and increased mRNA level of MMP-1 in SSc fibroblasts treated with 2-ME. CONCLUSION 2-ME could suppress SSc tissue fibrosis, which may be attributable to its inhibitory effect on the excessive proliferation, differentiation and production of collagen in fibroblasts. 2-ME is rising as a prospective agent for control of fibrosis in SSc.
Collapse
Affiliation(s)
- Lubing Zhu
- Department of Dermatology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Yinghua Song
- Department of Dermatology, Wuhan No.1 Hospital, Wuhan, 430022, China
| | - Ming Li
- Department of Dermatology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
| |
Collapse
|
21
|
Fujisawa C, Castellot JJ. Matrix production and remodeling as therapeutic targets for uterine leiomyoma. J Cell Commun Signal 2014; 8:179-94. [PMID: 25012731 DOI: 10.1007/s12079-014-0234-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 05/22/2014] [Indexed: 01/06/2023] Open
Abstract
Uterine leiomyoma, commonly known as fibroids, is a benign neoplasm of smooth muscle in women. The incidence of clinically symptomatic fibroids in reproductive-age women is approximately 20 %, with nearly 80 % of black women suffering from this condition. Symptoms include severe pain and hemorrhage; fibroids are also a major cause of infertility or sub-fertility in women. Uterine leiomyoma consist of hyperplastic smooth muscle cells and an excess deposition of extracellular matrix, specifically collagen, fibronectin, and sulfated proteoglycans. Extracellular matrix components interact and signal through integrin-β1 on the surface of uterine leiomyoma smooth muscle cells, provide growth factor storage, and act as co-receptors for growth factor-receptor binding. ECM and growth factor signaling through integrin-β1 and growth factor receptors significantly increases cell proliferation and ECM deposition in uterine leiomyoma. Growth factors TGF-β, IGF, PDGF, FGF and EGF are all shown to promote uterine leiomyoma progression and signal through multiple pathways to increase the expression of genes encoding matrix or matrix-modifying proteins. Decreasing integrin expression, reducing growth factor action and inhibiting ECM action on uterine leiomyoma smooth muscle cells are important opportunities to treat uterine leiomyoma without use of the current surgical procedures. Both natural compounds and chemicals are shown to decrease fibrosis and uterine leiomyoma progression, but further analysis is needed to make inroads in treating this common women's health issue.
Collapse
Affiliation(s)
- Caitlin Fujisawa
- Public Heath and Professional Degrees Program, Tufts University School of Medicine, Boston, MA, 02111, USA
| | | |
Collapse
|
22
|
Zhang D, Rajaratnam V, Al-Hendy O, Halder S, Al-Hendy A. Green Tea Extract Inhibition of Human Leiomyoma Cell Proliferation Is Mediated via Catechol- O-Methyltransferase. Gynecol Obstet Invest 2014; 78:109-18. [DOI: 10.1159/000363410] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 05/03/2014] [Indexed: 11/19/2022]
|
23
|
Li D, Xu X, Qian R, Geng J, Zhang Y, Xie X, Wang Y, Zou X. Effect of Lichong decoction on expression of Bcl-2 and Bcl-2-associated X protein mRNAs in hysteromyoma model rat. J TRADIT CHIN MED 2013; 33:238-42. [DOI: 10.1016/s0254-6272(13)60132-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
24
|
Abstract
BACKGROUND The purpose of this work was to engineer polymeric nanoparticles to encapsulate and deliver 2-methoxyestradiol, a potential antitumor drug for treatment of uterine leiomyoma (fibroids), the most common hormone-dependent pathology affecting women of reproductive age. METHODS/RESULTS Encapsulation efficiency and drug release from the nanoparticles were monitored by HPLC. Cell morphology and in vitro cytotoxicity experiments were carried out in a human leiomyoma cell line. The nanoparticles displayed high encapsulation efficiency (>86%), which was verified by differential scanning calorimetry and x-ray diffraction. Excellent long-term stability of the nanoparticles and gradual drug release without burst were also observed. Cellular uptake of fluorescent nanoparticles was confirmed by confocal imaging. The drug-loaded poly(lactic acid) and poly(lactic-co-glycolic acid) nanoparticles induced cytotoxicity in human leiomyoma cells to a significantly greater extent than the free drug at 0.35 µM. CONCLUSION This novel approach represents a potential fertility-preserving alternative to hysterectomy.
Collapse
|
25
|
Abstract
Sex differences in the incidence of liver cirrhosis and portal hypertension have been reported by epidemiological studies. Previous studies have indicated that estrogen therapy improved hepatic fibrosis, inhibited the activation of hepatic stellate cells, and reduced portal pressure, whereas the administration of exogenous estrogens resulted in some potential risks, limiting their clinical use. However, the biological actions of estrogens are mediated by three subtypes of estrogen receptors (ERs): ERα, ERβ, and G-protein-coupled ER. These ER subtypes act in distinct ways and exert different biological effects that mediate genomic and nongenomic events, resulting in tissue-specific responses. In addition, active estrogen metabolites, with little or no affinity for ERs, could mediate the fibrosuppressive effect of estrogens through an ER-independent pathway. Taken together, such specific estrogen derivatives as ER selective agonists, or active estrogen metabolites, would provide novel therapeutic opportunities, stratifying this hormonal treatment, thereby reducing undesired side-effects in the treatment of liver cirrhosis and portal hypertension.
Collapse
|
26
|
Machado-Linde F, Pelegrin P, Sanchez-Ferrer ML, Leon J, Cascales P, Parrilla JJ. 2-Methoxyestradiol in the Pathophysiology of Endometriosis: Focus on Angiogenesis and Therapeutic Potential. Reprod Sci 2012; 19:1018-29. [DOI: 10.1177/1933719112446080] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
| | - Pablo Pelegrin
- Inflammation and Experimental Surgery Group, Centro de Investigación Biomédica en Red en el Área temática de Enfermedades Hepáticas y Digestivas (CIBERehd), Hospital Virgen Arrixaca, Murcia, Spain
| | | | - Josefa Leon
- Department of Hospital Pharmacy, Hospital Morales Meseguer, Murcia, Spain
| | - Pedro Cascales
- Department of General Surgery, Hospital Virgen Arrixaca, Murcia, Spain
| | - Juan J. Parrilla
- Department of Gynecology and Obstetrics, Hospital Virgen Arrixaca, Murcia, Spain
| |
Collapse
|
27
|
Salama SA, Diaz-Arrastia CR, Kilic GS, Kamel MW. 2-Methoxyestradiol causes functional repression of transforming growth factor β3 signaling by ameliorating Smad and non-Smad signaling pathways in immortalized uterine fibroid cells. Fertil Steril 2012; 98:178-84. [PMID: 22579131 DOI: 10.1016/j.fertnstert.2012.04.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Revised: 03/30/2012] [Accepted: 04/03/2012] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To investigate the effects and the mechanism of action of 2-methoxyestradiol (2ME(2)) on transforming growth factor (TGF) β3-induced profibrotic response in immortalized human uterine fibroid smooth muscle (huLM) cells. DESIGN Laboratory study. SETTING University research laboratory. PATIENTS(S) Not applicable. INTERVENTIONS(S) Not applicable. MAIN OUTCOME MEASURE(S) huLM cells were treated with TGF-β3 (5 ηg/mL) in the presence or absence of specific Smad3 inhibitor SIS3 (1 μmol/L), inhibitor of the PI3K/Akt (LY294002, 10 μmol/L), or 2ME(2) (0.5 μmol/L), and the expression of collagen (Col) type I(αI), Col III(αI), plasminogen activator inhibitor (PAI) 1, connective tissue growth factor (CTGF), and α-smooth muscle actin (α-SMA) were determined by real-time reverse-transcription polymerase chain reaction and immunoblotting. The effect of 2ME(2) on Smad-microtubule binding was evaluated by coimmunoprecipitation. RESULT(S) Our data revealed that TGF-β3-induced fibrogenic response in huLM is mediated by both Smad-dependent and Smad-independent PI3K/Akt/mTOR signaling pathways. 2ME(2) abrogates TGF-β3-induced expression of Col I(αI), Col III(αI), PAI-1, CTGF, and α-SMA. Molecularly, 2ME(2) ameliorates TGF-β3-induced Smad2/3 phosphorylation and nuclear translocation. In addition, 2ME(2) inhibits TGF-β3-induced activation of the PI3K/Akt/mTOR pathway. CONCLUSION(S) TGF-β3-induced profibrotic response in fibroid cells is mediated by Smad-dependent and Smad-independent PI3K/Akt/mTOR pathways. 2ME(2) inhibits TGF-β3 profibrotic effects in huLM cells by ameliorating both Smad-dependent and Smad-independent signaling pathways.
Collapse
Affiliation(s)
- Salama A Salama
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, Texas 77030, USA.
| | | | | | | |
Collapse
|
28
|
Mosli HA, Al-Abd AM, El-Shaer MA, Khedr A, Gazzaz FS, Abdel-Naim AB. Local inflammation influences oestrogen metabolism in prostatic tissue. BJU Int 2011; 110:274-82. [PMID: 22145816 DOI: 10.1111/j.1464-410x.2011.10796.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
UNLABELLED What's known on the subject? and What does the study add? The role of oestrogen in prostatic inflammation has been extensively shown. The catechol oestrogens are known to be more potent oestrogenic moieties that not only aggravate the inflammatory response in situ, but are also believed to have oxidative stress and genotoxic effects. The present study highlights a significant role of inflammation in oestrogen metabolism and, particularly, in generating 'bad' oestrogen metabolites. This finding may pave the way for new therapeutic methods for the treatment and/or prevention of prostate diseases. OBJECTIVE • To investigate the impact of experimentally induced inflammation on oestrogen metabolism in rat prostate. MATERIALS AND METHODS • Prostatitis was induced in normal and oestrogen-treated male rats by injecting 2% carrageenan solution into the ventral prostate. After 48 h, the rats were killed and the ventral prostate was collected. • Prostatic inflammation and proliferation were confirmed by gross visual evidence, histology and elevated tumour necrosis factor-α, prostaglandin E(2) and cyclin-D(1) . • Expression of oestrogen-metabolizing enzymes was assessed using capillary electrophoresis, and oestrogen metabolites within prostate tissue were assayed using liquid chromatography mass spectrometry. RESULTS • Animals exposed to carrageenan insult combined with oestrogen treatment showed the most prominent inflammatory and proliferative response. • Treatment of animals with oestrogen alone induced moderate inflammation and proliferation. • Oestrogen-metabolizing enzymes were overexpressed in animals with experimental prostatitis with sequential accumulation of catechol oestrogens within prostatic tissues. • Oestrogen receptor-α was underexpressed in the prostatitis with oestrogen group, while oestrogen receptor-β was overexpressed. CONCLUSIONS • The present work provides experimental evidence that local inflammation enhances oestrogen synthesis and directs oestrogen metabolism to generate catechol oestrogens within prostatic tissues. • This may contribute, at least partly, to enhanced prostatic cell proliferation.
Collapse
Affiliation(s)
- Hisham A Mosli
- Department of Urology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | | | | | | | | |
Collapse
|
29
|
Salama S, Diaz-Arrastia C, Patel D, Botting S, Hatch S. 2-Methoxyestradiol, an Endogenous Estrogen Metabolite, Sensitizes Radioresistant MCF-7/FIR Breast Cancer Cells Through Multiple Mechanisms. Int J Radiat Oncol Biol Phys 2011; 80:231-9. [DOI: 10.1016/j.ijrobp.2010.10.080] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2010] [Revised: 10/12/2010] [Accepted: 10/13/2010] [Indexed: 10/18/2022]
|
30
|
Halder SK, Goodwin JS, Al-Hendy A. 1,25-Dihydroxyvitamin D3 reduces TGF-beta3-induced fibrosis-related gene expression in human uterine leiomyoma cells. J Clin Endocrinol Metab 2011; 96:E754-62. [PMID: 21289245 PMCID: PMC3070259 DOI: 10.1210/jc.2010-2131] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Uterine leiomyomas (fibroids) are the most common benign estrogen-dependent tumors of premenopausal women. TGF-β3 up-regulates the synthesis of many of extracellular matrix proteins that are associated with tissue fibrosis. OBJECTIVE To examine the effect of 1,25-dihydroxyvitamin D(3) (vitamin D(3)) on TGF-β3-induced fibrosis-related protein expression in immortalized human uterine leiomyoma (HuLM) cells. METHODS HuLM cells were treated with TGF-β3 with or without vitamin D(3). Western blot analyses were employed to test the effect of vitamin D(3) on TGF-β3-induced protein expression of collagen type 1, fibronectin, and plasminogen activator inhibitor-1 proteins. Western blots as well as immunofluorescence analyses were used to verify the effect of vitamin D(3) on TGF-β3-induced Smad activation involved in extracellular matrix protein synthesis and deposition, which ultimately lead to tissue fibrosis. RESULTS We observed that TGF-β3 induced fibronectin and collagen type 1 protein expression in HuLM cells, and that effect was suppressed by vitamin D(3). TGF-β3 also induced protein expression of plasminogen activator inhibitor-1, an important TGF-β target, in HuLM cells, which was also inhibited by vitamin D(3). Additionally, TGF-β3 induced phosphorylation of Smad2 as well as nuclear translocation of Smad2 and Smad3 in HuLM cells, whereas vitamin D significantly reduced all these TGF-β3-mediated effects. Therefore, our results suggest that vitamin D(3) has consistently reduced TGF-β3 effects that are involved in the process of fibrosis in human leiomyoma cells. CONCLUSION Vitamin D(3) is an antifibrotic factor that might be potentially useful as a novel therapeutic for nonsurgical treatment of benign uterine fibroids.
Collapse
Affiliation(s)
- Sunil K Halder
- Center for Women’s Health Research, Department of Obstetrics and Gynecology, Meharry Medical College, Nashville, Tennessee 37208, USA
| | | | | |
Collapse
|
31
|
Abstract
2-Methoxyestradiol (2MeO-E2) is an endogenous metabolite of estrogen which was initially considered to be inactive. During the last few decades it has been shown that 2MeO-E2 is a promising anticancer drug. In vitro experiments have demonstrated that it has several anticancer activities, and potential to alleviate hypertension, glomerulosclerosis, hypercholesterolemia, and other disorders. However, due to its low solubility and extensive glucuronidation, to achieve effective concentrations large doses of 2MeO-E2 would be required. Clinical studies reflected very high inter- and intrapatient variability and oral bioavailability of 1 to 2%. Thus, this review paper highlights the origin of this compound, its therapeutic promises, and possible mechanisms of action. It also discusses the pharmacokinetic properties of 2MeO-E2 as well as current developments to overcome low drug solubility and its extensive first pass metabolism.
Collapse
Affiliation(s)
- Svetlana Verenich
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University PO Box 980533, Richmond, VA
| | - Phillip M. Gerk
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University PO Box 980533, Richmond, VA
| |
Collapse
|
32
|
Mamalis A, Markopoulou C, Lagou A, Vrotsos I. Oestrogen regulates proliferation, osteoblastic differentiation, collagen synthesis and periostin gene expression in human periodontal ligament cells through oestrogen receptor beta. Arch Oral Biol 2010; 56:446-55. [PMID: 21130420 DOI: 10.1016/j.archoralbio.2010.11.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2010] [Revised: 10/25/2010] [Accepted: 11/08/2010] [Indexed: 11/26/2022]
Abstract
OBJECTIVE The present study was designed to examine how oestrogen regulates proliferation, osteoblastic differentiation, collagen synthesis and periostin gene expression in primary human periodontal ligament (hPDL) cells. DESIGN The short interfering RNA (siRNA) technique was used to inhibit oestrogen receptor beta (ERβ) expression hPDL cells. hPDL cell were isolated and fully characterized. A colorimetric assay was applied for the determination of alkaline phosphatase (ALP). An ELISA kit was used to detect osteocalcin (OCN) levels. Collagen synthesis was determined by measuring the incorporation of L-[3H] praline. RT-PCR was performed to detection of periostin mRNA relative gene expression. RESULTS ERβ mRNA was expressed in hPDL cells and significant inhibition of mRNA expression and ERβ mature protein of the ERβ was evident in the siRNA group. At 72h, there was a significant increase in non-transfected hPDL cell proliferation after estradiol stimulation. Addition of 17β-estradiol significantly enhanced ALP activity and production of OCN in non-transfected cells but had no effect on collagen synthesis. A clear increase in periostin mRNA expression levels was observed after incubating hPDL cells with estradiol. In hPDL-siERβ cells, the application of estradiol did not produce any evident differences in periostin mRNA expression CONCLUSIONS ERβ may play important roles in oestrogen-induced effects on hPDL cell proliferation, osteoblastic differentiation and expression of key molecules for the functional and structural integrity of the periodontium (i.e. periostin).
Collapse
Affiliation(s)
- Anastasios Mamalis
- Department of Periodontics, Dental School, University of Texas, Health Science Center, San Antonio, TX 78229-3900, USA.
| | | | | | | |
Collapse
|
33
|
Zhang D, Al-Hendy M, Richard-Davis G, Montgomery-Rice V, Sharan C, Rajaratnam V, Khurana A, Al-Hendy A. Green tea extract inhibits proliferation of uterine leiomyoma cells in vitro and in nude mice. Am J Obstet Gynecol 2010; 202:289.e1-9. [PMID: 20074693 DOI: 10.1016/j.ajog.2009.10.885] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2009] [Revised: 08/31/2009] [Accepted: 10/28/2009] [Indexed: 12/11/2022]
Abstract
OBJECTIVE The purpose of this study was to investigate the effect of epigallocatechin gallate (EGCG) on rat leiomyoma (ELT3) cells in vitro and in a nude mice model. STUDY DESIGN ELT3 cells were treated with various concentrations of EGCG. Cell proliferation, proliferation cell nuclear antigen (PCNA), and cyclin-dependent kinase 4 (Cdk4) protein levels were evaluated. ELT3 cells were inoculated subcutaneously in female athymic nude mice. Animals were fed 1.25 mg EGCG (in drinking water)/mouse/day. Tumors were collected and evaluated at 4 and 8 weeks after the treatment. RESULTS Inhibitory effect of EGCG (200 micromol/L) on ELT3 cells was observed after 24 hours of treatment (P < .05). At > or = 50 micromol/L, EGCG significantly decreased PCNA and Cdk4 protein levels (P < .05). In vivo, EGCG treatment dramatically reduced the volume and weight of tumors at 4 and 8 weeks after the treatment (P < .05). The PCNA and Cdk4 protein levels were significantly reduced in the EGCG-treated group (P < .05). CONCLUSION EGCG effectively inhibits proliferation and induces apoptosis in rat ELT3 uterine leiomyoma cells in vitro and in vivo.
Collapse
|
34
|
Zhang D, Al-Hendy M, Richard-Davis G, Montgomery-Rice V, Rajaratnam V, Al-Hendy A. Antiproliferative and proapoptotic effects of epigallocatechin gallate on human leiomyoma cells. Fertil Steril 2009; 94:1887-93. [PMID: 19819432 DOI: 10.1016/j.fertnstert.2009.08.065] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2009] [Revised: 08/26/2009] [Accepted: 08/27/2009] [Indexed: 01/12/2023]
Abstract
OBJECTIVE To investigate the effects of epigallocatechin gallate (EGCG), an extract of green tea on cultured human leiomyoma cells (HuLM). DESIGN Laboratory study. SETTING University hospitals. PATIENT(S) Not applicable. INTERVENTION(S) Not applicable. MAIN OUTCOME MEASURE(S) The HuLM cells were treated with various EGCG concentrations. Cell proliferation was assayed using Hoechst 33258 dye, and apoptosis by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay. Total RNA was isolated, and gene expression profiling was performed on 84 key genes related to 18 different signal transduction pathways. The protein levels of PCNA, CDK4, BCL2, and BAX were examined by Western blot analysis. RESULT(S) The HuLM cells treated with EGCG showed a dose-dependent and time-dependent inhibition of cell proliferation. The TUNEL staining indicated a significant increase in apoptosis in HuLM cells treated with 100 μM of EGCG compared with untreated control. Gene expression profiling indicated that EGCG treatment up-regulated representative genes from the transforming growth factor β (TGF-β) and stress pathways, while inhibiting the survival pathway and NFκB-dependent inflammatory pathway. Western blot analysis confirmed that EGCG at ≥50 μM significantly decreased the expression of PCNA, CDK4, and BCL2 as well as increased the expression of the proapoptotic BAX in a dose-dependent manner. CONCLUSION(S) Epigallocatechin gallate inhibits the proliferation of HuLM cells and induces apoptosis. These results suggest that EGCG may be a potential anti-uterine fibroid agent acting through multiple signal transduction pathways.
Collapse
Affiliation(s)
- Dong Zhang
- Department of Obstetrics and Gynecology, Center for Women's Health Research, Meharry Medical College, Nashville, Tennessee 37208, USA
| | | | | | | | | | | |
Collapse
|
35
|
Salama SA, Kamel MW, Botting S, Salih SM, Borahay MA, Hamed AA, Kilic GS, Saeed M, Williams MY, Diaz-Arrastia CR. Catechol-o-methyltransferase expression and 2-methoxyestradiol affect microtubule dynamics and modify steroid receptor signaling in leiomyoma cells. PLoS One 2009; 4:e7356. [PMID: 19809499 PMCID: PMC2752809 DOI: 10.1371/journal.pone.0007356] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2009] [Accepted: 08/27/2009] [Indexed: 11/18/2022] Open
Abstract
Context Development of optimal medicinal treatments of uterine leiomyomas represents a significant challenge. 2-Methoxyestradiol (2ME) is an endogenous estrogen metabolite formed by sequential action of CYP450s and catechol-O-methyltransferase (COMT). Our previous study demonstrated that 2ME is a potent antiproliferative, proapoptotic, antiangiogenic, and collagen synthesis inhibitor in human leiomyomas cells (huLM). Objectives Our objectives were to investigate whether COMT expression, by the virtue of 2ME formation, affects the growth of huLM, and to explore the cellular and molecular mechanisms whereby COMT expression or treatment with 2ME affect these cells. Results Our data demonstrated that E2-induced proliferation was less pronounced in cells over-expressing COMT or treated with 2ME (500 nM). This effect on cell proliferation was associated with microtubules stabilization and diminution of estrogen receptor α (ERα) and progesterone receptor (PR) transcriptional activities, due to shifts in their subcellular localization and sequestration in the cytoplasm. In addition, COMT over expression or treatment with 2ME reduced the expression of hypoxia-inducible factor -1α (HIF-1 α) and the basal level as well as TNF-α-induced aromatase (CYP19) expression. Conclusions COMT over expression or treatment with 2ME stabilize microtubules, ameliorates E2-induced proliferation, inhibits ERα and PR signaling, and reduces HIF-1 α and CYP19 expression in human uterine leiomyoma cells. Thus, microtubules are a candidate target for treatment of uterine leiomyomas. In addition, the naturally occurring microtubule-targeting agent 2ME represents a potential new therapeutic for uterine leiomyomas.
Collapse
Affiliation(s)
- Salama A Salama
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, Texas, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Malik M, Mendoza M, Payson M, Catherino WH. Curcumin, a nutritional supplement with antineoplastic activity, enhances leiomyoma cell apoptosis and decreases fibronectin expression. Fertil Steril 2009; 91:2177-84. [DOI: 10.1016/j.fertnstert.2008.03.045] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2008] [Revised: 03/17/2008] [Accepted: 03/17/2008] [Indexed: 01/28/2023]
|
37
|
Ahmed N, Garcia G, Ali H, van Lier JE. (18)F-labelling of A-ring substituted 16alpha-fluoro-estradiols as potential radiopharmaceuticals for PET imaging. Steroids 2009; 74:42-50. [PMID: 18845173 DOI: 10.1016/j.steroids.2008.09.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2007] [Revised: 09/03/2008] [Accepted: 09/05/2008] [Indexed: 11/20/2022]
Abstract
The 2-methoxy derivative of estradiol is currently in Phase II clinical trial as an anticancer agent while the 4-methyl derivative has been shown to interact with cytoplasmic and nuclear estrogen receptors in rat pituitary gland and hypothalamus. We hypothesize that the 16alpha-(18)F-analogs of these estrogens could be suitable radiotracers to evaluate action mechanisms of the parent compounds. In this study we report the synthesis of the 16alpha-(18)F and 16alpha-(19)F-analogs of the A-ring substituted estradiols in high yield via stereoselective opening of the intermediate 16beta,17beta-O-cyclic sulfones with [(18)F]F(-) or F(-) followed by deprotection.
Collapse
Affiliation(s)
- Naseem Ahmed
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada J1H 5N4
| | | | | | | |
Collapse
|
38
|
de Oliveira E, de Aquino Castro R, Vieira Gomes MT, Cotrim Guerreiro da Silva ID, Baracat EC, Rodrigues de Lima G, Ferreira Sartori MG, Batista Castello Girão MJ. Role of glutathione S-transferase (GSTM1) gene polymorphism in development of uterine fibroids. Fertil Steril 2008; 91:1496-8. [PMID: 18829006 DOI: 10.1016/j.fertnstert.2008.07.1767] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2008] [Revised: 07/23/2008] [Accepted: 07/24/2008] [Indexed: 10/21/2022]
Abstract
Glutathione S-transferase (GSTM1) plays an important role in the excretion of catechol estrogens and is therefore a candidate marker for fibroids. However, this case-control study demonstrated no association between GSTM1 polymorphism and the risk of leiomyoma in Brazilian women.
Collapse
Affiliation(s)
- Emerson de Oliveira
- Department of Gynecology, Federal University of São Paulo, São Paulo, Brazil.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
LPS-induced MCP-1 and IL-6 production is not reversed by oestrogen in human periodontal ligament cells. Arch Oral Biol 2008; 53:896-902. [DOI: 10.1016/j.archoralbio.2008.05.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2007] [Revised: 03/25/2008] [Accepted: 05/01/2008] [Indexed: 11/18/2022]
|
40
|
Guzeloglu-Kayisli O, Halis G, Taskiran S, Kayisli UA, Arici A. DNA-binding ability of NF-kappaB is affected differently by ERalpha and ERbeta and its activation results in inhibition of estrogen responsiveness. Reprod Sci 2008; 15:493-505. [PMID: 18579858 DOI: 10.1177/1933719108317583] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Estrogenic effects involve interactions between estrogen receptors (ERs), response elements, and nuclear proteins. It is hypothesized that interaction between ER and NF-kappa B may affect the regulation of responsive genes. Electrophoretic mobility shift assay (EMSA) was performed to assess if the interaction of ERs and NF- kappaB affect their respective DNA-binding activities, and alkaline phosphatase assay was done to evaluate estrogenic activity. EMSA revealed that ERs inhibit DNA-binding of p50 and p65, whereas p50 did not impair ER alpha binding. Stimulation with estradiol inhibited DNA binding of NF-kappaB in ERalpha-transfected endometrial stromal cells (ESCs). Moreover, activation of NF-kappaB significantly decreased estrogen responsiveness of Ishikawa cells and ERalpha-transfected ESC. Our results suggest that ERs downregulate NF-kappaB-dependent gene activation by directly preventing DNA binding. However, NF-kappaB-mediated inhibition of ER-dependent gene activation may be carried out indirectly rather than through a direct inhibition of ER-DNA binding. These findings offer new insight into the specific role of ERalpha and could eventually help in developing therapeutics for endometriosis.
Collapse
Affiliation(s)
- Ozlem Guzeloglu-Kayisli
- Division of Reproductive Endocrinology, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut 06520-8063, USA
| | | | | | | | | |
Collapse
|
41
|
Grisaru D, Keidar R, Schreiber L, Lessing JB, Deutsch V. The effect of the readthrough acetylcholinesterase variant (AChE-R) on uterine muscle and leiomyomas. Mol Hum Reprod 2007; 13:351-4. [PMID: 17350961 DOI: 10.1093/molehr/gam010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Acetylcholine signaling and acetylcholinesterase (AChE) function(s) are pivotal elements in muscle development. The effects of the stimulus-dependent readthrough AChE variant, AChE-R, on leiomyomas and normal myometrium proliferation were assessed in vivo and in vitro. Histological preparations and cell cultures therefrom were obtained during hysterectomies or myomectomies and included both the leiomyoma sample and the adjacent normal uterine muscle as control. In situ hybridization procedures were performed using AChE cRNA probes complementary to the human AChE-R transcript. Antibodies against the AChE-R variant served for immunohistochemical staining. To determine the biological function of AChE-R on the uterine muscle cell cultures, we used a synthetic peptide representing the potentially cleavable morphogenically active C-terminus of AChE-R (ARP). Cell proliferation was assessed using the incorporation of 5'-bromo-2-deoxyuridine (BrDU). Leiomyomas expressed an excess of AChE-R mRNA and the AChE-R protein compared with the normal myometrium. Cell cultures originating from leiomyomas proliferated significantly faster than cultures from the adjacent myometrium (P = 0.027 at BrDU incorporation). Addition of ARP (2-200 nM) caused a dose-dependent decrease in the proliferation of cell cultures from both leiomyomas and the myometrium. The effect on the myometrium reached statistical significance (at 20 and 200 nM, P = 0.02), whereas the variability of the rapidly proliferating primary cultures was high and precluded statistical significance in the leiomyoma cultures. AChE-R is involved in the proliferation of the myometrium. The inhibitory effect of ARP on the myometrium may suggest a future therapeutic role of ARP.
Collapse
Affiliation(s)
- Dan Grisaru
- Department of Obstetrics-Gynecology, Tel Aviv University, Tel-Aviv, Israel.
| | | | | | | | | |
Collapse
|