1
|
Takahashi K, Takiwaki M, Fukuzawa S, Kikutani Y, Abe K, Higashi T, Kobayashi H. Development of an accurate and sensitive assay for 2-methoxyestradiol using derivatization and liquid chromatography-tandem mass spectrometry. Pract Lab Med 2025; 44:e00447. [PMID: 39882397 PMCID: PMC11772991 DOI: 10.1016/j.plabm.2024.e00447] [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: 11/06/2024] [Revised: 12/10/2024] [Accepted: 12/27/2024] [Indexed: 01/31/2025] Open
Abstract
2-Methoxyestradiol (2ME) is involved in the pathogenesis of preeclampsia and antitumor activity. In addition to its low concentration in healthy human serum, presence of isomers makes quantification of 2ME for clinical research and laboratory medicine difficult. The objective of this study was to develop a highly sensitive and accurate method for quantifying 2ME using LC-MS/MS combined with derivatization with 1-(2,4-dinitro-5-fluorophenyl)-4,4-dimethylpiperazinium iodide (MPDNP-F). This approach significantly enhanced the detectability of 2ME in positive electrospray ionization-tandem mass spectrometry (ESI-MS/MS) and enabled the chromatographic separation of 2ME from isomeric metabolites possessing a methoxy group, including 4-methoxyestradiol, 3-O-methyl 2-hydroxyestradiol, and 3-O-methyl 4-hydroxyestradiol (3M4OH). Although the derivatized 2ME and 3M4OH were closely eluted under the optimized LC conditions, the different fragmentation patterns of these isomers during MS/MS allowed their distinction. The lower limit of quantification for 2ME was 2.5 pg/mL, indicating a satisfactory sensitivity. These findings demonstrated that this LC-MS/MS method combined with the MPDNP-F derivatization can provide accurate and highly sensitive quantification of 2ME.
Collapse
Affiliation(s)
- Koji Takahashi
- Medical Equipment Business Operations, JEOL Ltd., Tokyo, Japan
| | - Masaki Takiwaki
- Medical Equipment Business Operations, JEOL Ltd., Tokyo, Japan
| | | | | | - Kentaro Abe
- Medical Equipment Business Operations, JEOL Ltd., Tokyo, Japan
| | - Tatsuya Higashi
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | | |
Collapse
|
2
|
Liu N, Sun Y, Liu J, Zhang Y, Yi X, Yan W, Cui X, Guo T, Zhao W, Han S, Ma W, Cao Y, Chen L. Nobiletin: a potential erythropoietin receptor activator protects renal cells against hypoxia. Apoptosis 2025; 30:842-860. [PMID: 39755823 DOI: 10.1007/s10495-024-02067-9] [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] [Accepted: 12/20/2024] [Indexed: 01/06/2025]
Abstract
Tangerine peel is a traditional Chinese herb and has been widely applied in foods and medicine for its multiple pharmacological effects. Erythropoietin receptor (EPOR), a member of the cytokine receptor family, is widely expressed in multiple tissues in especial kidney and plays protective effects in adverse physiological and pathological conditions. We hypothesized that it might be EPOR agonists existing in Tangerine peel bring such renal benefits. To test our hypothesis, an EPOR/cell membrane chromatography (CMC)-high performance liquid chromatography (HPLC)-mass spectrometry (MS) analytical system was developed to screen EPOR targeted compounds from tangerine peel extra out. A fraction was retained on the EPOR/CMC column, separated, and further identified as nobiletin. Frontal analysis, non-linear chromatography, and molecular docking assay were applied to determine the binding force and sites between nobiletin and EPOR. Intracellular Ca2+ mobilization, cAMP accumulation, and phosphorylation of JAK2 and STAT5 were determined to confirm the EPOR activation effect of nobiletin. CoCl2 was applied to construct a renal hypoxic cell model, and cell viability and apoptosis of human glomerular mesangial cells (HMC) were carried out to assess the pharmacological effect of nobiletin. Apoptosis-related proteins including Bcl-2, Bcl-xL, Bax, Cleaved caspase 3, caspase 3, caspase 9, and Cytochrome C were determined. SiRNA and lentivirus were used to silence or overexpress EPOR. Our results indicated that nobiletin is a potential EPOR agonist, reflected on its explicit binding force and downstream signal activating effects. Furthermore, EPOR-overexpressing enhanced the hypoxia-tolerance of renal cells. Our mechanism research indicated that the protective effect of nobiletin against hypoxia was depended on its pro-proliferation and anti-apoptosis effects. In conclusion, nobiletin, a potential small molecular agonist of EPOR, protects HMC against hypoxia through positively activating EPOR.
Collapse
Affiliation(s)
- Na Liu
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yuzhuo Sun
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jieyun Liu
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yangyang Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xinyao Yi
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Wenhui Yan
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xin Cui
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Tingli Guo
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Wenzhuo Zhao
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Shengli Han
- School of Pharmacy, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Weina Ma
- School of Pharmacy, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Yapeng Cao
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, 710061, China.
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China.
| | - Lina Chen
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710061, China.
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, 710061, China.
- Cardiometabolic Innovation Center, Ministry of Education, Xi'an, 710061, China.
| |
Collapse
|
3
|
Mercier AE, Joubert AM, Prudent R, Viallet J, Desroches-Castan A, De Koning L, Mabeta P, Helena J, Pepper MS, Lafanechère L. Sulfamoylated Estradiol Analogs Targeting the Actin and Microtubule Cytoskeletons Demonstrate Anti-Cancer Properties In Vitro and In Ovo. Cancers (Basel) 2024; 16:2941. [PMID: 39272798 PMCID: PMC11394244 DOI: 10.3390/cancers16172941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/30/2024] [Accepted: 08/14/2024] [Indexed: 09/15/2024] Open
Abstract
The microtubule-disrupting agent 2-methoxyestradiol (2-ME) displays anti-tumor and anti-angiogenic properties, but its clinical development is halted due to poor pharmacokinetics. We therefore designed two 2-ME analogs in silico-an ESE-15-one and an ESE-16 one-with improved pharmacological properties. We investigated the effects of these compounds on the cytoskeleton in vitro, and their anti-angiogenic and anti-metastatic properties in ovo. Time-lapse fluorescent microscopy revealed that sub-lethal doses of the compounds disrupted microtubule dynamics. Phalloidin fluorescent staining of treated cervical (HeLa), metastatic breast (MDA-MB-231) cancer, and human umbilical vein endothelial cells (HUVECs) displayed thickened, stabilized actin stress fibers after 2 h, which rearranged into a peripheral radial pattern by 24 h. Cofilin phosphorylation and phosphorylated ezrin/radixin/moesin complexes appeared to regulate this actin response. These signaling pathways overlap with anti-angiogenic, extra-cellular communication and adhesion pathways. Sub-lethal concentrations of the compounds retarded both cellular migration and invasion. Anti-angiogenic and extra-cellular matrix signaling was evident with TIMP2 and P-VEGF receptor-2 upregulation. ESE-15-one and ESE-16 exhibited anti-tumor and anti-metastatic properties in vivo, using the chick chorioallantoic membrane assay. In conclusion, the sulfamoylated 2-ME analogs displayed promising anti-tumor, anti-metastatic, and anti-angiogenic properties. Future studies will assess the compounds for myeloproliferative effects, as seen in clinical applications of other drugs in this class.
Collapse
Affiliation(s)
- Anne Elisabeth Mercier
- Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0028, South Africa
| | - Anna Margaretha Joubert
- Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0028, South Africa
| | - Renaud Prudent
- Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, 38000 Grenoble, France
| | - Jean Viallet
- Inovotion SAS France, Biopolis, 38700 La Tronche, France
| | - Agnes Desroches-Castan
- Laboratoire Biosanté U1292, Université Grenoble Alpes, Inserm, CEA, 38000 Grenoble, France
| | - Leanne De Koning
- Institut Curie Centre de Recherche, PSL Research University, 75248 Paris Cedex 05, France
| | - Peace Mabeta
- Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0028, South Africa
| | - Jolene Helena
- Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0028, South Africa
| | - Michael Sean Pepper
- Institute for Cellular and Molecular Medicine, Department of Immunology, and South African Medical Research Council Extramural Unit for Stem Cell Research and Therapy, School of Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0028, South Africa
| | - Laurence Lafanechère
- Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0028, South Africa
- Team Cytoskeleton Dynamics and Nuclear Functions, Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, 38000 Grenoble, France
| |
Collapse
|
4
|
Schaufelberger SA, Schaettin M, Azzarito G, Rosselli M, Leeners B, Dubey RK. 2-Methoxyestradiol, an Endogenous 17β-Estradiol Metabolite, Induces Antimitogenic and Apoptotic Actions in Oligodendroglial Precursor Cells and Triggers Endoreduplication via the p53 Pathway. Cells 2024; 13:1086. [PMID: 38994940 PMCID: PMC11240791 DOI: 10.3390/cells13131086] [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: 04/17/2024] [Revised: 06/14/2024] [Accepted: 06/20/2024] [Indexed: 07/13/2024] Open
Abstract
The abnormal growth of oligodendrocyte precursor cells (OPCs) significantly contributes to the progression of glioblastoma tumors. Hence, molecules that block OPC growth may be of therapeutic importance in treating gliomas. 2-Methoxyestradiol (2ME), an endogenous tubulin-interacting metabolite of estradiol, is effective against multiple proliferative disorders. Based on its anti-carcinogenic and anti-angiogenic actions, it is undergoing phase II clinical trials. We hypothesize that 2ME may prevent glioma growth by targeting OPC growth. Here, we tested this hypothesis by assessing the impact of 2ME on the growth of an OPC line, "Oli-neu", and dissected the underlying mechanism(s). Treatment with 2ME inhibited OPC growth in a concentration-dependent manner, accompanied by significant upregulation in the expression of p21 and p27, which are negative cell-cycle regulators. Moreover, treatment with 2ME altered OPC morphology from multi-arm processes to rounded cells. At concentrations of 1uM and greater, 2ME induced apoptosis, with increased expressions of caspase 3, PARP, and caspase-7 fragments, externalized phosphatidylserine staining/APOPercentage, and increased mitochondrial activity. Flow cytometry and microscopic analysis demonstrated that 2ME triggers endoreduplication in a concentration-dependent fashion. Importantly, 2ME induced cyclin E, JNK1/2, and p53 expression, as well as OPC fusion, which are key mechanisms driving endoreduplication and whole-genome duplication. Importantly, the inhibition of p53 with pifithrin-α rescued 2ME-induced endoreduplication. The pro-apoptotic and endoreduplication actions of 2ME were accompanied by the upregulation of survivin, cyclin A, Cyclin B, Cyclin D2, and ppRB. Similar growth inhibitory, apoptotic, and endoreduplication effects of 2ME were observed in CG4 cells. Taken together, our findings provide evidence that 2ME not only inhibits OPC growth and triggers apoptosis, but also activates OPCs into survival (fight or flight) mode, leading to endoreduplication. This inherent survival characteristic of OPCs may, in part, be responsible for drug resistance in gliomas, as observed for many tubulin-interacting drugs. Importantly, the fate of OPCs after 2ME treatment may depend on the cell-cycle status of individual cells. Combining tubulin-interfering molecules with drugs such as pifithrin-α that inhibit endoreduplication may help inhibit OPC/glioma growth and limit drug resistance.
Collapse
Affiliation(s)
- Sara. A. Schaufelberger
- Department of Obstetrics and Gynaecology, Clinic for Reproductive Endocrinology, University Hospital Zurich, 8091 Zurich, Switzerland (G.A.); (M.R.); (B.L.)
| | - Martina Schaettin
- Department of Obstetrics and Gynaecology, Clinic for Reproductive Endocrinology, University Hospital Zurich, 8091 Zurich, Switzerland (G.A.); (M.R.); (B.L.)
| | - Giovanna Azzarito
- Department of Obstetrics and Gynaecology, Clinic for Reproductive Endocrinology, University Hospital Zurich, 8091 Zurich, Switzerland (G.A.); (M.R.); (B.L.)
| | - Marinella Rosselli
- Department of Obstetrics and Gynaecology, Clinic for Reproductive Endocrinology, University Hospital Zurich, 8091 Zurich, Switzerland (G.A.); (M.R.); (B.L.)
| | - Brigitte Leeners
- Department of Obstetrics and Gynaecology, Clinic for Reproductive Endocrinology, University Hospital Zurich, 8091 Zurich, Switzerland (G.A.); (M.R.); (B.L.)
| | - Raghvendra K. Dubey
- Department of Obstetrics and Gynaecology, Clinic for Reproductive Endocrinology, University Hospital Zurich, 8091 Zurich, Switzerland (G.A.); (M.R.); (B.L.)
- Zurich Centre for Integrative Human Physiology (ZIHP), University of Zurich, 8006 Zurich, Switzerland
| |
Collapse
|
5
|
Zacharia LC, Eleftheriou C, Gkretsi V. Effects of 2-methoxyestradiol on hydrogen peroxide induced neuronal cell death and tau hyperphosphorylation. Life Sci 2022; 309:121047. [DOI: 10.1016/j.lfs.2022.121047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 09/25/2022] [Accepted: 10/02/2022] [Indexed: 11/29/2022]
|
6
|
Hargrave SD, Joubert AM, Potter BVL, Dohle W, Marais S, Mercier AE. Cell Fate following Irradiation of MDA-MB-231 and MCF-7 Breast Cancer Cells Pre-Exposed to the Tetrahydroisoquinoline Sulfamate Microtubule Disruptor STX3451. Molecules 2022; 27:3819. [PMID: 35744942 PMCID: PMC9228122 DOI: 10.3390/molecules27123819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/26/2022] [Accepted: 05/30/2022] [Indexed: 11/16/2022] Open
Abstract
A tetrahydroisoquinoline (THIQ) core is able to mimic the A and B rings of 2-methoxyestradiol (2ME2), an endogenous estrogen metabolite that demonstrates promising anticancer properties primarily by disrupting microtubule dynamic instability parameters, but has very poor pharmaceutical properties that can be improved by sulfamoylation. The non-steroidal THIQ-based microtubule disruptor 2-(3-bromo-4,5-dimethoxybenzyl)-7-methoxy-6-sulfamoyloxy-1,2,3,4-tetrahydroisoquinoline (STX3451), with enhanced pharmacokinetic and pharmacodynamic profiles, was explored for the first time in radiation biology. We investigated whether 24 h pre-treatment with STX3451 could pre-sensitize MCF-7 and MDA-MB-231 breast cancer cells to radiation. This regimen showed a clear increase in cytotoxicity compared to the individual modalities, results that were contiguous in spectrophotometric analysis, flow cytometric quantification of apoptosis induction, clonogenic studies and microscopy techniques. Drug pre-treatment increased radiation-induced DNA damage, with statistically more double-strand (ds) DNA breaks demonstrated. The latter could be due to the induction of a radiation-sensitive metaphase block or the increased levels of reactive oxygen species, both evident after compound exposure. STX3451 pre-exposure may also delay DNA repair mechanisms, as the DNA damage response element ataxia telangiectasia mutated (ATM) was depressed. These in vitro findings may translate into in vivo models, with the ultimate aim of reducing both radiation and drug doses for maximal clinical effect with minimal adverse effects.
Collapse
Affiliation(s)
- Scott D. Hargrave
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (S.D.H.); (A.M.J.); (S.M.)
| | - Anna M. Joubert
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (S.D.H.); (A.M.J.); (S.M.)
| | - Barry V. L. Potter
- Medicinal Chemistry & Drug Discovery, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK; (B.V.L.P.); (W.D.)
| | - Wolfgang Dohle
- Medicinal Chemistry & Drug Discovery, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK; (B.V.L.P.); (W.D.)
| | - Sumari Marais
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (S.D.H.); (A.M.J.); (S.M.)
| | - Anne E. Mercier
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (S.D.H.); (A.M.J.); (S.M.)
| |
Collapse
|
7
|
Zhang Y, Ogola BO, Iyer L, Karamyan VT, Thekkumkara T. Estrogen Metabolite 2-Methoxyestradiol Attenuates Blood Pressure in Hypertensive Rats by Downregulating Angiotensin Type 1 Receptor. Front Physiol 2022; 13:876777. [PMID: 35586713 PMCID: PMC9108484 DOI: 10.3389/fphys.2022.876777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/18/2022] [Indexed: 11/13/2022] Open
Abstract
The therapeutic potential of 2-Methoxyestradiol (2ME2) is evident in cardiovascular disease. Our laboratory has previously demonstrated the mechanism involved in the 2ME2 regulation of angiotensin type 1 receptor (AT1R) in vitro. However, 2ME2 regulation of angiotensin receptors and its effects on blood pressure (BP) and resting heart rate (RHR) are uncertain. In this study, male and female Wistar-Kyoto (WKY) rats infused with angiotensin II (65 ng/min) and male spontaneously hypertensive rats (SHR) were surgically implanted with telemetric probes to continuously assess arterial BP and RHR. In both male and female WKY rats, 2ME2 treatment (20 mg/kg/day for 2 weeks) resulted in a significant reduction of Ang II-induced systolic, diastolic, and mean arterial BP. Moreover, significant weight loss and RHR were indicated in all groups. In a separate set of experiments, prolonged 2ME2 exposure in male SHR (20 mg/kg/day for 5 weeks) displayed a significant reduction in diastolic and mean arterial BP along with RHR. We also found downregulation of angiotensin receptors and angiotensinogen (AGT) in the kidney and liver and a reduction of plasma Ang II levels. Collectively, we demonstrate that 2ME2 attenuated BP and RHR in hypertensive rats involves downregulation of angiotensin receptors and body weight loss.
Collapse
Affiliation(s)
- Yong Zhang
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center School of Pharmacy, Amarillo, TX, United States
| | - Benard O. Ogola
- Department of Pharmacology, Tulane University, New Orleans, LA, United States
- *Correspondence: Benard O. Ogola,
| | - Laxmi Iyer
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center School of Pharmacy, Amarillo, TX, United States
| | - Vardan T. Karamyan
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center School of Pharmacy, Amarillo, TX, United States
| | - Thomas Thekkumkara
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center School of Pharmacy, Amarillo, TX, United States
| |
Collapse
|
8
|
Samargandy S, Matthews KA, Brooks MM, Barinas-Mitchell E, Magnani JW, Thurston RC, El Khoudary SR. Trajectories of Blood Pressure in Midlife Women: Does Menopause Matter? Circ Res 2022; 130:312-322. [PMID: 35113663 PMCID: PMC8814466 DOI: 10.1161/circresaha.121.319424] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Whether changes in blood pressure (BP) over women's midlife are more driven by chronological aging or the menopause transition has been debated. We sought to determine whether women can be classified into distinct trajectory groups based on pattern and level of systolic BP (SBP), diastolic BP, pulse pressure (PP), and mean arterial pressure (MAP) over the menopause transition, and to assess whether menopause-related factors predict the group and level of BP measures. METHODS Participants were from the SWAN (Study of Women's Health Across the Nation). Group-based trajectory modeling was used to identify women who shared distinct BP trajectories over time relative to menopause onset and to assess associations of menopause-related factors with trajectory group and level of BP measures. An accelerated rise relative to menopause onset suggests a menopause contribution. RESULTS The study included 3302 multiracial and multiethnic women with BP measures over 17 follow-up visits (baseline age [SD]: 46.3 [2.7]). Women were classified into either low, medium, or high trajectory group in each BP measure. The low SBP, PP, and MAP trajectories (in 35%, 53%, and 28% of the cohort, respectively) were rising slowly before menopause but showed a significant accelerated rise 1 year after menopause, indicating a menopause contribution. The remaining BP trajectories were rising up until menopause and either continued with the same rise or declined after menopause. A younger menopause age predicted the low SBP, PP, and MAP trajectories. A greater follicle-stimulating hormone level predicted lower SBP and PP levels, while vasomotor symptoms occurrence predicted higher SBP, PP, and MAP levels over time. Estradiol did not predict trajectory or level of any BP measure. CONCLUSIONS Distinct BP trajectories over the menopause transition exist that revealed a group of women whose SBP, PP, and MAP trajectories are consistent with a menopause contribution. Our findings support frequent monitoring of BP during the menopause transition.
Collapse
Affiliation(s)
- Saad Samargandy
- Department of Community Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Maria M. Brooks
- Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA
| | | | - Jared W. Magnani
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | | | | |
Collapse
|
9
|
Ibata T, Lyu J, Imachi H, Fukunaga K, Sato S, Kobayashi T, Saheki T, Yoshimura T, Murao K. Effects of 2-Methoxyestradiol, a Main Metabolite of Estradiol on Hepatic ABCA1 Expression in HepG2 Cells. Nutrients 2022; 14:nu14020288. [PMID: 35057469 PMCID: PMC8779252 DOI: 10.3390/nu14020288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 02/04/2023] Open
Abstract
ATP-binding cassette transporter A1 (ABCA1) is a key regulator of lipid efflux, and the absence of ABCA1 induces hepatic lipid accumulation, which is one of the major causes of fatty liver. 2-Methoxyestradiol (2-ME2) has been demonstrated to protect against fatty liver. In this study, we investigated the effects of 2-ME2 on the hepatic lipid content and ABCA1 expression. We found that 2-ME2 dose-dependently increased ABCA1 expression, and therefore, the lipid content was significantly decreased in HepG2 cells. 2-ME2 enhanced the ABCA1 promoter activity; however, this effect was reduced after the inhibition of the PI3K pathway. The overexpression of Akt or p110 induced ABCA1 promoter activity, while dominant-negative Akt diminished the ability of 2-ME2 on ABCA1 promoter activity. Further, 2-ME2 stimulated the rapid phosphorylation of Akt and FoxO1 and reduced the nuclear accumulation of FoxO1. Chromatin immunoprecipitation confirmed that FoxO1 bonded to the ABCA1 promoter region. The binding was reduced by 2-ME2, which facilitated ABCA1 gene transcription. Furthermore, mutating FoxO1-binding sites in the ABCA1 promoter region or treatment with FoxO1-specific siRNA disrupted the effect of 2-ME2 on ABCA1 expression. All of our results demonstrated that 2-ME2 might upregulate ABCA1 expression via the PI3K/Akt/FoxO1 pathway, which thus reduces the lipid content in hepatocytes.
Collapse
Affiliation(s)
- Tomohiro Ibata
- Department of Endocrinology and Metabolism, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho 761-0793, Kagawa, Japan; (T.I.); (J.L.); (H.I.); (K.F.); (S.S.); (T.K.); (T.S.); (T.Y.)
| | - Jingya Lyu
- Department of Endocrinology and Metabolism, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho 761-0793, Kagawa, Japan; (T.I.); (J.L.); (H.I.); (K.F.); (S.S.); (T.K.); (T.S.); (T.Y.)
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou 510632, China
| | - Hitomi Imachi
- Department of Endocrinology and Metabolism, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho 761-0793, Kagawa, Japan; (T.I.); (J.L.); (H.I.); (K.F.); (S.S.); (T.K.); (T.S.); (T.Y.)
| | - Kensaku Fukunaga
- Department of Endocrinology and Metabolism, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho 761-0793, Kagawa, Japan; (T.I.); (J.L.); (H.I.); (K.F.); (S.S.); (T.K.); (T.S.); (T.Y.)
| | - Seisuke Sato
- Department of Endocrinology and Metabolism, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho 761-0793, Kagawa, Japan; (T.I.); (J.L.); (H.I.); (K.F.); (S.S.); (T.K.); (T.S.); (T.Y.)
| | - Toshihiro Kobayashi
- Department of Endocrinology and Metabolism, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho 761-0793, Kagawa, Japan; (T.I.); (J.L.); (H.I.); (K.F.); (S.S.); (T.K.); (T.S.); (T.Y.)
| | - Takanobu Saheki
- Department of Endocrinology and Metabolism, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho 761-0793, Kagawa, Japan; (T.I.); (J.L.); (H.I.); (K.F.); (S.S.); (T.K.); (T.S.); (T.Y.)
| | - Takafumi Yoshimura
- Department of Endocrinology and Metabolism, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho 761-0793, Kagawa, Japan; (T.I.); (J.L.); (H.I.); (K.F.); (S.S.); (T.K.); (T.S.); (T.Y.)
| | - Koji Murao
- Department of Endocrinology and Metabolism, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho 761-0793, Kagawa, Japan; (T.I.); (J.L.); (H.I.); (K.F.); (S.S.); (T.K.); (T.S.); (T.Y.)
- Correspondence:
| |
Collapse
|
10
|
Song CY, Singh P, Motiwala M, Shin JS, Lew J, Dutta SR, Gonzalez FJ, Bonventre JV, Malik KU. 2-Methoxyestradiol Ameliorates Angiotensin II-Induced Hypertension by Inhibiting Cytosolic Phospholipase A 2α Activity in Female Mice. Hypertension 2021; 78:1368-1381. [PMID: 34628937 PMCID: PMC8516072 DOI: 10.1161/hypertensionaha.121.18181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Supplemental Digital Content is available in the text. We tested the hypothesis that CYP1B1 (cytochrome P450 1B1)-17β-estradiol metabolite 2-methoxyestradiol protects against Ang II (angiotensin II)–induced hypertension by inhibiting group IV cPLA2α (cytosolic phospholipase A2α) activity and production of prohypertensive eicosanoids in female mice. Ang II (700 ng/kg per minute, SC) increased mean arterial blood pressure (BP), systolic and diastolic BP measured by radiotelemetry, renal fibrosis, and reactive oxygen species production in wild-type mice (cPLA2α+/+/Cyp1b1+/+) that were enhanced by ovariectomy and abolished in intact and ovariectomized-cPLA2α−/−/Cyp1b1+/+ mice. Ang II–induced increase in SBP measured by tail-cuff, renal fibrosis, reactive oxygen species production, and cPLA2α activity measured by its phosphorylation in the kidney, and urinary excretion of prostaglandin E2 and thromboxane A2 metabolites were enhanced in ovariectomized-cPLA2α+/+/Cyp1b1+/+ and intact cPLA2α+/+/Cyp1b1−/− mice. 2-Methoxyestradiol and arachidonic acid metabolism inhibitor 5,8,11,14-eicosatetraynoic acid attenuated the Ang II–induced increase in SBP, renal fibrosis, reactive oxygen species production, and urinary excretion of prostaglandin E2, and thromboxane A2 metabolites in ovariectomized-cPLA2α+/+/Cyp1b1+/+ and intact cPLA2α+/+/Cyp1b1−/− mice. Antagonists of prostaglandin E2 and thromboxane A2 receptors EP1 and EP3 and TP, respectively, inhibited Ang II–induced increases in SBP and reactive oxygen species production and renal fibrosis in ovariectomized-cPLA2α+/+/Cyp1b1+/+ and intact cPLA2α+/+/Cyp1b1−/− mice. These data suggest that CYP1B1-generated metabolite 2-methoxyestradiol mitigates Ang II–induced hypertension and renal fibrosis by inhibiting cPLA2α activity, reducing prostaglandin E2, and thromboxane A2 production and stimulating EP1 and EP3 and TP receptors, respectively. Thus, 2-methoxyestradiol and the drugs that selectively block EP1 and EP3 and TP receptors could be useful in treating hypertension and its pathogenesis in females.
Collapse
Affiliation(s)
- Chi Young Song
- From the Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee HSC, Memphis (C.Y.S., P.S., M.M., J.S.S., J.L., S.R.D., K.U.M.)
| | - Purnima Singh
- From the Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee HSC, Memphis (C.Y.S., P.S., M.M., J.S.S., J.L., S.R.D., K.U.M.)
| | - Mustafa Motiwala
- From the Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee HSC, Memphis (C.Y.S., P.S., M.M., J.S.S., J.L., S.R.D., K.U.M.)
| | - Ji Soo Shin
- From the Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee HSC, Memphis (C.Y.S., P.S., M.M., J.S.S., J.L., S.R.D., K.U.M.)
| | - Jessica Lew
- From the Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee HSC, Memphis (C.Y.S., P.S., M.M., J.S.S., J.L., S.R.D., K.U.M.)
| | - Shubha R. Dutta
- From the Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee HSC, Memphis (C.Y.S., P.S., M.M., J.S.S., J.L., S.R.D., K.U.M.)
| | - Frank J. Gonzalez
- Laboratory of Metabolism, National Cancer Institute, Bethesda, MD (F.J.G.)
| | - Joseph V. Bonventre
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA (J.V.B.)
| | - Kafait U. Malik
- From the Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee HSC, Memphis (C.Y.S., P.S., M.M., J.S.S., J.L., S.R.D., K.U.M.)
| |
Collapse
|
11
|
Jackson EK, Gillespie DG, Tofovic SP. DPP4 Inhibition, NPY 1-36, PYY 1-36, SDF-1 α, and a Hypertensive Genetic Background Conspire to Augment Cell Proliferation and Collagen Production: Effects That Are Abolished by Low Concentrations of 2-Methoxyestradiol. J Pharmacol Exp Ther 2020; 373:135-148. [PMID: 32015161 PMCID: PMC7174788 DOI: 10.1124/jpet.119.263467] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 01/30/2020] [Indexed: 12/13/2022] Open
Abstract
By reducing their metabolism, dipeptidyl peptidase 4 inhibition (DPP4I) enhances the effects of numerous peptides including neuropeptide Y1-36 (NPY1-36), peptide YY1-36 (PYY1-36), and SDF-1α Studies show that separately NPY1-36, PYY1-36 and SDF-1α stimulate proliferation of, and collagen production by, cardiac fibroblasts (CFs), preglomerular vascular smooth muscle cells (PGVSMCs), and glomerular mesangial cells (GMCs), particularly in cells isolated from genetically hypertensive rats. Whether certain combinations of these factors, in the absence or presence of DPP4I, are more profibrotic than others is unknown. Here we contrasted 24 different combinations of conditions (DPP4I, hypertensive genotype and physiologic levels [3 nM] of NPY1-36, PYY1-36, or SDF-1α) on proliferation of, and [3H]-proline incorporation by, CFs, PGVSMCs, and GMCs. In all three cell types, the various treatment conditions differentially increased proliferation and [3H]-proline incorporation, with a hypertensive genotype + DPP4I + NPY1-36 + SDF-1α being the most efficacious combination. Although the effects of this four-way combination were similar in male versus female CFs, physiologic (1 nM) concentrations of 2-methoxyestradiol (2ME; nonestrogenic metabolite of 17β-estradiol), abolished the effects of this combination in both male and female CFs. In conclusion, this study demonstrates that CFs, PGVSMCs, and GMCs are differentially activated by various combinations of NPY1-36, PYY1-36, SDF-1α, a hypertensive genetic background and DPP4I. We hypothesize that as these progrowth conditions accumulate, a tipping point would be reached that manifests in the long term as organ fibrosis and that 2ME would obviate any profibrotic effects of DPP4I, even under the most profibrotic conditions (i.e., hypertensive genotype with high NPY1-36 + SDF-1α levels and low 2ME levels). SIGNIFICANCE STATEMENT: This work elucidates combinations of factors that could contribute to long-term profibrotic effects of dipeptidyl peptidase 4 inhibitors and suggests a novel drug combination that could prevent any potential profibrotic effects of dipeptidyl peptidase 4 inhibitors while augmenting the protective effects of this class of antidiabetic agents.
Collapse
Affiliation(s)
- Edwin K Jackson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Delbert G Gillespie
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Stevan P Tofovic
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
12
|
Docherty CK, Nilsen M, MacLean MR. Influence of 2-Methoxyestradiol and Sex on Hypoxia-Induced Pulmonary Hypertension and Hypoxia-Inducible Factor-1-α. J Am Heart Assoc 2019; 8:e011628. [PMID: 30819028 PMCID: PMC6474940 DOI: 10.1161/jaha.118.011628] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 01/18/2019] [Indexed: 12/14/2022]
Abstract
Background Women are at greater risk of developing pulmonary arterial hypertension, with estrogen and its downstream metabolites playing a potential role in the pathogenesis of the disease. Hypoxia-inducible factor-1-α (HIF 1α) is a pro-proliferative mediator and may be involved in the development of human pulmonary arterial hypertension . The estrogen metabolite 2-methoxyestradiol (2 ME 2) has antiproliferative properties and is also an inhibitor of HIF 1α. Here, we examine sex differences in HIF 1α signaling in the rat and human pulmonary circulation and determine if 2 ME 2 can inhibit HIF 1α in vivo and in vitro. Methods and Results HIF 1α signaling was assessed in male and female distal human pulmonary artery smooth muscle cells ( hPASMC s), and the effects of 2 ME 2 were also studied in female hPASMC s. The in vivo effects of 2 ME 2 in the chronic hypoxic rat (male and female) model of pulmonary hypertension were also determined. Basal HIF 1α protein expression was higher in female hPASMC s compared with male. Both factor-inhibiting HIF and prolyl hydroxylase-2 (hydroxylates HIF leading to proteosomal degradation) protein levels were significantly lower in female hPASMC s when compared with males. In vivo, 2 ME 2 ablated hypoxia-induced pulmonary hypertension in male and female rats while decreasing protein expression of HIF 1α. 2 ME 2 reduced proliferation in hPASMC s and reduced basal protein expression of HIF 1α. Furthermore, 2 ME 2 caused apoptosis and significant disruption to the microtubule network. Conclusions Higher basal HIF 1α in female hPASMC s may increase susceptibility to developing pulmonary arterial hypertension . These data also demonstrate that the antiproliferative and therapeutic effects of 2 ME 2 in pulmonary hypertension may involve inhibition of HIF 1α and/or microtubular disruption in PASMC s.
Collapse
MESH Headings
- 2-Methoxyestradiol/pharmacology
- Animals
- Apoptosis/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Cytoskeleton/drug effects
- Cytoskeleton/metabolism
- Cytoskeleton/pathology
- Disease Models, Animal
- Female
- Humans
- Hypoxia/complications
- Hypoxia/metabolism
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Pulmonary Arterial Hypertension/drug therapy
- Pulmonary Arterial Hypertension/etiology
- Pulmonary Arterial Hypertension/metabolism
- Pulmonary Arterial Hypertension/pathology
- Pulmonary Artery/drug effects
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Rats, Sprague-Dawley
- Sex Factors
- Signal Transduction/drug effects
- Vascular Remodeling/drug effects
Collapse
Affiliation(s)
- Craig K. Docherty
- Research Institute of Cardiovascular and Medical SciencesCollege of Medical, Veterinary and Life SciencesUniversity of GlasgowUnited Kingdom
| | - Margaret Nilsen
- Research Institute of Cardiovascular and Medical SciencesCollege of Medical, Veterinary and Life SciencesUniversity of GlasgowUnited Kingdom
| | - Margaret R. MacLean
- Research Institute of Cardiovascular and Medical SciencesCollege of Medical, Veterinary and Life SciencesUniversity of GlasgowUnited Kingdom
| |
Collapse
|
13
|
2-Methoxyestradiol attenuates liver fibrosis in mice: implications for M2 macrophages. Naunyn Schmiedebergs Arch Pharmacol 2018; 392:381-391. [PMID: 30535572 DOI: 10.1007/s00210-018-1577-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 10/31/2018] [Indexed: 02/06/2023]
Abstract
Liver fibrosis is a major health problem worldwide due to its serious complications including cirrhosis and liver cancer. 2-Methoxyestradiol (2-ME) is an end metabolite of estradiol with anti-proliferative, antioxidant, and anti-inflammatory activities. However, the protective role of 2-ME in liver fibrosis has not been fully investigated. The aim of this study was to determine the protective effect of 2-ME in carbon tetrachloride (CCl4)-induced liver fibrosis in mice. Animals were injected intraperitoneally with CCl4 twice weekly for 6 weeks. 2-ME 50 mg/kg or 100 mg/kg was administrated intraperitoneally every day over the same period. Our data showed that 2-ME reduced the extent of liver toxicity and fibrosis due to CCl4 exposure. It restored the elevated serum liver enzymes aspartate aminotransferase (AST), alanine aminotransferase (ALT), and alkaline phosphatase (ALP) levels and ameliorated oxidative status. In addition, 2-ME significantly reduced collagen deposition and alpha-smooth muscle actin (α-SMA) protein expressions. Furthermore, 2-ME markedly lowered macrophage infiltration and macrophage alternative activation marker chitinase-like molecules (CHI3L3/YM1). The results of this study indicate an important protective activity of 2-ME in liver fibrosis and highlight the role of macrophage recruitment and alternative activation as a possible target.
Collapse
|
14
|
Tripathi V, Jaiswar SP, Deo S, Shankhwar P. Association of 2-Methoxyestradiol (2ME) Plasma Levels with Clinical Severity Indices and Biomarkers of Preeclampsia. J Obstet Gynaecol India 2018; 69:122-127. [PMID: 31686744 DOI: 10.1007/s13224-018-1154-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Accepted: 07/02/2018] [Indexed: 11/29/2022] Open
Abstract
Introduction Preeclampsia is an important cause of maternal and perinatal morbidity and mortality. The pathophysiology of preeclampsia has been extensively studied. 2ME seems to maintain placental homeostasis necessary for appropriate cytotrophoblast invasion of the maternal deciduas. We plan this study to compare its value in the plasma of normotensive and preeclamptic women and studying its correlation with the severity indices of preeclampsia. Aim and Objectives The aim of this study was to estimate plasma levels of 2ME in preeclamptic and normotensive pregnant women and correlate it with the severity in terms of clinical indices, laboratory investigations, and fetomaternal outcome. Materials and Methods This study was a case-control study conducted in KGMU, Lucknow, August 2015-2016. Sample size was 90 pregnant women including 30 normotensive, 30 non-severe preeclampsia, and 30 severe preeclampsia patients. Plasma 2ME levels were analyzed using ELISA kit and the outcome was compared in terms of systolic and diastolic blood pressure, proteinuria, liver and renal function tests, serum LDH, uric acid, plasma 2ME levels, and fetomaternal outcome. Results There was a significant (p = 0.0001) difference in 2ME plasma levels among the groups (normotensive, non severe and severe preeclampsia patients). 2ME plasma levels significantly negatively correlated with SBP (r = - 0.62, p = 0.0001) and DBP (r = - 0.63, p = 0.0001). With one-unit increase in SBP, 2ME will decrease by 2445.14 units, and in one-unit increase in DBP, 2ME will decrease by 1807.28 units. The sensitivity and specificity of 2ME for preeclampsia were found to be high. Conclusion It was seen that there was a statistically significant difference between plasma 2ME levels among the three groups. The ROC curve obtained showed that sensitivity was predicted to be 98.3%, specificity 76.7%, positive predictive value of 89.4%, negative predictive value of 95.8% and accuracy of 91.1%.
Collapse
Affiliation(s)
- Vartika Tripathi
- Department of Obstetrics and Gynecology, King George Medical University, Lucknow, India
| | - S P Jaiswar
- Department of Obstetrics and Gynecology, King George Medical University, Lucknow, India
| | - Sujata Deo
- Department of Obstetrics and Gynecology, King George Medical University, Lucknow, India
| | - Pushplata Shankhwar
- Department of Obstetrics and Gynecology, King George Medical University, Lucknow, India
| |
Collapse
|
15
|
Novel p-carborane-containing multitarget anticancer agents inspired by the metabolism of 17β-estradiol. Bioorg Med Chem 2017; 25:6371-6378. [DOI: 10.1016/j.bmc.2017.10.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 10/10/2017] [Indexed: 12/11/2022]
|
16
|
Raghava N, Das BC, Ray SK. Neuroprotective effects of estrogen in CNS injuries: insights from animal models. ACTA ACUST UNITED AC 2017; 6:15-29. [PMID: 28845391 PMCID: PMC5567743 DOI: 10.2147/nan.s105134] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Among the estrogens that are biosynthesized in the human body, 17β-estradiol (estradiol or E2) is the most common and the best estrogen for neuroprotection in animal models of the central nervous system (CNS) injuries such as spinal cord injury (SCI), traumatic brain injury (TBI), and ischemic brain injury (IBI). These CNS injuries are not only serious health problems, but also enormous economic burden on the patients, their families, and the society at large. Studies from animal models of these CNS injuries provide insights into the multiple neuroprotective mechanisms of E2 and also suggest the possibility of translating the therapeutic efficacy of E2 in the treatment SCI, TBI, and IBI in humans in the near future. The pathophysiology of these injuries includes loss of motor function in the limbs, arms and their extremities, cognitive deficit, and many other serious consequences including life-threatening paralysis, infection, and even death. The potential application of E2 therapy to treat the CNS injuries may become a trend as the results are showing significant therapeutic benefits of E2 for neuroprotection when administered into the animal models of SCI, TBI, and IBI. This article describes the plausible mechanisms how E2 works with or without the involvement of estrogen receptors and provides an overview of the known neuroprotective effects of E2 in these three CNS injuries in different animal models. Because activation of estrogen receptors has profound implications in maintaining and also affecting normal physiology, there are notable impediments in translating E2 therapy to the clinics for neuroprotection in CNS injuries in humans. While E2 may not yet be the sole molecule for the treatment of CNS injuries due to the controversies surrounding it, the neuroprotective effects of its metabolite and derivative or combination of E2 with another therapeutic agent are showing significant impacts in animal models that can potentially shape the new treatment strategies for these CNS injuries in humans.
Collapse
Affiliation(s)
- Narayan Raghava
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Bhaskar C Das
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Swapan K Ray
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, USA
| |
Collapse
|
17
|
Affiliation(s)
- Raghvendra K. Dubey
- From the Department of Reproductive Endocrinology, University Hospital Zurich, Switzerland; Zurich Center for Integrative Human Physiology, University of Zurich, Switzerland; and Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, PA
| |
Collapse
|
18
|
Said AMA, Zaki RGE, Salah Eldin RA, Nasr M, Azab SS, Elzankalony YA. Efficacy of Intravitreal injection of 2-Methoxyestradiol in regression of neovascularization of a retinopathy of prematurity rat model. BMC Ophthalmol 2017; 17:38. [PMID: 28376733 PMCID: PMC5379748 DOI: 10.1186/s12886-017-0433-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 03/31/2017] [Indexed: 12/04/2022] Open
Abstract
Background Retinopathy of prematurity (ROP) is one of the targets for early detection and treatment to prevent childhood blindness in world health organization programs. The purpose of study was to evaluate the efficacy of intravitreal injection of 2-Methoxyestradiol (2-ME) nanoemulsion in regressing neovascularization of a ROP rat model. Methods A prospective comparative case - control animal study conducted on 56 eyes of 28 healthy new born Sprague Dawley male albino rat. ROP was induced in 21 rats then two concentrations of 2-ME nanoparticles were injected in right eyes of 14 rats (low dose; study group I, high dose; study group II). A blank nanoemulsion was injected in the right eyes of seven rats (control positive group I). No injections performed in contralateral left eyes (control positive group II). Seven rats (14 eyes) were kept in room air (control negative group). On postnatal day 17, eyeballs were enucleated. Histological structure of the retina was examined using Hematoxylin and eosin staining. Vascular endothelial growth factor (VEGF) and glial fibrillary acidic protein (GFAP) expressions were detected by immunohistochemical studies. Results Intravitreal injection of 2-ME (in the two concentrations) caused marked regression of the new vascular tufts on the vitreal side with normal organization and thickness of the retina especially in study group II, which also show negative VEGF immunoreaction. Positive GFAP expression was detected in the control positive groups and study group (I). Conclusion Intravitreal injection of 2-Methoxyestradiol nanoemulsion is a promising effective method in reduction of neovascularization of a ROP rat model. Electronic supplementary material The online version of this article (doi:10.1186/s12886-017-0433-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Azza Mohamed Ahmed Said
- Ophthalmology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt. .,, 10th Fawzy Elmoteay Street, Heliopolis, Cairo, 11361, Egypt.
| | | | - Rania A Salah Eldin
- Anatomy and Embryology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Maha Nasr
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Samar Saad Azab
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | | |
Collapse
|
19
|
Berkane N, Liere P, Oudinet JP, Hertig A, Lefèvre G, Pluchino N, Schumacher M, Chabbert-Buffet N. From Pregnancy to Preeclampsia: A Key Role for Estrogens. Endocr Rev 2017; 38:123-144. [PMID: 28323944 DOI: 10.1210/er.2016-1065] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 02/28/2017] [Indexed: 02/08/2023]
Abstract
Preeclampsia (PE) results in placental dysfunction and is one of the primary causes of maternal and fetal mortality and morbidity. During pregnancy, estrogen is produced primarily in the placenta by conversion of androgen precursors originating from maternal and fetal adrenal glands. These processes lead to increased plasma estrogen concentrations compared with levels in nonpregnant women. Aberrant production of estrogens could play a key role in PE symptoms because they are exclusively produced by the placenta and they promote angiogenesis and vasodilation. Previous assessments of estrogen synthesis during PE yielded conflicting results, possibly because of the lack of specificity of the assays. However, with the introduction of reliable analytical protocols using liquid chromatography/mass spectrometry or gas chromatography/mass spectrometry, more recent studies suggest a marked decrease in estradiol levels in PE. The aim of this review is to summarize current knowledge of estrogen synthesis, regulation in the placenta, and biological effects during pregnancy and PE. Moreover, this review highlights the links among the occurrence of PE, estrogen biosynthesis, angiogenic factors, and cardiovascular risk factors. A close link between estrogen dysregulation and PE occurrence might validate estrogen levels as a biomarker but could also reveal a potential approach for prevention or cure of PE.
Collapse
Affiliation(s)
- Nadia Berkane
- Department of Gynecology and Obstetrics of University Hospital of Geneva, 1205, Genève, Switzerland.,U1195, INSERM and University Paris Sud, 94276 Kremlin Bicêtre, France
| | - Philippe Liere
- U1195, INSERM and University Paris Sud, 94276 Kremlin Bicêtre, France
| | - Jean-Paul Oudinet
- U1195, INSERM and University Paris Sud, 94276 Kremlin Bicêtre, France
| | - Alexandre Hertig
- Department of Nephrology, Tenon Hospital, APHP, 75020 Paris, France.,University of Pierre and Marie Curie, Sorbonne University, Paris 06, 75005 Paris, France.,Unité Mixte de Recherche Scientifique 1155, F-75020 Paris, France
| | - Guillaume Lefèvre
- University of Pierre and Marie Curie, Sorbonne University, Paris 06, 75005 Paris, France.,Department of Biochemistry and Hormonology, Tenon Hospital, APHP, F-75020 Paris, France
| | - Nicola Pluchino
- Department of Gynecology and Obstetrics of University Hospital of Geneva, 1205, Genève, Switzerland
| | | | - Nathalie Chabbert-Buffet
- University of Pierre and Marie Curie, Sorbonne University, Paris 06, 75005 Paris, France.,Department of Obstetrics, Gynecology and Reproductive Medicine, Tenon Hospital, APHP, F-75020 Paris, France.,INSERM, UMR-S938, Centre de Recherche Saint-Antoine, F-75012 Paris, France
| |
Collapse
|
20
|
Zhang Y, Li P, Gao Q, Simoncini T, Fu X. 2-Methoxyestradiol prevents monocyte adhesion to vascular endothelial cells via downregulation of VCAM-1 expression. Gynecol Endocrinol 2016; 32:571-6. [PMID: 26880304 DOI: 10.3109/09513590.2016.1141880] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
2-Methoxyestradiol (2-ME) reduces atherosclerotic lesion formation. However, the underlying mechanisms remain largely unknown. In this work, we investigated the effect of 2-ME on monocyte adhesion to vascular endothelial cells. Lipopolysaccharides (LPS) greatly increased the attachment of monocyte onto cultured human umbilical vascular endothelial cells (HUVECs), which was inhibited by 2-ME in a dose- and time-dependent manner, or by the vascular cell adhesion protein-1 (VCAM-1) neutralizing antibody, suggesting that a functional releationship between 2-ME and VCAM-1 may exist. In accordance with this, treatment with 2-ME (10(-)(7)-10(-)(5) M) for 6-48 h downregulated VCAM-1 protein expression. Meanwhile, the nuclear factor κB (NF-κB) p65 subunit activity and its nuclear translocation was inhibited by 2-ME in HUVECs. The PI3K inhibitor wortmannin or the specific Akt siRNA both inhibited the effects of 2-ME, suggesting that 2-ME inhibited p65 activity via PI3K/Akt signaling. In conclusion, 2-ME inhibits VCAM-1 expression and thus prevents monocyte adhesion to vascular endothelial cells via regulation of PI3K/Akt and NF-κB signaling. These findings will be helpful for better understanding the mechanisms through which 2-ME improves endothelial function.
Collapse
Affiliation(s)
- Yongfu Zhang
- a Department of Anesthesiology , Guangzhou Women and Children's Medical Center , Guangzhou, Guangdong Province , China
| | - Ping Li
- b School of Basic Sciences, Guangzhou Medical University , Guangzhou, Guangdong Province , China
| | - Qi Gao
- c Department of Anesthesiology , Longhua Hospital , Shenzhen, Guangdong Province , China , and
| | - Tommaso Simoncini
- d Molecular and Cellular Gynecological Endocrinology Laboratory (MCGEL), Department of Reproductive Medicine and Child Development, University of Pisa , Pisa , Italy
| | - Xiaodong Fu
- b School of Basic Sciences, Guangzhou Medical University , Guangzhou, Guangdong Province , China
| |
Collapse
|
21
|
Abstract
The estrogen receptors, ERα, ERβ, and GPER, mediate the effects of estrogenic compounds on their target tissues. Estrogen receptors are located in the tissues of the female reproductive tract and breast as one would expect, but also in tissues as diverse as bone, brain, liver, colon, skin, and salivary gland. The purpose of this discussion of the estrogen receptors is to provide a brief overview of the estrogen receptors and estrogen action from perspectives such as the historical, physiological, pharmacological, pathological, structural, and ligand perspectives.
Collapse
Affiliation(s)
- Kathleen M Eyster
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, 414 E. Clark St., Vermillion, SD, 57069, USA.
| |
Collapse
|
22
|
Rigassi L, Barchiesi Bozzolo F, Lucchinetti E, Zaugg M, Fingerle J, Rosselli M, Imthurn B, Jackson EK, Dubey RK. 2-Methoxyestradiol blocks the RhoA/ROCK1 pathway in human aortic smooth muscle cells. Am J Physiol Endocrinol Metab 2015; 309:E995-1007. [PMID: 26487003 PMCID: PMC4816197 DOI: 10.1152/ajpendo.00267.2015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 10/08/2015] [Indexed: 11/22/2022]
Abstract
2-Methoxyestradiol (2-ME), a metabolite of estradiol with little affinity for estrogen receptors, inhibits proliferation of vascular smooth muscle cells; however, the molecular mechanisms underlying this effect are incompletely understood. Our previous work shows that 2-ME inhibits initiation (blocks phosphorylation of ERK and Akt) and progression (reduces cyclin expression and increases expression of cyclin inhibitors) of the mitogenic pathway and interferes with mitosis (disrupts tubulin organization). Because the RhoA/ROCK1 pathway (RhoA → ROCK1 → myosin phosphatase targeting subunit → myosin light chain) is involved in cytokinesis, herein we tested the concept that 2-ME also blocks the RhoA/ROCK1 pathway. Because of the potential importance of 2-ME for preventing/treating vascular diseases, experiments were conducted in female human aortic vascular smooth muscle cells. Microarray transcriptional profiling suggested an effect of 2-ME on the RhoA/ROCK1 pathway. Indeed, 2-ME blocked mitogen-induced GTP-bound RhoABC expression and membrane-bound RhoA, suggesting interference with the activation of RhoA. 2-ME also reduced ROCK1 expression, suggesting reduced production of the primary downstream signaling kinase of the RhoA pathway. Moreover, 2-ME inhibited RhoA/ROCK1 pathway downstream signaling, including phosphorylated myosin phosphatase targeting subunit and myosin light chain; the ROCK1 inhibitor H-1152 mimicked these effects of 2-ME; both 2-ME and H-1152 blocked cytokinesis. 2-ME also reduced the expression of tissue factor, yet another downstream signaling component of the RhoA/ROCK1 pathway. We conclude that 2-ME inhibits the pathway RhoA → ROCK1 → myosin phosphatase targeting subunit → myosin light chain, and this likely contributes to the reduced cytokinesis in 2-ME treated HASMCs.
Collapse
Affiliation(s)
- Lisa Rigassi
- Department of Reproductive Endocrinology, University Hospital Zurich, Zurich, Switzerland
| | | | - Eliana Lucchinetti
- Department of Anesthesiology and Pain Medicine and Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada; and
| | - Michael Zaugg
- Department of Anesthesiology and Pain Medicine and Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada; and
| | - Jürgen Fingerle
- Preclinical Pharma Research 60/209, F Hoffmann-La-Roche, Basel, Switzerland
| | - Marinella Rosselli
- Department of Reproductive Endocrinology, University Hospital Zurich, Zurich, Switzerland
| | - Bruno Imthurn
- Department of Reproductive Endocrinology, University Hospital Zurich, Zurich, Switzerland
| | - Edwin K Jackson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Raghvendra K Dubey
- Department of Reproductive Endocrinology, University Hospital Zurich, Zurich, Switzerland; Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland;
| |
Collapse
|
23
|
Chen W, Cui Y, Zheng S, Huang J, Li P, Simoncini T, Zhang Y, Fu X. 2-methoxyestradiol induces vasodilation by stimulating NO release via PPARγ/PI3K/Akt pathway. PLoS One 2015; 10:e0118902. [PMID: 25748432 PMCID: PMC4351983 DOI: 10.1371/journal.pone.0118902] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 01/21/2015] [Indexed: 11/18/2022] Open
Abstract
The endogenous estradiol metabolite 2-methoxyestradiol (2-ME) reduces atherosclerotic lesion formation, while the underlying mechanisms remain obscure. In this work, we investigated the vasodilatory effect of 2-ME and the role of nitric oxide (NO) involved. In vivo studies using noninvasive tail-cuff methods showed that 2-ME decreased blood pressure in Sprague Dawley rats. Furthermore, in vitro studies showed that cumulative addition of 2-ME to the aorta caused a dose- and endothelium-dependent vasodilation. This effect was unaffected by the pretreatment with the pure estrogen receptor antagonist ICI 182,780, but was largely impaired by endothelial nitric oxide synthase (eNOS) inhibitor NG-nitro-L-arginine methyl ester (L-NAME) or by phosphoinositide 3-kinase (PI3K) inhibitor wortmannin (WM). Moreover, 2-ME(10−7 ∼10−5 M)enhanced phosphorylation of Akt and eNOS and promoted NO release from cultured human umbilical endothelial cells (HUVECs). These effects were blocked by PI3K inhibitor WM, or by the transfection with Akt specific siRNA, indicating that endothelial Akt/eNOS/NO cascade plays a crucial role in 2-ME-induced vasodilation. The peroxisome proliferator-activated receptor γ (PPARγ) mRNA and protein expression were detected in HUVECs and the antagonist GW9662 or the transfection with specific PPARγ siRNA inhibited 2-ME-induced eNOS and Akt phosphorylation, leading to the impairment of NO production and vasodilation. In conclusion, 2-ME induces vasodilation by stimulating NO release. These actions may be mediated by PPARγ and the subsequent activation of Akt/eNOS cascade in vascular endothelial cells.
Collapse
Affiliation(s)
- Weiyu Chen
- School of Basic Sciences, Guangzhou Medical University, Guangzhou, 510182, Guangdong Province, China; Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong Province, China
| | - Yuhong Cui
- School of Basic Sciences, Guangzhou Medical University, Guangzhou, 510182, Guangdong Province, China
| | - Shuhui Zheng
- Research Center of Translational Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, Guangdong Province, China
| | - Jinghe Huang
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong Province, China
| | - Ping Li
- School of Basic Sciences, Guangzhou Medical University, Guangzhou, 510182, Guangdong Province, China
| | - Tommaso Simoncini
- Molecular and Cellular Gynecological Endocrinology Laboratory (MCGEL), Department of Reproductive Medicine and Child Development, University of Pisa, Pisa, 56100, Italy
| | - Yongfu Zhang
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou, 510180, Guangdong Province, China
| | - Xiaodong Fu
- School of Basic Sciences, Guangzhou Medical University, Guangzhou, 510182, Guangdong Province, China
| |
Collapse
|
24
|
Jennings BL, Moore JA, Pingili AK, Estes AM, Fang XR, Kanu A, Gonzalez FJ, Malik KU. Disruption of the cytochrome P-450 1B1 gene exacerbates renal dysfunction and damage associated with angiotensin II-induced hypertension in female mice. Am J Physiol Renal Physiol 2015; 308:F981-92. [PMID: 25694484 DOI: 10.1152/ajprenal.00597.2014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 02/11/2015] [Indexed: 11/22/2022] Open
Abstract
Recently, we demonstrated in female mice that protection against ANG II-induced hypertension and associated cardiovascular changes depend on cytochrome P-450 (CYP)1B1. The present study was conducted to determine if Cyp1b1 gene disruption ameliorates renal dysfunction and organ damage associated with ANG II-induced hypertension in female mice. ANG II (700 ng·kg(-1)·min(-1)) infused by miniosmotic pumps for 2 wk in female Cyp1b1(+/+) mice did not alter water consumption, urine output, Na(+) excretion, osmolality, or protein excretion. However, in Cyp1b1(-/-) mice, ANG II infusion significantly increased (P < 0.05) water intake (5.50 ± 0.42 ml/24 h with vehicle vs. 8.80 ± 0.60 ml/24 h with ANG II), urine output (1.44 ± 0.37 ml/24 h with vehicle vs. 4.30 ± 0.37 ml/24 h with ANG II), and urinary Na(+) excretion (0.031 ± 0.016 mmol/24 h with vehicle vs. 0.099 ± 0.010 mmol/24 h with ANG II), decreased osmolality (2,630 ± 79 mosM/kg with vehicle vs. 1,280 ± 205 mosM/kg with ANG II), and caused proteinuria (2.60 ± 0.30 mg/24 h with vehicle vs. 6.96 ± 0.55 mg/24 h with ANG II). Infusion of ANG II caused renal fibrosis, as indicated by an accumulation of renal interstitial α-smooth muscle actin, collagen, and transforming growth factor-β in Cyp1b1(-/-) but not Cyp1b1(+/+) mice. ANG II also increased renal production of ROS and urinary excretion of thiobarburic acid-reactive substances and reduced the activity of antioxidants and urinary excretion of nitrite/nitrate and the 17β-estradiol metabolite 2-methoxyestradiol in Cyp1b1(-/-) but not Cyp1b1(+/+) mice. These data suggest that Cyp1b1 plays a critical role in female mice in protecting against renal dysfunction and end-organ damage associated with ANG II-induced hypertension, in preventing oxidative stress, and in increasing activity of antioxidant systems, most likely via generation of 2-methoxyestradiol from 17β-estradiol.
Collapse
Affiliation(s)
- Brett L Jennings
- Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Joseph A Moore
- Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Ajeeth K Pingili
- Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Anne M Estes
- Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Xiao R Fang
- Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Alie Kanu
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee; and
| | - Frank J Gonzalez
- Laboratory of Metabolism, National Cancer Institute, Bethesda, Maryland
| | - Kafait U Malik
- Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee;
| |
Collapse
|
25
|
Pertegal M, Fenoy FJ, Bonacasa B, Mendiola J, Delgado JL, Hernández M, Salom MG, Bosch V, Hernández I. 2-methoxyestradiol plasma levels are associated with clinical severity indices and biomarkers of preeclampsia. Reprod Sci 2014; 22:198-206. [PMID: 24899468 DOI: 10.1177/1933719114537716] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
We investigated whether clinical severity indices and biomarkers for preeclampsia (PE) are associated with low plasmatic 2-methoxyestradiol (2ME) in the third trimester of gestation. Blood was collected from 53 women with PE and 73 control pregnant women before parturition. The concentration of 2ME was significantly higher in controls than in patients with PE (2906.43 ± 200.69 pg/mL vs 1818.41 ± 189.25 pg/mL). The risk of PE decreased as 2ME levels increased. The 2ME values were negatively correlated with systolic peak arterial pressure and proteinuria in PE. Additionally, those women with PE with lower 2ME had a more serious clinical situation and needed a more aggressive therapy. Finally, 2ME levels (in patients with PE and total population) were significantly correlated with concentrations of soluble fms-like tyrosine kinase 1 and placental growth factor . Summarizing, patients with PE had lower 2ME levels that were correlated with different clinical indices and biomarkers of severity, indicating that 2ME could be taken into account for the clinical management of this syndrome.
Collapse
Affiliation(s)
- Miriam Pertegal
- Department of Gynaecology and Obstetrics, "Virgen de la Arrixaca" Clinical Universitary Hospital, El Palmar (Murcia), Spain
| | - Francisco J Fenoy
- Department of Physiology, School of Medicine, University of Murcia, Espinardo (Murcia) Spain
| | - Barbara Bonacasa
- Department of Physiology, School of Medicine, University of Murcia, Espinardo (Murcia) Spain
| | - Jaime Mendiola
- Division of Preventive Medicine and Public Health, Department of Health and Social Sciences, School of Medicine, University of Murcia, Espinardo (Murcia), Spain
| | - Juan L Delgado
- Department of Gynaecology and Obstetrics, "Virgen de la Arrixaca" Clinical Universitary Hospital, El Palmar (Murcia), Spain
| | - Moises Hernández
- Department of Physiology, School of Medicine, University of Murcia, Espinardo (Murcia) Spain
| | - Miguel G Salom
- Department of Physiology, School of Medicine, University of Murcia, Espinardo (Murcia) Spain
| | - Vicente Bosch
- Division of Preventive Medicine and Public Health, Department of Health and Social Sciences, School of Medicine, University of Murcia, Espinardo (Murcia), Spain Department of Pediatrics, "Virgen de la Arrixaca" Clinical Universitary Hospital, El Palmar (Murcia), Spain
| | - Isabel Hernández
- Department of Physiology, School of Medicine, University of Murcia, Espinardo (Murcia) Spain
| |
Collapse
|
26
|
Jennings BL, George LW, Pingili AK, Khan NS, Estes AM, Fang XR, Gonzalez FJ, Malik KU. Estrogen metabolism by cytochrome P450 1B1 modulates the hypertensive effect of angiotensin II in female mice. Hypertension 2014; 64:134-40. [PMID: 24777982 DOI: 10.1161/hypertensionaha.114.03275] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
To determine the role of cytochrome P450 (CYP) 1B1 in the sex difference in response to angiotensin II (Ang II)-induced hypertension, female Cyp1b1(+/+) and Cyp1b1(-/-) mice were infused with Ang II (700 ng/kg per minute) or vehicle with or without ovariectomy. In addition, mice were treated with the CYP1B1 inhibitor, 2,3',4,5'-tetramethoxystilbene (TMS; 300 μg/kg IP, every third day), and 17-β estradiol metabolites, 2-hydroxyestradiol (2-OHE), 4-OHE, or 2-methoxyestradiol (1.5 mg/kg per day IP, for 2 weeks) and systolic blood pressure (SBP) measured. Ang II increased SBP more in Cyp1b1(-/-) than in Cyp1b1(+/+) mice (119±3-171±11 versus 120±4-149±4 mm Hg; P<0.05). Ang II caused cardiovascular remodeling and endothelial dysfunction and increased vascular reactivity and oxidative stress in Cyp1b1(-/-) but not in Cyp1b1(+/+)mice. The Ang II-induced increase in SBP was enhanced by ovariectomy and TMS in Cyp1b1(+/+) but not in Cyp1b1(-/-) mice. 2-OHE did not alter Ang II-induced increase in SBP in Cyp1b1(+/+) mice but minimized it in Cyp1b1(-/-) mice, whereas 4-OHE enhanced Ang II-induced increase in SBP in Cyp1b1(+/+) mice but did not alter the increased SBP in Cyp1b1(-/-) mice. 2-OHE-derived catechol-O-methyltransferase metabolite, 2-methoxyestradiol, inhibited Ang II-induced increase in SBP in Cyp1b1(-/-) mice. Ang II increased plasma levels of 2-methoxyestradiol in Cyp1b1(+/+) but not in Cyp1b1(-/-) mice and increased activity of cardiac extracellular signal-regulated kinase 1/2, p38 mitogen-activated kinase, c-Src, and Akt in Cyp1b1(-/-) but not in Cyp1b1(+/+) mice. These data suggest that CYP1B1 protects against Ang II-induced hypertension and associated cardiovascular changes in female mice, most likely mediated by 2-methoxyestradiol-inhibiting oxidative stress and the activity of these signaling molecules.
Collapse
Affiliation(s)
- Brett L Jennings
- From the Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis (B.L.J., L.W.G., A.K.P., N.S.K., A.M.E., X.R.F., K.U.M.); and Laboratory of Metabolism, National Cancer Institute, Bethesda, MD (F.J.G.)
| | - L Watson George
- From the Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis (B.L.J., L.W.G., A.K.P., N.S.K., A.M.E., X.R.F., K.U.M.); and Laboratory of Metabolism, National Cancer Institute, Bethesda, MD (F.J.G.)
| | - Ajeeth K Pingili
- From the Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis (B.L.J., L.W.G., A.K.P., N.S.K., A.M.E., X.R.F., K.U.M.); and Laboratory of Metabolism, National Cancer Institute, Bethesda, MD (F.J.G.)
| | - Nayaab S Khan
- From the Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis (B.L.J., L.W.G., A.K.P., N.S.K., A.M.E., X.R.F., K.U.M.); and Laboratory of Metabolism, National Cancer Institute, Bethesda, MD (F.J.G.)
| | - Anne M Estes
- From the Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis (B.L.J., L.W.G., A.K.P., N.S.K., A.M.E., X.R.F., K.U.M.); and Laboratory of Metabolism, National Cancer Institute, Bethesda, MD (F.J.G.)
| | - Xiao R Fang
- From the Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis (B.L.J., L.W.G., A.K.P., N.S.K., A.M.E., X.R.F., K.U.M.); and Laboratory of Metabolism, National Cancer Institute, Bethesda, MD (F.J.G.)
| | - Frank J Gonzalez
- From the Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis (B.L.J., L.W.G., A.K.P., N.S.K., A.M.E., X.R.F., K.U.M.); and Laboratory of Metabolism, National Cancer Institute, Bethesda, MD (F.J.G.)
| | - Kafait U Malik
- From the Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis (B.L.J., L.W.G., A.K.P., N.S.K., A.M.E., X.R.F., K.U.M.); and Laboratory of Metabolism, National Cancer Institute, Bethesda, MD (F.J.G.).
| |
Collapse
|
27
|
Affiliation(s)
- Sarah H Lindsey
- From the Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA.
| |
Collapse
|
28
|
Evans CE, Grover SP, Humphries J, Saha P, Patel AP, Patel AS, Lyons OT, Waltham M, Modarai B, Smith A. Antiangiogenic Therapy Inhibits Venous Thrombus Resolution. Arterioscler Thromb Vasc Biol 2014; 34:565-70. [DOI: 10.1161/atvbaha.113.302998] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Colin E. Evans
- From the Academic Department of Vascular Surgery, Cardiovascular Division, Kings College London, BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, St Thomas’ Hospital, London, United Kingdom
| | - Steven P. Grover
- From the Academic Department of Vascular Surgery, Cardiovascular Division, Kings College London, BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, St Thomas’ Hospital, London, United Kingdom
| | - Julia Humphries
- From the Academic Department of Vascular Surgery, Cardiovascular Division, Kings College London, BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, St Thomas’ Hospital, London, United Kingdom
| | - Prakash Saha
- From the Academic Department of Vascular Surgery, Cardiovascular Division, Kings College London, BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, St Thomas’ Hospital, London, United Kingdom
| | - Anant P. Patel
- From the Academic Department of Vascular Surgery, Cardiovascular Division, Kings College London, BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, St Thomas’ Hospital, London, United Kingdom
| | - Ashish S. Patel
- From the Academic Department of Vascular Surgery, Cardiovascular Division, Kings College London, BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, St Thomas’ Hospital, London, United Kingdom
| | - Oliver T. Lyons
- From the Academic Department of Vascular Surgery, Cardiovascular Division, Kings College London, BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, St Thomas’ Hospital, London, United Kingdom
| | - Matt Waltham
- From the Academic Department of Vascular Surgery, Cardiovascular Division, Kings College London, BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, St Thomas’ Hospital, London, United Kingdom
| | - Bijan Modarai
- From the Academic Department of Vascular Surgery, Cardiovascular Division, Kings College London, BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, St Thomas’ Hospital, London, United Kingdom
| | - Alberto Smith
- From the Academic Department of Vascular Surgery, Cardiovascular Division, Kings College London, BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, St Thomas’ Hospital, London, United Kingdom
| |
Collapse
|
29
|
Koganti S, Snyder R, Gumaste U, Karamyan VT, Thekkumkara T. 2-methoxyestradiol binding of GPR30 down-regulates angiotensin AT(1) receptor. Eur J Pharmacol 2013; 723:131-40. [PMID: 24262995 DOI: 10.1016/j.ejphar.2013.10.064] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Revised: 10/22/2013] [Accepted: 10/31/2013] [Indexed: 11/26/2022]
Abstract
Controlling angiotensin AT1 receptor function has been shown to be protective for many pathophysiological disorders. Although estrogen metabolite, 2-methoxyestradiol (2ME2) can down-regulate angiotensin AT1 receptor expression independently of nuclear receptors, no specific cellular targets have been identified. This study was focused on identification and validation of a cellular target responsible for 2ME2-mediated angiotensin AT1 receptor down-regulation in a continuously passaged rat liver epithelial cell line. Cell membranes were isolated and used to determine 2ME2 specific binding. Cell membranes exposed to [(3)H]2ME2 showed specific saturable binding, which was found to be pertussis toxin (PTx) sensitive. Under similar conditions, G-protein coupled receptor 30 (GPR30) agonist (G1) and antagonist (G15) inhibited 2ME2 specific binding. In these cells GPR30 was found localized to endoplasmic reticulum (ER) membranes. In intact cells, G1 down-regulated angiotensin AT1 receptor expression and this effect was reversed by G15. Furthermore, 2ME2 mediated activation of epidermal growth factor receptor (EGFR) followed by ERK1/2 phosphorylation, an essential signaling step in angiotensin AT1 receptor down-regulation, was abrogated by G15, suggesting that this signal is GPR30 dependent. Additionally, EGF was found to independently down-regulate angiotensin AT1 receptor in an ERK1/2-dependent manner. In summary, our results demonstrate for the first time that 2ME2 down-regulation of angiotensin AT1 receptor is dependent on ER membrane-associated GRP30. Moreover, this effect is facilitated by GPR30 dependent transactivation of EGFR and ERK1/2 phosphorylation. This study provides further understanding of the physiological significance of 2ME2 and its role in modulating angiotensin AT1 receptor expression.
Collapse
Affiliation(s)
- Sivaramakrishna Koganti
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center, 1300 Coulter Drive, Amarillo, TX 79106, United States
| | - Russell Snyder
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center, 1300 Coulter Drive, Amarillo, TX 79106, United States
| | - Upendra Gumaste
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center, 1300 Coulter Drive, Amarillo, TX 79106, United States
| | - Vardan T Karamyan
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX 79106, United States
| | - Thomas Thekkumkara
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center, 1300 Coulter Drive, Amarillo, TX 79106, United States.
| |
Collapse
|
30
|
Moon JY, Kim SJ, Moon MH, Chung BC, Choi MH. Differential estimation of isomeric 2- and 4-methoxylated estrogens in serum by matrix-assisted laser desorption ionization-tandem mass spectrometry. ANAL SCI 2013; 29:345-51. [PMID: 23474725 DOI: 10.2116/analsci.29.345] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
As a simple and rapid method for small-molecule analysis, matrix-assisted laser desorption ionization coupled with linear ion-trap tandem mass spectrometry (MALDI-LTQ-MS/MS) was used to differentially quantify 2- and 4-methoxyestrogens in spiked serum samples. Both 2- and 4-methoxyestrogens were consistently identified using distinct analyte-matrix adduct [M+H+CHCA](+) precursor ions when a mixture of 10 mg mL(-1) α-cyano-4-hydroxycinnamic acid (CHCA) in 0.2% trifluoroacetic acid/70% acetonitrile was used as the MALDI matrix. Especially, unusual isomeric product ions from each precursor ion were characterized during MALDI-MS/MS analysis, which permitted differential estimations of isomeric 2- and 4-methoxylated estrone and 17β-estradiol. The calibration linearity was higher than 0.99 in the dynamic range, while the inter-day precision and accuracy ranged from 6.8 to 30.6%, and from 91.4 to 108.5%, respectively. The present technique could be used as a screening assay for 2- and 4-methoxyestrogens without the need for prior chromatographic separation, opening up possible applications in large-scale pharmacokinetic studies.
Collapse
Affiliation(s)
- Ju-Yeon Moon
- Future Convergence Research Division, Korea Institute of Science and Technology, Seoul, Korea
| | | | | | | | | |
Collapse
|
31
|
Theron AE, Nolte EM, Lafanechère L, Joubert AM. Molecular crosstalk between apoptosis and autophagy induced by a novel 2-methoxyestradiol analogue in cervical adenocarcinoma cells. Cancer Cell Int 2013; 13:87. [PMID: 23977838 PMCID: PMC3766685 DOI: 10.1186/1475-2867-13-87] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 08/23/2013] [Indexed: 12/24/2022] Open
Abstract
Background 2-Methoxyestradiol has been shown to induce both autophagy and apoptosis in various carcinogenic cell lines. Although a promising anti-cancer agent, it has poor bioavailability and rapid in vivo metabolism which decreases its efficiency. In order to improve 2-methoxyestradiol’s anti-proliferative properties, a novel 2-methoxyestradiol analogue, 2-ethyl-3-O-sulphamoyl-estra-1,3,5 (10)16-tetraene (ESE-16), was previously in silico-designed in our laboratory. This study investigated ESE-16 for its anti-proliferative potential on a cervical adenocarcinoma cell (HeLa) cell line. Additionally, the possible intracellular crosstalk mechanisms between the two types of cell death were investigated. Methods and results HeLa cells exposed to 0.5 μM ESE-16 for 24 hours showed morphological evidence of both apoptotic and autophagic death pathways as assessed by polarization-optical transmitted light differential interference contrast microscopy, fluorescent microscopy and transmission electron microscopy. Flow cytometric cyclin B1 quantification revealed induction of programmed cell death after halting cell cycle progression in metaphase. Confocal microscopy demonstrated that ESE-16 caused microtubule fragmentation. Flow cytometric analysis of cell cycle progression and phosphatidylserine flip determination confirmed induction of apoptosis. Moreover, an increase in aggresome formation and microtubule-associated protein light chain, LC3, was demonstrated indicative of autophagy. Both caspase 8 and 3 were upregulated in a spectrophotometric analysis, indicating the involvement of the extrinsic pathway of apoptotic induction. Conclusions We conclude that the novel in silico-designed compound, ESE-16, exerts its anti-proliferative effect on the tumorigenic human epithelial cervical (HeLa) cells by sequentially targeting microtubule integrity, resulting in a metaphase block, causing induction of both autophagic and apoptotic cell death via a crosstalk mechanism that involves the extrinsic pathway. Future investigations will expand on signal transduction pathways involved in both apoptosis and autophagy for assessment of ESE-16 effects on microtubule dynamic instability parameters.
Collapse
Affiliation(s)
- Anne E Theron
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Private Bag X323, Arcadia, 0007 Gauteng, Pretoria, South Africa.
| | | | | | | |
Collapse
|
32
|
Quezada M, Alvarez M, Peña OA, Henríquez S, d' Alençon CA, Lange S, Oliva B, Owen GI, Allende ML. Antiangiogenic, antimigratory and antiinflammatory effects of 2-methoxyestradiol in zebrafish larvae. Comp Biochem Physiol C Toxicol Pharmacol 2013; 157:141-9. [PMID: 23142146 DOI: 10.1016/j.cbpc.2012.10.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2012] [Revised: 10/30/2012] [Accepted: 10/31/2012] [Indexed: 12/13/2022]
Abstract
2-Methoxyestradiol (2ME), an endogenous metabolite of 17β-estradiol, has been previously reported to possess antiangiogenic and antitumor properties. Herein, we demonstrate that the effects of this antiangiogenic steroid can be readily assayed in live zebrafish, introducing a convenient and robust new model system as a screening tool for both single cell and collective cell migration assays. Using the in vitro mammalian endothelial cell line EA.hy926, we first show that cell migration and angiogenesis, as estimated by wound assay and tube formation respectively, are antagonized by 2ME. In zebrafish (Danio rerio) larvae, dose-dependent exposure to 2ME diminishes (1) larval angiogenesis, (2) leukocyte recruitment to damaged lateral line neuromasts and (3) retards the lateral line primordium in its migration along the body. Our results indicate that 2ME has an effect on collective cell migration in vivo as well as previously reported anti-tumorigenic activity and suggests that the molecular mechanisms governing cell migration in a variety of contexts are conserved between fish and mammals. Moreover, we exemplify the versatility of the zebrafish larvae for testing diverse physiological processes and screening for antiangiogenic and antimigratory drugs in vivo.
Collapse
Affiliation(s)
- Marisol Quezada
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Zhou M, Hamza A, Zhan CG, Thorson JS. Assessing the regioselectivity of OleD-catalyzed glycosylation with a diverse set of acceptors. JOURNAL OF NATURAL PRODUCTS 2013; 76:279-86. [PMID: 23360118 PMCID: PMC3607945 DOI: 10.1021/np300890h] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
To explore the acceptor regioselectivity of OleD-catalyzed glucosylation, the products of OleD-catalyzed reactions with six structurally diverse acceptors flavones- (daidzein), isoflavones (flavopiridol), stilbenes (resveratrol), indole alkaloids (10-hydroxycamptothecin), and steroids (2-methoxyestradiol)-were determined. This study highlights the first synthesis of flavopiridol and 2-methoxyestradiol glucosides and confirms the ability of OleD to glucosylate both aromatic and aliphatic nucleophiles. In all cases, molecular dynamics simulations were consistent with the determined product distribution and suggest the potential to develop a virtual screening model to identify additional OleD substrates.
Collapse
Affiliation(s)
- Maoquan Zhou
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, Wisconsin 53705, USA
| | - Adel Hamza
- Department of Pharmaceutical Sciences, Center for Pharmaceutical Research and Innovation, College of Pharmacy, University of Kentucky, 789 S. Limestone St. Lexington, Kentucky 40536 USA
| | - Chang-Guo Zhan
- Department of Pharmaceutical Sciences, Center for Pharmaceutical Research and Innovation, College of Pharmacy, University of Kentucky, 789 S. Limestone St. Lexington, Kentucky 40536 USA
| | - Jon S. Thorson
- Department of Pharmaceutical Sciences, Center for Pharmaceutical Research and Innovation, College of Pharmacy, University of Kentucky, 789 S. Limestone St. Lexington, Kentucky 40536 USA
| |
Collapse
|
34
|
Hsieh YC, Jeng JS, Lin HJ, Hu CJ, Yu CC, Lien LM, Peng GS, Chen CI, Tang SC, Chi NF, Tseng HP, Chern CM, Hsieh FI, Bai CH, Chen YR, Chiou HY. Epistasis analysis for estrogen metabolic and signaling pathway genes on young ischemic stroke patients. PLoS One 2012; 7:e47773. [PMID: 23112845 PMCID: PMC3480403 DOI: 10.1371/journal.pone.0047773] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Accepted: 09/17/2012] [Indexed: 12/19/2022] Open
Abstract
Background Endogenous estrogens play an important role in the overall cardiocirculatory system. However, there are no studies exploring the hormone metabolism and signaling pathway genes together on ischemic stroke, including sulfotransferase family 1E (SULT1E1), catechol-O-methyl-transferase (COMT), and estrogen receptor α (ESR1). Methods A case-control study was conducted on 305 young ischemic stroke subjects aged ≦ 50 years and 309 age-matched healthy controls. SULT1E1 -64G/A, COMT Val158Met, ESR1 c.454−397 T/C and c.454−351 A/G genes were genotyped and compared between cases and controls to identify single nucleotide polymorphisms associated with ischemic stroke susceptibility. Gene-gene interaction effects were analyzed using entropy-based multifactor dimensionality reduction (MDR), classification and regression tree (CART), and traditional multiple regression models. Results COMT Val158Met polymorphism showed a significant association with susceptibility of young ischemic stroke among females. There was a two-way interaction between SULT1E1 -64G/A and COMT Val158Met in both MDR and CART analysis. The logistic regression model also showed there was a significant interaction effect between SULT1E1 -64G/A and COMT Val158Met on ischemic stroke of the young (P for interaction = 0.0171). We further found that lower estradiol level could increase the risk of young ischemic stroke for those who carry either SULT1E1 or COMT risk genotypes, showing a significant interaction effect (P for interaction = 0.0174). Conclusions Our findings support that a significant epistasis effect exists among estrogen metabolic and signaling pathway genes and gene-environment interactions on young ischemic stroke subjects.
Collapse
Affiliation(s)
- Yi-Chen Hsieh
- School of Public Health, Taipei Medical University, Taipei, Taiwan
| | - Jiann-Shing Jeng
- Stroke Center and Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Huey-Juan Lin
- Department of Neurology, Chi-Mei Medical Center, Tainan, Taiwan
| | - Chaur-Jong Hu
- Department of Neurology, Taipei Medical University Hospital and Shuang Ho Hospital, Taipei, Taiwan
| | - Chia-Chen Yu
- School of Public Health, Taipei Medical University, Taipei, Taiwan
| | - Li-Ming Lien
- Department of Neurology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Giia-Sheun Peng
- Department of Neurology, Tri-Service General Hospital, Taipei, Taiwan
| | - Chin-I Chen
- Department of Neurology, Wanfang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Sung-Chun Tang
- Stroke Center and Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Nai-Fang Chi
- Department of Neurology, Taipei Medical University Hospital and Shuang Ho Hospital, Taipei, Taiwan
| | - Hung-Pin Tseng
- Department of Neurology, Lotung Poh-Ai Hospital, I-Lan, Taiwan
| | - Chang-Ming Chern
- Department of Neurology, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Fang-I Hsieh
- School of Public Health, Taipei Medical University, Taipei, Taiwan
| | - Chyi-Huey Bai
- School of Public Health, Taipei Medical University, Taipei, Taiwan
| | - Yi-Rhu Chen
- School of Public Health, Taipei Medical University, Taipei, Taiwan
| | - Hung-Yi Chiou
- School of Public Health, Taipei Medical University, Taipei, Taiwan
- Dr. Chi-Hsing Huang Stroke Research Center, Taipei Medical University, Taipei, Taiwan
- * E-mail:
| | | |
Collapse
|
35
|
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
|
36
|
Abstract
Estrogens not only play a pivotal role in sexual development but are also involved in several physiological processes in various tissues including vasculature. While several epidemiological studies documented an inverse relationship between plasma estrogen levels and the incidence of cardiovascular disease and related it to the inhibition of atherosclerosis, an interventional trial showed an increase in cardiovascular events among postmenopausal women on estrogen treatment. The development of atherosclerotic lesions involves complex interplay between various pro- or anti-atherogenic processes that can be effectively studied only in vivo in appropriate animal models. With the advent of genetic engineering, transgenic mouse models of atherosclerosis have supplemented classical dietary cholesterol-induced disease models such as the cholesterol-fed rabbit. In the last two decades, these models were widely applied along with in vitro cell systems to specifically investigate the influence of estrogens on the development of early and advanced atherosclerotic lesions. The present review summarizes the results of these studies and assesses their contribution toward better understanding of molecular mechanisms underlying anti- and/or pro-atherogenic effects of estrogens in humans.
Collapse
Affiliation(s)
- Jerzy-Roch Nofer
- Center for Laboratory Medicine, University Hospital Münster, Albert Schweizer Campus 1, Gebäude A1, 48129 Münster, Germany.
| |
Collapse
|
37
|
Basini G, Grasselli F, Bussolati S, Baioni L, Bianchi F, Musci M, Careri M, Mangia A. Hypoxia stimulates the production of the angiogenesis inhibitor 2-methoxyestradiol by swine granulosa cells. Steroids 2011; 76:1433-6. [PMID: 21827779 DOI: 10.1016/j.steroids.2011.07.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Revised: 07/08/2011] [Accepted: 07/23/2011] [Indexed: 11/30/2022]
Abstract
We previously demonstrated the presence of 2-methoxyestradiol (2-ME) in swine follicular fluid. Present study was aimed first of all to investigate if swine granulosa cell produce 2-ME; in addition, we tried to assess a potential effect of hypoxia in modulating 2-ME output. Finally, we explored the effect of 2-ME in an angiogenesis bioassay set up in our lab. Our data show that cultured granulosa cells are able to produce 2-ME; interestingly, the secretion of the hormone appeared to be stimulated by hypoxia. Angiogenesis bioassay points out that 2-ME displays an inhibitory effect on neovascularisation. Therefore our data suggest that 2-ME could be a local effector in determining the fine tuning responsible for follicle angiogenesis. These data deserve special attention since the ovary is a valuable experimental model in angiogenesis research.
Collapse
Affiliation(s)
- Giuseppina Basini
- Dipartimento di Produzioni Animali, Biotecnologie Veterinarie, Qualità e Sicurezza degli Alimenti, Sezione di Fisiologia Veterinaria, Università degli Studi di Parma, Via del Taglio 8, 43126 Parma, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Wilhelmson AS, Bourghardt-Fagman J, Gogos JA, Fogelstrand P, Tivesten A. Catechol-O-methyltransferase is dispensable for vascular protection by estradiol in mouse models of atherosclerosis and neointima formation. Endocrinology 2011; 152:4683-90. [PMID: 22009725 DOI: 10.1210/en.2011-1458] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Estradiol is converted to the biologically active metabolite 2-methoxyestradiol via the activity of the enzyme catechol-O-methyltransferase (COMT). Exogenous administration of both estradiol and 2-methoxyestradiol reduces experimental atherosclerosis and neointima formation, and COMT-dependent formation of 2-methoxyestradiol likely mediates the antimitogenic effect of estradiol on smooth muscle cells in vitro. This study evaluated whether 2-methoxyestradiol mediates the vasculoprotective actions of estradiol in vivo. Wild-type (WT) and COMT knockout (COMTKO) mice on an apolipoprotein E-deficient background were gonadectomized and treated with estradiol or placebo. Exogenous estradiol reduced atherosclerotic lesion formation in both females (WT, -78%; COMTKO, -82%) and males (WT, -48%; COMTKO, -53%) and was equally effective in both genotypes. We further evaluated how exogenous estradiol affected neointima formation after ligation of the carotid artery in ovariectomized female mice; estradiol reduced intimal hyperplasia to a similar extent in both WT (-80%) and COMTKO (-77%) mice. In ovarian-intact female COMTKO mice, atherosclerosis was decreased (-25%) compared with WT controls. In conclusion, the COMT enzyme is dispensable for vascular protection by exogenous estradiol in experimental atherosclerosis and neointima formation in vivo. Instead, COMT deficiency in virgin female mice with intact endogenous production of estradiol results in relative protection against atherosclerosis.
Collapse
Affiliation(s)
- Anna S Wilhelmson
- Wallenberg Laboratory for Cardiovascular Research, Institute of Medicine, Sahlgrenska University Hospital, University of Gothenburg, Bruna Stråket 16, SE-413 45 Gothenburg, Sweden
| | | | | | | | | |
Collapse
|
39
|
Zhang Q, Ma Y, Cheng YF, Li WJ, Zhang Z, Chen SY. Involvement of reactive oxygen species in 2-methoxyestradiol-induced apoptosis in human neuroblastoma cells. Cancer Lett 2011; 313:201-10. [PMID: 21978530 DOI: 10.1016/j.canlet.2011.09.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2011] [Revised: 09/05/2011] [Accepted: 09/05/2011] [Indexed: 12/27/2022]
Abstract
Neuroblastoma is the most common extra-cranial solid tumor in children. Despite advances in the treatment of childhood cancer, outcomes for children with advanced-stage neuroblastoma remain poor. Here we reported that 2-methoxyestradiol (2-ME) inhibited the proliferation and induced apoptosis in human neuroblastoma SK-N-SH and SH-SY5Y cells. 2-ME treatment also resulted in the generation of ROS and the loss of mitochondrial membrane potential in SK-N-SH and SH-SY5Y, indicating that 2-ME-induced apoptosis is mediated by ROS. This is supported by the results that have shown that co-treatment with antioxidants, VC, L-GSH and MitoQ(10), decreased 2-ME-induced generation of ROS and the loss of the mitochondrial membrane potential, increased the Bcl-2/Bax ratio, decreased 2-ME-induced activation of caspase-9 and caspase-3 and the up-regulation of apoptosis-inducing factor (AIF), and prevented 2-ME-induced apoptosis in SK-N-SH and SH-SY5Y cells. These results suggested that oxidative stress plays an important role in 2-ME-induced apoptotic death of human neuroblastoma cells.
Collapse
Affiliation(s)
- Qi Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, China
| | | | | | | | | | | |
Collapse
|
40
|
Serum 2-methoxyestradiol, an estrogen metabolite, is positively associated with serum HDL-C in a population-based sample. Lipids 2011; 47:35-8. [PMID: 21809102 DOI: 10.1007/s11745-011-3600-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Accepted: 07/12/2011] [Indexed: 01/27/2023]
Abstract
Serum HDL cholesterol (HDL-C) is inversely associated with coronary artery disease, ischemic stroke, and atherosclerosis in men and women. Among postmenopausal women, oral conjugated equine estrogen (CEE) increases serum HDL-C. This is due to activation of hepatic nuclear estrogen receptors, resulting in increased HDL-C expression, as well as modulation of proteins which metabolize HDL-C. 2-methoxyestradiol (2-MeOE2), an estrogen metabolite, has several vasculoprotective effects and may play a role in HDL-C production. 2-MeOE2 inhibits HMG-CoA reductase in vitro but no study has examined the relationship between serum 2-MeOE2 and serum HDL-C. A population-based sample provided information regarding demographic characteristics and use of antihyperlipidemic medications. Serum was analyzed for 17β-estradiol (E2), estrogen metabolites (EMs), and lipoproteins. Results included serum EM data from 51 men and 47 postmenopausal women. Preliminary analysis revealed no correlation between 2-MeOE2 and serum HDL-C in men so the current analysis includes only women (N = 40) with no missing demographic, medication, EM, or lipoprotein data. Linear regression revealed that serum 2-MeOE2 and antihyperlipidemic medications were positively associated with serum HDL-C (β = 0.276, P = 0.043, and β = 0.307, P = 0.047, respectively) when age, race/ethnicity, and body mass index were held constant. Prospective studies are needed to determine if 2-MeOE2 is causally related to HDL-C in women.
Collapse
|
41
|
Roten LT, Fenstad MH, Forsmo S, Johnson MP, Moses EK, Austgulen R, Skorpen F. A low COMT activity haplotype is associated with recurrent preeclampsia in a Norwegian population cohort (HUNT2). Mol Hum Reprod 2011; 17:439-46. [PMID: 21355050 PMCID: PMC3116680 DOI: 10.1093/molehr/gar014] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Revised: 01/23/2011] [Accepted: 02/17/2011] [Indexed: 01/15/2023] Open
Abstract
The etiology of preeclampsia is complex, with susceptibility being attributable to multiple environmental factors and a large genetic component. Although many candidate genes for preeclampsia have been suggested and studied, the specific causative genes still remain to be identified. Catechol-O-methyltransferase (COMT) is an enzyme involved in catecholamine and estrogen degradation and has recently been ascribed a role in development of preeclampsia. In the present study, we have examined the COMT gene by genotyping the functional Val108/158Met polymorphism (rs4680) and an additional single-nucleotide polymorphism, rs6269, predicting COMT activity haplotypes in a large Norwegian case/control cohort (n(cases)= 1135, n(controls)= 2262). A low COMT activity haplotype is associated with recurrent preeclampsia in our cohort. This may support the role of redox-regulated signaling and oxidative stress in preeclampsia pathogenesis as suggested by recent studies in a genetic mouse model. The COMT gene might be a genetic risk factor shared between preeclampsia and cardiovascular diseases.
Collapse
Affiliation(s)
- L T Roten
- Department of Cancer Research and Molecular Medicine, Faculty of Medicine, Norwegian University of Science and Technology (NTNU), Trondheim 7006, Norway.
| | | | | | | | | | | | | |
Collapse
|
42
|
Synergistic therapeutic effects of 2-methoxyestradiol with either sildenafil or bosentan on amelioration of monocrotaline-induced pulmonary hypertension and vascular remodeling. J Cardiovasc Pharmacol 2011; 56:475-83. [PMID: 20881615 DOI: 10.1097/fjc.0b013e3181f215e7] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
2-Methoxyestradiol (2ME) is a major nonestrogenic metabolite of estradiol. Our previous studies suggest that 2ME, in several models of cardiac and/or vascular injury, strongly inhibits cardiac and vascular remodeling. Furthermore, our most recent study shows that in male rats, 2ME attenuates the development and retards the progression of monocrotaline (MCT)-induced pulmonary arterial hypertension (PAH), and in female rats, 2ME eliminates the exacerbation of PAH and the increased mortality due to ovariectomy. The current standard of care of patients with PAH includes treatment with an endothelin receptor antagonist (eg, bosentan) or a phosphodiesterase5 inhibitor (eg, sildenafil). Moreover, combination therapy is often prescribed. Therefore, in the present study, we compared the efficacy of 2ME (10 μg · kg(-1) · h(-1), 2ME-10) to the effects of bosentan (200 mg/kg; BOS), sildenafil (50 mg/kg; SIL), and their respective combinations with 2ME-10 (2ME + BOS and 2ME + SIL groups, respectively). Treatments were initiated 12 days after administration of MCT (60 mg/kg). Twenty-eight days after MCT administration, right ventricular peak systolic pressure was measured and morphometric analysis was conducted. 2ME exhibited beneficial effects in pulmonary hypertensive animals and had efficacy comparable to that of BOS and SIL. Importantly, combination treatments had favorable effects on survival, vascular remodeling, and inflammatory response, and the 2ME + SIL combination was significantly more efficacious than any other treatment. These results indicate that 2ME is effective in experimental PAH and suggests that 2ME may provide additional therapeutic benefit over existing drugs used for the treatment of pulmonary hypertension.
Collapse
|
43
|
Sarkanen JR, Mannerström M, Vuorenpää H, Uotila J, Ylikomi T, Heinonen T. Intra-Laboratory Pre-Validation of a Human Cell Based in vitro Angiogenesis Assay for Testing Angiogenesis Modulators. Front Pharmacol 2011; 1:147. [PMID: 21779245 PMCID: PMC3134867 DOI: 10.3389/fphar.2010.00147] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2010] [Accepted: 12/31/2010] [Indexed: 11/13/2022] Open
Abstract
The developed standardized human cell based in vitro angiogenesis assay was intra-laboratory pre-validated to verify that the method is reliable and relevant for routine testing of modulators of angiogenesis, e.g., pharmaceuticals and industrial chemicals. This assay is based on the earlier published method but it was improved and shown to be more sensitive and rapid than the previous assay. The performance of the assay was assessed by using six reference chemicals, which are widely used pharmaceuticals that inhibit angiogenesis: acetyl salicylic acid, erlotinib, 2-methoxyestradiol, levamisole, thalidomide, and anti-vascular endothelial growth factor. In the intra-laboratory pre-validation, the sensitivity of the assay (upper and lower limits of detection and linearity of response in tubule formation), batch to batch variation in tubule formation between different Master cell bank batches, and precision as well as the reliability of the assay (reproducibility and repeatability) were tested. The pre-set acceptance criteria for the intra-laboratory pre-validation study were met. The relevance of the assay in man was investigated by comparing the effects of reference chemicals and their concentrations to the published human data. The comparison showed a good concordance, which indicates that this human cell based angiogenesis model predicts well the effects in man and has the potential to be used to supplement and/or replace of animal tests.
Collapse
Affiliation(s)
- Jertta-Riina Sarkanen
- Finnish Center for Alternative Methods, Medical School, University of Tampere Tampere, Finland
| | | | | | | | | | | |
Collapse
|
44
|
Verenich S, Gerk PM. Therapeutic promises of 2-methoxyestradiol and its drug disposition challenges. Mol Pharm 2010; 7:2030-9. [PMID: 20831190 DOI: 10.1021/mp100190f] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
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, Virginia 23298-0581, USA
| | | |
Collapse
|
45
|
Barchiesi F, Lucchinetti E, Zaugg M, Ogunshola OO, Wright M, Meyer M, Rosselli M, Schaufelberger S, Gillespie DG, Jackson EK, Dubey RK. Candidate genes and mechanisms for 2-methoxyestradiol-mediated vasoprotection. Hypertension 2010; 56:964-72. [PMID: 20921434 DOI: 10.1161/hypertensionaha.110.152298] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
2-Methoxyestradiol (2-ME; estradiol metabolite) inhibits vascular smooth muscle cell (VSMC) growth and protects against atherosclerosis and vascular injury; however, the mechanisms by which 2-ME induces these actions remain obscure. To assess the impact of 2-ME on biochemical pathways regulating VSMC biology, we used high-density oligonucleotide microarrays to identify differentially expressed genes in cultured human female aortic VSMCs treated with 2-ME acutely (4 hours) or long term (30 hours). Both single gene analysis and Gene Set Enrichment Analysis revealed 2-ME-induced downregulation of genes involved in mitotic spindle assembly and function in VSMCs. Also, Gene Set Enrichment Analysis identified effects of 2-ME on genes regulating cell-cycle progression, cell migration/adhesion, vasorelaxation, inflammation, and cholesterol metabolism. Transcriptional changes were associated with changes in protein expression, including inhibition of cyclin D1, cyclin B1, cyclin-dependent kinase 6, cyclin-dependent kinase 4, tubulin polymerization, cholesterol and steroid synthesis, and upregulation of cyclooxygenase 2 and matrix metalloproteinase 1. Microarray data suggested that 2-ME may activate peroxisome proliferator-activated receptors (PPARs) in VSMCs, and 2-ME has structural similarities with rosiglitazone (PPARγ agonist). However, our finding of weak activation and lack of binding of 2-ME to PPARs suggests that 2-ME may modulate PPAR-associated genes via indirect mechanisms, potentially involving cyclooxygenase 2. Indeed, the antimitogenic effects of 2-ME at concentrations that do not inhibit tubulin polymerization were blocked by the PPAR antagonist GW9662 and the cyclooxygenase 2 inhibitor NS398. Finally, we demonstrated that 2-ME inhibited hypoxia-inducible factor 1α. Identification of candidate genes that are positively or negatively regulated by 2-ME provides important leads to investigate and better understand the mechanisms by which 2-ME induces its vasoprotective actions.
Collapse
Affiliation(s)
- Federica Barchiesi
- Department of Obstetrics and Gynecology, Clinic for Reproductive Endocrinology, University Hospital Zurich, Zurich, Switzerland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Shenoy V, Kanasaki K, Kalluri R. Pre-eclampsia: connecting angiogenic and metabolic pathways. Trends Endocrinol Metab 2010; 21:529-36. [PMID: 20646932 DOI: 10.1016/j.tem.2010.05.002] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2010] [Revised: 05/04/2010] [Accepted: 05/06/2010] [Indexed: 10/19/2022]
Abstract
Pre-eclampsia is a hypertensive disease of pregnancy with a worldwide incidence of 5-8%. This review focuses on recent developments in pre-eclampsia research related to angiogenesis and metabolism. We first address the 'angiogenic imbalance' theory, which hypothesizes that pre-eclampsia results from an imbalance of factors that promote or antagonize angiogenesis, such as soluble fms-like tyrosine kinase (sFlt1), 2-methoxyestradiol (2-ME) and catechol-O-methyltransferase (COMT). Next, we analyze the association between pre-eclampsia and dysfunctional metabolism of both homocysteine and placental glycogen. We hope that illuminating some of the various connections existing between angiogenesis and metabolism in pre-eclampsia will facilitate the update or reconsideration of old models of pathogenesis.
Collapse
Affiliation(s)
- Vivek Shenoy
- Division of Matrix Biology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | | | | |
Collapse
|
47
|
The in vitro effects of 2-methoxyestradiol-bis-sulphamate on cell numbers, membrane integrity and cell morphology, and the possible induction of apoptosis and autophagy in a non-tumorigenic breast epithelial cell line. Cell Mol Biol Lett 2010; 15:564-81. [PMID: 20697831 PMCID: PMC6275594 DOI: 10.2478/s11658-010-0030-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Accepted: 08/02/2010] [Indexed: 11/20/2022] Open
Abstract
2-methoxyestradiol (2ME2) exerts estrogen receptor-independent anti-proliferative, anti-angiogenic and anti-tumor activity in vitro and in vivo. Due to its low bioavailability and rapid metabolic degradation, several analogues have been developed in recent years. 2-methoxyestradiol-bis-sulphamate (2-MeOE2bisMATE) is a bis-sulphamoylated derivative of 2ME2 with anti-proliferative activity. The aim of this study was to investigate cell signaling events induced by 2-MeOE2bisMATE in a non-tumorigenic cell line (MCF-12A) by analysing its influence on cell number, morphology and membrane integrity, and the possible induction of apoptosis and autophagy. Dose- and time-dependent studies revealed that 48 h exposure to 2-MeOE2bisMATE (0.4 μM) resulted in a decrease in cell numbers to 79%. A slight increase in the level of lactate dehydrogenase production was observed in the 2-MeOE2bisMATE-treated cells. Morphological studies revealed an increase in the number of cells in metaphase. Hallmarks of apoptosis were also found, namely nuclear fragmentation and apoptotic bodies. In addition, increased lysosomal staining was observed via fluorescent microscopy, suggesting the induction of another type of cell death, namely autophagy. Since 2-MeOE2bisMATE is regarded as a potential anti-cancer agent, it is also imperative to investigate the susceptibility of non-tumorigenic cells to its influence. The data generated from this study contributes to the understanding of the action that 2-MeOE2bisMATE exerts on the non-tumorigenic MCF-12A breast epithelial cell line.
Collapse
|
48
|
Stander BA, Marais S, Vorster CJJ, Joubert AM. In vitro effects of 2-methoxyestradiol on morphology, cell cycle progression, cell death and gene expression changes in the tumorigenic MCF-7 breast epithelial cell line. J Steroid Biochem Mol Biol 2010; 119:149-60. [PMID: 20193762 DOI: 10.1016/j.jsbmb.2010.02.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2009] [Revised: 02/04/2010] [Accepted: 02/22/2010] [Indexed: 02/06/2023]
Abstract
In the present study, the antiproliferative mechanism of action of 1 microM 2-methoxyestradiol (2ME) was investigated in the MCF-7 cell line. Measurement of intracellular cyclin B and cytochrome c protein levels, reactive oxygen species formation, cell cycle progression and apoptosis induction were conducted by means of flow cytometry. Morphological changes were evaluated using transmission electron microscopy and fluorescent microscopy by employing Hoechst 33342 and acridine orange. Gene expression changes were conducted by means of microarrays. 2ME-treated cells demonstrated an increase in cyclin B protein levels, hydrogen peroxide formation, intracellular levels of cytochrome c, as well as an increase in early and late stages of apoptosis. In addition, morphological data revealed the presence of autophagic processes. Fluorescent microscopy showed an increase in acridine orange staining and electron microscopy revealed an increase in vacuolar formation in 2ME-treated cells. The gene expression of several genes associated with mRNA translation, autophagy-related processes and genes involved in microtubule dynamics were affected. The study contributes to the mechanistic understanding of 2ME's growth inhibition in MCF-7 cells and highlights the possibility of both apoptotic and autophagic processes being activated in response to 2ME treatment in this cell line.
Collapse
Affiliation(s)
- B A Stander
- Department of Physiology, University of Pretoria, Pretoria, South Africa
| | | | | | | |
Collapse
|