Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 123
Filter
1.
Best Pract Res Clin Endocrinol Metab ; 38(1): 101815, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37634998

ABSTRACT

Menopausal women with an intact uterus choosing estrogens for menopausal symptom relief require a progestogen for endometrial protection. The aim of this systematic review was to evaluate the risks of endometrial hyperplasia resp. malignancy with different progestogens used in combined MHT. Overall, 84 RCTs were included. We found that 1) most studies were done with NETA, followed by MPA, MP and DYD and LNG, 2) most progestogens were only available as oral formulations, 3) the most frequently studied progestogens (oral MP, DYD, MPA, oral and transdermal NETA, transdermal LNG) were assessed in continuously as well as in sequentially combined MHT regimens, 4) FDA endometrial safety criteria were only fulfilled for some progestogen formulations, 5) most studies demonstrated endometrial protection for the progestogen dose and time period examined. However, 6) study quality varied which should be taken into account, when choosing a combined MHT, especially if off-label-use is chosen.


Subject(s)
Endometrial Hyperplasia , Progestins , Female , Humans , Progestins/therapeutic use , Endometrium/pathology , Hormone Replacement Therapy , Endometrial Hyperplasia/chemically induced , Endometrial Hyperplasia/prevention & control , Endometrial Hyperplasia/drug therapy , Menopause , Estrogen Replacement Therapy/adverse effects
2.
Minerva Obstet Gynecol ; 75(5): 491-497, 2023 Oct.
Article in English | MEDLINE | ID: mdl-36255159

ABSTRACT

Insertion of a LNG-Intra-uterine System (LNG-IUS) has many gynecological indications. The approved indications worldwide are contraception, treatment of abnormal uterine bleeding depending on not organic disease, and endometrial protection in case of an estrogenic therapy. Instead adenomyosis, fibroids, and fertility-sparing management of endometrial hyperplasia or early endometrial cancer in patients with desire of pregnancy are off label indications. Hydroureteronephrosis is an uncommon complication during LNG-IUS insertion. There are few cases described in the literature. The first diagnostic approach for this complication is an abdominal-pelvic ultrasound scan to identify the abnormal position of the device. Diagnostic management includes computed tomography (CT) or magnetic resonance imaging (MRI), which are necessary to confirm hydroureteronephrosis and to assess the exact location of the LNG-IUS in the abdomen. A minimally invasive approach is the standard of care with the removal of the device, while the therapeutic management of the hydroureteronephrosis depends on ureteral and kidney involvement. We report the history of a dislocated LNG-IUS in the left paracervical space with subsequent ipsilateral hydroureteronephrosis. In our case we removed the device through hysteroscopy and later inserted a J-J stent. Follow-up at three months revealed the persistence of left hydroureteronephrosis, so we performed ureter reimplantation. We also performed a review of the literature to analyze common diagnostic and therapeutic pathways for this rare complication.


Subject(s)
Endometrial Hyperplasia , Endometrial Neoplasms , Intrauterine Devices, Medicated , Pregnancy , Female , Humans , Levonorgestrel/therapeutic use , Intrauterine Devices, Medicated/adverse effects , Endometrial Hyperplasia/drug therapy , Endometrial Hyperplasia/prevention & control , Endometrial Neoplasms/complications , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/prevention & control , Endometrium
3.
Nutrients ; 14(9)2022 Apr 30.
Article in English | MEDLINE | ID: mdl-35565857

ABSTRACT

Endometrial hyperplasia (EH) is the most common risk factor for endometrial malignancy in females. The pathogenesis of EH has been directly linked to uterine inflammation, which can result in abnormal cell division and decreased apoptosis. Piceatannol (PIC), a natural polyphenolic stilbene, is known to exert anti-inflammatory, antioxidant and anti-proliferative activities. The aim of the present study was to examine the potential preventive role of PIC in estradiol benzoate (EB)-induced EH in rats. A self-nanoemulsifying drug delivery system (SNEDDS) was prepared to improve the solubility of the PIC. Therefore, thirty female Wistar rats were divided into five groups: (1) control, (2) PIC SNEDDS (10 mg/kg), (3) EB (0.6 mg/kg), (4) EB + PIC SNEDDS (5 mg/kg) and (5) EB + PIC SNEDDS (10 mg/kg). The administration of PIC SNEDDS prevented EB-induced increases in uterine weights and histopathological changes. Additionally, it displayed pro-apoptotic and antioxidant activity in the endometrium. Immunohistochemical staining of uterine sections co-treated with PIC SNEDDS showed significantly decreased expression of tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6) and nuclear transcription factor-kappa B (NF-κB). This anti-inflammatory effect was further confirmed by a significant increase in Nrf2 and heme oxygenase-1 (HO-1) expression. These results indicate that SNEDDS nanoformulation of PIC possesses protective effects against experimentally induced EH.


Subject(s)
Endometrial Hyperplasia , Stilbenes , Animals , Anti-Inflammatory Agents/therapeutic use , Antioxidants/metabolism , Endometrial Hyperplasia/chemically induced , Endometrial Hyperplasia/drug therapy , Endometrial Hyperplasia/prevention & control , Estradiol/pharmacology , Female , Heme Oxygenase-1/metabolism , Humans , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Oxidative Stress , Rats , Rats, Wistar , Stilbenes/therapeutic use
4.
Post Reprod Health ; 28(1): 40-46, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34841960

ABSTRACT

Progestogen administration is required to oppose naturally produced or administered estrogens to provide endometrial protection. Within HRT regimens, this should be delivered for at least the same duration as that produced during the luteal phase of the monthly cycle and in the recommended doses to protect against the risk of endometrial hyperplasia and endometrial cancer. This includes progestogens administered for 12-14 days a month in sequential regimens and continuous daily intake in continuous combined HRT regimens. Shorter durations and lower doses of progestogen intake are likely to be associated with an increased risk of breakthrough bleeding, endometrial hyperplasia, and endometrial cancer.


Subject(s)
Endometrial Hyperplasia , Progestins , Endometrial Hyperplasia/prevention & control , Endometrium , Estrogens , Female , Humans , Menopause , Progestins/therapeutic use
5.
Toxicol Mech Methods ; 31(1): 43-52, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32967526

ABSTRACT

Early detection and treatment of endometrial hyperplasia (EH) is mandatory for endometrial cancer prevention. Several bioactive agents of plant origin have been shown to elicit their chemotherapeutic effect against tumors and cancer via induction of mitochondrial permeability transition(mPT) pore opening. This research was therefore aimed at evaluating the potential chemopreventive effect of methyl palmitate (MP), on estradiol benzoate(EB)-induced EH, looking at the mitochondrial-mediated pathway and other possible mechanisms of action. Mitochondria were isolated using differential centrifugation. The mPT pore, mitochondrial ATPase (mATPase) activity, lipid peroxidation and cytochrome c release were determined by standard methods using spectrophotometer. Uterine interleukin 1b, MDA levels and SOD, GSH activities, were determined using commercially available kits. The uterine histological and immunohistochemical assessment of estrogen receptor (ERα), IL-1b and caspas-3 were carried out. The fibroblast cell count density was determined using histomorphometry. At all the concentrations of MP used, there was no significant induction of mPT pore opening, neither any enhancement of mATPase activity nor release of cytochrome c when compared to the control. Similar pattern of results were recorded for the in vivo study. However, there was marked increase in the uterine MDA and interleukin 1b levels, with concurrent decrease in SOD and GSH activities, in the EB-treated group, which was significantly reversed by MP co-administration. Endometrial Hyperplasia observed in the EB-treated group was ameliorated by MP co-administration. The immunoexpression of ERα and IL-1b in the EB-treated group was reversed by MP co-administration. This study suggests anti-inflammatory, antioxidant and anti-proliferative potential of MP against EB-induced EH.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Cell Proliferation/drug effects , Endometrial Hyperplasia/prevention & control , Endometrium/drug effects , Estradiol/analogs & derivatives , Mitochondria/drug effects , Palmitates/pharmacology , Animals , Cytochromes c/metabolism , Endometrial Hyperplasia/chemically induced , Endometrial Hyperplasia/metabolism , Endometrial Hyperplasia/pathology , Endometrium/metabolism , Endometrium/pathology , Estradiol/toxicity , Estrogen Receptor alpha/metabolism , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Interleukin-1beta/metabolism , Lipid Peroxidation/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Proton-Translocating ATPases/metabolism , Oxidative Stress/drug effects , Rats, Wistar , Signal Transduction
6.
Cochrane Database Syst Rev ; 12: CD007245, 2020 12 21.
Article in English | MEDLINE | ID: mdl-33348436

ABSTRACT

BACKGROUND: Adjuvant tamoxifen reduces the risk of breast cancer recurrence in women with oestrogen receptor-positive breast cancer. Tamoxifen also increases the risk of postmenopausal bleeding, endometrial polyps, hyperplasia, and endometrial cancer. The levonorgestrel-releasing intrauterine system (LNG-IUS) causes profound endometrial suppression. This systematic review considered the evidence that the LNG-IUS prevents the development of endometrial pathology in women taking tamoxifen as adjuvant endocrine therapy for breast cancer. OBJECTIVES: To determine the effectiveness and safety of the levonorgestrel intrauterine system (LNG-IUS) in pre- and postmenopausal women taking adjuvant tamoxifen following breast cancer for the outcomes of endometrial and uterine pathology including abnormal vaginal bleeding or spotting, and secondary breast cancer events. SEARCH METHODS: We searched the following databases on 29 June 2020; The Cochrane Gynaecology and Fertility Group specialised register, Cochrane Central Register of Controlled Trials, MEDLINE, Embase, PsycINFO and Cumulative Index to Nursing and Allied Health Literature. We searched the Cochrane Breast Cancer Group specialised register on 4 March 2020. We also searched two trials registers, checked references for relevant trials and contacted study authors and experts in the field to identify additional studies. SELECTION CRITERIA: We included randomised controlled trials (RCTs) of women with breast cancer on adjuvant tamoxifen that compared the effectiveness of the LNG-IUS with endometrial surveillance versus endometrial surveillance alone on the incidence of endometrial pathology. DATA COLLECTION AND ANALYSIS: We used standard methodological procedures recommended by Cochrane. The primary outcome measure was endometrial pathology (including polyps, endometrial hyperplasia, or endometrial cancer), diagnosed at hysteroscopy or endometrial biopsy. Secondary outcome measures included fibroids, abnormal vaginal bleeding or spotting, breast cancer recurrence, and breast cancer-related deaths. We rated the overall certainty of evidence using GRADE methods. MAIN RESULTS: We included four RCTs (543 women analysed) in this review. We judged the certainty of the evidence to be moderate for all of the outcomes, due to imprecision (i.e. limited sample sizes and low event rates). In the included studies, the active treatment arm was the 20 µg/day LNG-IUS plus endometrial surveillance; the control arm was endometrial surveillance alone. In tamoxifen users, the LNG-IUS probably reduces the incidence of endometrial polyps compared to the control group over both a 12-month period (Peto odds ratio (OR) 0.22, 95% confidence interval (CI) 0.08 to 0.64, I² = 0%; 2 RCTs, n = 212; moderate-certainty evidence) and over a long-term follow-up period (24 to 60 months) (Peto OR 0.22, 95% CI 0.13 to 0.39; I² = 0%; 4 RCTs, n = 417; moderate-certainty evidence). For long-term follow-up, this suggests that if the incidence of endometrial polyps following endometrial surveillance alone is assumed to be 23.5%, the incidence following LNG-IUS with endometrial surveillance would be between 3.8% and 10.7%.  The LNG-IUS probably slightly reduces the incidence of endometrial hyperplasia compared with controls over a long-term follow-up period (24 to 60 months) (Peto OR 0.13, 95% CI 0.03 to 0.67; I² = 0%; 4 RCTs, n = 417; moderate-certainty evidence). This suggests that if the chance of endometrial hyperplasia following endometrial surveillance alone is assumed to be 2.8%, the chance following LNG-IUS with endometrial surveillance would be between 0.1% and 1.9%. However, it should be noted that there were only six cases of endometrial hyperplasia. There was insufficient evidence to reach a conclusion regarding the incidence of endometrial cancer in tamoxifen users, as no studies reported cases of endometrial cancer. At 12 months of follow-up, the LNG-IUS probably increases abnormal vaginal bleeding or spotting compared to the control group (Peto OR 7.26, 95% CI 3.37 to 15.66; I² = 0%; 3 RCTs, n = 376; moderate-certainty evidence). This suggests that if the chance of abnormal vaginal bleeding or spotting following endometrial surveillance alone is assumed to be 1.7%, the chance following LNG-IUS with endometrial surveillance would be between 5.6% and 21.5%. By 24 months of follow-up, abnormal vaginal bleeding or spotting occurs less frequently than at 12 months of follow-up, but is still more common in the LNG-IUS group than the control group (Peto OR 2.72, 95% CI 1.04 to 7.10; I² = 0%; 2 RCTs, n = 233; moderate-certainty evidence). This suggests that if the chance of abnormal vaginal bleeding or spotting following endometrial surveillance alone is assumed to be 4.2%, the chance following LNG-IUS with endometrial surveillance would be between 4.4% and 23.9%. By 60 months of follow-up, there were no cases of abnormal vaginal bleeding or spotting in either group. The numbers of events for the following outcomes were low: fibroids (n = 13), breast cancer recurrence (n = 18), and breast cancer-related deaths (n = 16). As a result, there is probably little or no difference in these outcomes between the LNG-IUS treatment group and the control group.  AUTHORS' CONCLUSIONS: The LNG-IUS probably slightly reduces the incidence of benign endometrial polyps and endometrial hyperplasia in women with breast cancer taking tamoxifen. At 12 and 24 months of follow-up, the LNG-IUS probably increases abnormal vaginal bleeding or spotting among women in the treatment group compared to those in the control. Data were lacking on whether the LNG-IUS prevents endometrial cancer in these women. There is no clear evidence from the available RCTs that the LNG-IUS affects the risk of breast cancer recurrence or breast cancer-related deaths. Larger studies are necessary to assess the effects of the LNG-IUS on the incidence of endometrial cancer, and to determine whether the LNG-IUS might have an impact on the risk of secondary breast cancer events.


Subject(s)
Breast Neoplasms/prevention & control , Endometrial Hyperplasia/prevention & control , Endometrial Neoplasms/prevention & control , Intrauterine Devices, Medicated , Levonorgestrel/administration & dosage , Neoplasm Recurrence, Local/prevention & control , Adenocarcinoma/chemically induced , Adenocarcinoma/prevention & control , Antineoplastic Agents, Hormonal/adverse effects , Breast Neoplasms/chemistry , Breast Neoplasms/mortality , Chemotherapy, Adjuvant , Confidence Intervals , Contraceptive Agents, Female/administration & dosage , Endometrial Hyperplasia/chemically induced , Endometrial Hyperplasia/epidemiology , Endometrial Neoplasms/chemically induced , Endometrial Neoplasms/epidemiology , Female , Humans , Levonorgestrel/adverse effects , Neoplasm Recurrence, Local/mortality , Polyps/chemically induced , Polyps/epidemiology , Polyps/prevention & control , Randomized Controlled Trials as Topic , Tamoxifen/adverse effects , Uterine Hemorrhage/chemically induced , Uterine Hemorrhage/epidemiology , Uterus/drug effects
7.
Menopause ; 27(4): 410-417, 2020 04.
Article in English | MEDLINE | ID: mdl-31913228

ABSTRACT

OBJECTIVE: The aim of the study was to evaluate the effect of a single-capsule 17ß-estradiol/progesterone (E2/P4), TX-001HR, on endometrial safety, to report on amenorrhea and bleeding patterns of users, and to identify predictors of amenorrhea. METHODS: The REPLENISH trial (NCT01942668) evaluated use of TX-001HR in menopausal women (40-65 y) with vasomotor symptoms (VMS) and a uterus. Women were randomized to daily E2/P4 (mg/mg: 1/100, 0.5/100, 0.5/50, or 0.25/50), or placebo for 12 months. Incidence rate of endometrial hyperplasia was calculated from endometrial biopsies conducted at screening and study completion. Women reported bleeding and spotting in daily diaries. The number of bleeding and/or spotting days and the proportion of women with no bleeding or amenorrhea were compared between treatment and placebo using the Fisher exact test. Predictors of cumulative amenorrhea were assessed by univariate analyses. RESULTS: Women (n = 1,835) who took at least one study dose comprised the safety population; 1,255 had baseline and 12-month biopsies and comprised the endometrial safety population. Incidence of endometrial hyperplasia was ≤0.36% with any dose of TX-001HR after 1 year of use (one-sided upper 95% confidence interval ≤4%). Cumulative amenorrhea (no bleeding/spotting) rates increased over time and were relatively high from cycle 1 to 13 with TX-001HR (56%-73%; placebo 79%; P < 0.05 except with 0.25/50 dose). Few vaginal bleeding adverse events (1.0%-4.6% TX-001HR vs 0.7% placebo) were reported and discontinuations due to bleeding were low (0.4%-1.4% vs 0%). Cumulative amenorrhea was significantly more frequent in older women, those further from their last menstrual period, and those with lower baseline E2 concentrations (all; P < 0.01). CONCLUSIONS: All doses of TX-001HR provided endometrial protection and were associated with an improved bleeding profile over time; older age, further last menstrual period, or lower baseline E2 may predict amenorrhea with TX-001HR.


Subject(s)
Amenorrhea/chemically induced , Endometrial Hyperplasia/prevention & control , Estradiol/administration & dosage , Estrogens/administration & dosage , Progesterone/administration & dosage , Receptors, Progesterone/administration & dosage , Adult , Aged , Drug Combinations , Female , Hot Flashes/drug therapy , Humans , Menopause/physiology , Metrorrhagia/prevention & control , Middle Aged
8.
Int J Gynecol Pathol ; 39(4): 313-320, 2020 Jul.
Article in English | MEDLINE | ID: mdl-31851061

ABSTRACT

Lynch syndrome (LS) is associated with an increased risk for colorectal, endometrial, and ovarian carcinomas in women. Risk-reducing hysterectomy and bilateral salpingo-oophorectomy (RRHSO) has been shown to be a cost-effective form of management and prevention of gynecological malignancies in patients with LS. Studies of incidental gynecologic malignancies identified in RRHSO are limited. In addition, recommendations on optimal handling of this type of specimen have ranged from submitting for microscopic examination the entire endometrium, fallopian tubes and ovaries to submitting only routine representative sections of these organs. In this study, we present the clinicopathologic findings of 29 cases of LS patients that underwent risk-reducing gynecologic surgery at our institution over a period of 13 yr. Clinical-pathologic information was obtained from the patients' charts and pathology reports. Significant pathologic abnormalities were identified in 17% (5/29) of cases, all showing endometrial hyperplasia. Four of them with atypical and 1 without atypical. All of our cases with endometrial pathology had significant findings on preoperative endometrial sampling. To further study the recommendation of in toto submission of the endometrium, ovaries and fallopian tubes and the utility of preoperative endometrial sampling, we undertook a literature review of all the reported cases of incidental pathologic findings identified in RRHSO. The findings of our cohort and the literature reviewed support in toto submission of endometrium, and adnexal structures in the absence of gross lesions. In addition, our findings show a definite benefit for preoperative endometrial sampling as part of the workup for LS patients undergoing RRHSO.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis/surgery , Colorectal Neoplasms/prevention & control , Endometrial Hyperplasia/prevention & control , Endometrial Neoplasms/prevention & control , Ovarian Neoplasms/prevention & control , Adult , Aged , Cohort Studies , Colorectal Neoplasms/etiology , Colorectal Neoplasms/pathology , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , Endometrial Hyperplasia/etiology , Endometrial Hyperplasia/pathology , Endometrial Neoplasms/etiology , Endometrial Neoplasms/pathology , Endometrium/pathology , Endometrium/surgery , Fallopian Tubes/pathology , Fallopian Tubes/surgery , Female , Humans , Hysterectomy , Middle Aged , Neoplasm Staging , Ovarian Neoplasms/etiology , Ovarian Neoplasms/pathology , Ovary/pathology , Ovary/surgery , Prophylactic Surgical Procedures , Risk , Salpingo-oophorectomy
9.
J Vet Sci ; 20(4): e39, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31364324

ABSTRACT

Cystic endometrial hyperplasia-pyometra complex (CEH/P) is a challenge in canine reproduction. Present study aimed to assess fertility after medical treatment. One-hundred-seventy-four bitches affected by CEH/P received aglepristone on days 1, 2, 8, then every 7 days until blood progesterone < 1.2 ng/mL; cloprostenol was administered on days 3 to 5. Records were grouped according to bodyweight (BW): small (< 10 kg, n = 33), medium (10 ≥ BW < 25 kg, n = 44), large (25 ≥ BW < 40 kg, n = 52), and giant bitches (BW ≥ 40 kg, n = 45). Age; success rate; aglepristone treatments number; relapse, pregnancy rates; diagnosis-relapse, -first, -last litter intervals; litters number after treatment, and LS were analyzed by ANOVA. Overall age was 5.14 ± 1.75 years, without difference among groups. Treatment was 100% successful, without difference in treatments number (4.75 ± 1.18), relapse (15/174, 8.62%) and pregnancy (129/140 litters, 92.14%) rates, intervals diagnosis-relapse (409.63 ± 254.9 days) or -last litter (418.62 ± 129.03 days). The interval diagnosis-first litter was significantly shorter (163.52 ± 51.47 days) and longer (225.17 ± 90.97 days) in small and giant bitches, respectively. Overall, 1.47 ± 0.65 litters were born after treatment. Expected LS was achieved in each group, as shown by ΔLS (actual-expected LS by breed, overall -0.40 ± 1.62) without differences among groups. Concluding, CEH/P affects younger dogs than previously described. Relapses were rarer than previously reported. Medical treatment with aglepristone+cloprostenol is effective and safe, preserving subsequent fertility, as demonstrated by negligible changes in LS.


Subject(s)
Dog Diseases/prevention & control , Endometrial Hyperplasia/veterinary , Fertility , Litter Size , Luteolytic Agents/therapeutic use , Pregnancy Rate , Pyometra/veterinary , Animals , Cloprostenol/therapeutic use , Dog Diseases/physiopathology , Dogs , Endometrial Hyperplasia/physiopathology , Endometrial Hyperplasia/prevention & control , Estrenes/therapeutic use , Female , Pregnancy , Pyometra/physiopathology , Pyometra/prevention & control , Recurrence
10.
Int Immunopharmacol ; 62: 326-333, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30056375

ABSTRACT

OBJECTIVE: To study the possible protective role of montelukast in endometrial hyperplesia (EH) rat model, induced by estradiol valerate (EV). METHODS/MATERIALS: Thirty six female albino Wistar rats were classified into 7 groups: normal control, EV (2 mg/kg/day, p.o.), montelukast (10 mg/kg/day, p.o.), montelukast (1 mg/kg/day, p.o.) + EV (2 mg/kg/day, p.o.), montelukast (10 mg/kg/day, p.o.) + EV (2 mg/kg/day, p.o.), montelukast (20 mg/kg/day, p.o.) + EV (2 mg/kg/day, p.o.) groups. Uterine malondialdehyde (MDA), superoxide dismutase (SOD), total nitrites (NO) and serum total antioxidant capacity (TAC) were determined. Uterine, serum total cholesterol, high density lipoprotein (HDL) and tumor necrosis factor (TNF)-α were measured. Histopathological examination of the uterine tissue was also done. In addition, immunohistochemistry was done using Phosphatase and tensin homolog (PTEN) and inducible nitric oxide synthase (iNOS) antibodies. RESULTS: Our results showed that montelukast in dose dependant manner improves oxidative stress, lipids profile and TNF α which were affected by EV. Moreover, immunohistochemical examination revealed that montelukast markedly reduced iNOS expression, while expression of PTEN was markedly enhanced, as compared to EV group. The protective effects of montelukast were also verified histopathologically. CONCLUSIONS: Montelukast in dose dependant manner provided biochemical and histo-pathological improvement in EV induced EH, through its anti-inflammatory, antioxidant activity and inhibition of iNOS expression with induction of PTEN expression.


Subject(s)
Acetates/pharmacology , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Endometrial Hyperplasia/prevention & control , Quinolines/pharmacology , Animals , Cholesterol/metabolism , Cyclopropanes , Disease Models, Animal , Dose-Response Relationship, Drug , Endometrial Hyperplasia/immunology , Endometrial Hyperplasia/pathology , Estradiol/pharmacology , Female , Nitric Oxide Synthase Type II/antagonists & inhibitors , Oxidative Stress/drug effects , Rats , Rats, Wistar , Sulfides , Tumor Necrosis Factor-alpha/blood
11.
Climacteric ; 21(4): 315-320, 2018 08.
Article in English | MEDLINE | ID: mdl-29790373

ABSTRACT

Progesterone is a steroid hormone that is essential for the regulation of reproductive function. Progesterone has been approved for several indications including the treatment of anovulatory menstrual cycles, assisted reproductive technology, contraception during lactation and, when combined with estrogen, for the prevention of endometrial hyperplasia in postmenopausal hormonal therapy. In addition to its role in reproduction, progesterone regulates a number of biologically distinct processes in other tissues, particularly in the nervous system. This physiological hormone is poorly absorbed when administered in a crystalline form and is not active when given orally, unless in micronized form, or from different non-oral delivery systems that allow a more constant delivery rate. A limited number of preclinical studies have been conducted to document the toxicity, carcinogenicity and overall animal safety of progesterone delivered from different formulations, and these rather old studies showed no safety concern. More recently, it has been shown in animal experiments that progesterone, its metabolite allopregnanolone and structurally related progestins have positive effects on neuroregeneration and repair of brain damage, as well as myelin repair. These recent preclinical findings have the potential to accelerate therapeutic translation for multiple unmet neurological needs.


Subject(s)
Brain/drug effects , Endometrial Hyperplasia/prevention & control , Progesterone/pharmacology , Progestins/pharmacology , Animals , Brain/metabolism , Endometrial Hyperplasia/chemically induced , Estrogens/adverse effects , Female , Humans , Models, Animal , Progesterone/metabolism , Progesterone/toxicity , Progestins/metabolism , Progestins/toxicity
12.
Climacteric ; 21(5): 454-461, 2018 10.
Article in English | MEDLINE | ID: mdl-29526116

ABSTRACT

Major advances in menopause hormone therapy (MHT) hold promise in the future of better and safer care for women at and after the menopause. The principal advances are: (1) the critical window or 'window of opportunity' in the 10 years or so after the menopause, during which the benefits of MHT in healthy women exceed any risks; (2) use of transdermal instead of oral administration of estrogen to reduce the risk of venous thromboembolism; (c) investigation of the use of oral micronized progesterone (MP) and vaginal MP to prevent endometrial hyperplasia and carcinoma without any increased risk of breast cancer and venous thromboembolism in postmenopausal women receiving estrogens; vaginal MP prevents endometrial proliferation in the short term but the long-term effects in MHT remain to be established; (4) investigation into the use of intrauterine levonorgestrel-releasing devices (LNG-IUDs), which are an attractive form of MHT in perimenopausal women, providing contraception and reducing uterine bleeding, although the risk of breast cancer with LNG-IUDs requires clarification. Women in the future can look forward to a symptom-free menopause and to safer and more beneficial MHT.


Subject(s)
Endometrial Hyperplasia/prevention & control , Hormone Replacement Therapy/trends , Menopause/drug effects , Venous Thromboembolism/prevention & control , Administration, Cutaneous , Administration, Oral , Endometrial Hyperplasia/chemically induced , Estrogens/administration & dosage , Female , Hormone Replacement Therapy/adverse effects , Humans , Intrauterine Devices, Medicated , Levonorgestrel/administration & dosage , Progesterone/administration & dosage , Progestins/administration & dosage , Venous Thromboembolism/chemically induced
13.
Oncotarget ; 8(5): 7265-7275, 2017 Jan 31.
Article in English | MEDLINE | ID: mdl-27980219

ABSTRACT

During aging, uncontrolled epithelial cell proliferation in the uterus results in endometrial hyperplasia and/or cancer development. The mTOR signaling pathway is one of the major regulators of aging as suppression of this pathway prolongs lifespan in model organisms. Genetic alterations in this pathway via mutations and/or amplifications are often encountered in endometrial cancers. However, the exact contribution of mTOR signaling and uterine aging to endometrial pathologies is currently unclear. This study examined the role of mTOR signaling in uterine aging and its implications in the development of endometrial hyperplasia. The hyperplastic endometrium of both postmenopausal women and aged mice exhibited elevated mTOR activity as seen with increased expression of the pS6 protein. Analysis of uteri from Pten heterozygous and Pten overexpressing mice further confirmed that over-activation of mTOR signaling leads to endometrial hyperplasia. Pharmacological inhibition of mTOR signaling using rapamycin treatment suppressed endometrial hyperplasia in aged mice. Furthermore, treatment with mTOR inhibitors reduced colony size and proliferation of a PTEN negative endometrial cancer cell line in 3D culture. Collectively, this study suggests that hyperactivation of the mTOR pathway is involved in the development of endometrial hyperplasia in aged women and mice.


Subject(s)
Cell Proliferation , Endometrial Hyperplasia/enzymology , Endometrium/enzymology , Epithelial Cells/enzymology , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Endometrial Hyperplasia/genetics , Endometrial Hyperplasia/pathology , Endometrial Hyperplasia/prevention & control , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/enzymology , Endometrial Neoplasms/pathology , Endometrium/drug effects , Endometrium/pathology , Epithelial Cells/drug effects , Epithelial Cells/pathology , Female , Genetic Predisposition to Disease , Humans , Mice, Inbred C57BL , Mice, Knockout , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phenotype , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Sirolimus/pharmacology , Spheroids, Cellular , TOR Serine-Threonine Kinases/antagonists & inhibitors
14.
Oncology ; 92(2): 109-114, 2017.
Article in English | MEDLINE | ID: mdl-27931017

ABSTRACT

OBJECTIVES: Obesity is a major risk factor for endometrial cancer. We evaluated whether obesity exacerbates progression of endometrial hyperplasia (EH) using the PRCre/+ PTENflox/+ mouse model and examined if the type 2 diabetes drug, metformin, could prevent EH. METHODS: Twenty obese (PRCre/+ PTENflox/+) mice were maintained on a high-fat diet, while 20 lean mice ate a matching low-fat diet. Ten mice from each group received metformin (1,000 mg/day) in drinking water. Mice were euthanized at 26 weeks. Uterine tissue was scored for degree of EH. Immunohistochemical staining for Ki67 was used to evaluate cellular proliferation. Markers of PI3K/AKT/mTOR activity were evaluated by immunohistochemistry using activation-specific antibodies. Serum adiponectin was quantified by ELISA. RESULTS: Obesity had no effect on the extent of EH in (PRCre/+ PTENflox/+) mice. While metformin significantly altered circulating adiponectin levels in obese and lean animals, it had no effect on EH. There were no differences in endometrial proliferation as measured by Ki67 staining. Neither obesity nor metformin altered PI3K/AKT/mTOR activity in these animals. CONCLUSIONS: Weight and metformin did not affect the severity of EH resulting from PTEN inactivation. Alternative mouse models of early endometrial cancer are required for preclinical cancer prevention studies.


Subject(s)
Body Weight , Endometrial Hyperplasia/prevention & control , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Mutation , PTEN Phosphohydrolase/genetics , Adiponectin/metabolism , Animals , Endometrial Hyperplasia/genetics , Female , Mice
15.
Eur J Obstet Gynecol Reprod Biol ; 207: 220-226, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27773356

ABSTRACT

One of the most effective methods to tackle obesity and its related comorbidities is bariatric surgery. Polycystic ovary syndrome (PCOS) and endometrial hyperplasia (EH), which are associated with increased risk of endometrial carcinoma, have been identified as potentially new indications for bariatric surgery. PCOS is the most common endocrine disorder in women in the reproductive age and is associated with several components of the metabolic syndrome such as obesity, insulin resistance and hypertension. EH is a pre-cancerous condition which arises in the presence of chronic exposure to estrogen unopposed by progesterone such as both in PCOS and obesity. The main bariatric procedures are Roux-en-Y gastric bypass, laparoscopic sleeve gastrectomy and laparoscopic adjustable gastric banding. These procedures are well established and when correctly selected and performed by experienced bariatric surgeons, they can achieve significant weight loss and remission of obesity related co-morbidities. Studies have shown that bariatric surgery can play an important role in the management of patients with PCOS and improve fertility. Similarly, bariatric surgery has a positive effect on endometrial hyperplasia, making surgically induced weight loss a potentially attractive option for endometrial cancer prevention and treatment. Obesity has an adverse impact on spontaneous pregnancy, assisted reproduction methods and feto-maternal outcomes. After bariatric surgery obese women with subfertility can achieve spontaneous pregnancy. However, while bariatric surgery reduces the risk of pre-eclampsia and gestational diabetes, there is an increased risk of small for gestational age and possible increased risk of stillborn or neonatal death. In this article we will review the evidence regarding the use of bariatric surgery as a treatment modality in patients with PCOS and EH. We also provide an overview of the common bariatric procedures.


Subject(s)
Bariatric Surgery , Endometrial Hyperplasia/prevention & control , Evidence-Based Medicine , Infertility, Female/prevention & control , Obesity, Morbid/surgery , Polycystic Ovary Syndrome/prevention & control , Adult , Combined Modality Therapy , Comorbidity , Endometrial Hyperplasia/epidemiology , Endometrial Hyperplasia/physiopathology , Female , Healthy Lifestyle , Humans , Infertility, Female/etiology , Infertility, Female/therapy , Obesity/epidemiology , Obesity/physiopathology , Obesity/surgery , Obesity/therapy , Obesity, Morbid/epidemiology , Obesity, Morbid/physiopathology , Obesity, Morbid/therapy , Polycystic Ovary Syndrome/epidemiology , Polycystic Ovary Syndrome/physiopathology , Pregnancy , Reproductive Techniques, Assisted , Weight Loss
16.
J Appl Toxicol ; 36(6): 769-76, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26178146

ABSTRACT

Sulpiride and ethylene glycol monomethyl ether (EGME) are known ovarian toxicants that stimulate prolactin (PRL) secretion, resulting in hypertrophy of the corpora lutea and increased progesterone (P4) production. The purpose of the present study was to investigate how the PRL stimulatory agents affected uterine carcinogenesis and to clarify the effects of PRL on endometrial adenocarcinoma progression in rats. Ten-week-old female Donryu rats were treated once with N-ethyl-N'-nitro-N-nitrosoguanidine (20 mg kg(-1) ), followed by treatment with sulpiride (200 ppm) or EGME (1250 ppm) from 11 weeks of age to 12 months of age. Sulpiride treatment inhibited the incidence of uterine adenocarcinoma and precancerous lesions of atypical endometrial hyperplasia, whereas EGME had no effect on uterine carcinogenesis. Sulpiride markedly prevented the onset of persistent estrus throughout the study period, and EGME delayed and inhibited the onset of persistent estrus. Moreover, sulpiride-treated animals showed high PRL and P4 serum levels without changes in the levels of estradiol-17ß, low uterine weights and histological luteal cell hypertrophy. EGME did not affect serum PRL and P4 levels. These results suggest that the prolonged low estradiol-17ß to P4 ratio accompanied by persistent estrous cycle abnormalities secondary to the luteal stimulatory effects of PRL may explain the inhibitory effects of sulpiride on uterine carcinogenesis in rats. Copyright © 2015 John Wiley & Sons, Ltd.


Subject(s)
Adenocarcinoma/prevention & control , Anticarcinogenic Agents/therapeutic use , Carcinogenesis/drug effects , Endometrial Neoplasms/prevention & control , Ethylene Glycols/therapeutic use , Prolactin/agonists , Sulpiride/therapeutic use , Adenocarcinoma/blood , Adenocarcinoma/chemically induced , Adenocarcinoma/pathology , Animals , Anticarcinogenic Agents/adverse effects , Carcinogenesis/chemically induced , Carcinogens/chemistry , Carcinogens/toxicity , Endometrial Hyperplasia/blood , Endometrial Hyperplasia/chemically induced , Endometrial Hyperplasia/pathology , Endometrial Hyperplasia/prevention & control , Endometrial Neoplasms/blood , Endometrial Neoplasms/chemically induced , Endometrial Neoplasms/pathology , Endometrium/drug effects , Endometrium/pathology , Estrus/drug effects , Ethylene Glycols/adverse effects , Female , Infertility, Female/blood , Infertility, Female/chemically induced , Infertility, Female/pathology , Infertility, Female/prevention & control , Methylnitronitrosoguanidine/analogs & derivatives , Methylnitronitrosoguanidine/chemistry , Methylnitronitrosoguanidine/toxicity , Organ Size/drug effects , Ovary/drug effects , Ovary/pathology , Precancerous Conditions/blood , Precancerous Conditions/chemically induced , Precancerous Conditions/pathology , Precancerous Conditions/prevention & control , Progesterone/agonists , Progesterone/blood , Progesterone/metabolism , Prolactin/blood , Prolactin/metabolism , Rats, Inbred Strains , Sulpiride/adverse effects , Uterus/drug effects , Uterus/pathology , Weight Gain/drug effects
17.
Cochrane Database Syst Rev ; (12): CD007245, 2015 Dec 09.
Article in English | MEDLINE | ID: mdl-26649916

ABSTRACT

BACKGROUND: Adjuvant tamoxifen reduces the risk of breast cancer recurrence in women with oestrogen receptor-positive breast cancer. Tamoxifen also increases the risk of postmenopausal bleeding, endometrial polyps, hyperplasia, and endometrial cancer. The levonorgestrel-releasing intrauterine system (LNG-IUS) causes profound endometrial suppression. This systematic review considered the evidence that the LNG-IUS prevents the development of endometrial pathology in women taking tamoxifen as adjuvant endocrine therapy for breast cancer. OBJECTIVES: To determine the effectiveness and safety of levonorgestrel intrauterine system (LNG-IUS) in pre- and postmenopausal women taking adjuvant tamoxifen following breast cancer for the outcomes of endometrial and uterine pathology including abnormal vaginal bleeding or spotting, and secondary breast cancer events. SEARCH METHODS: We searched the following databases: Cochrane Menstrual Disorders and Subfertility Group Specialised Register (MDSG), Cochrane Breast Cancer Group Specialised Register (CBCG), Cochrane Central Register of Controlled Trials (CENTRAL), Cochrane Database of Abstracts of Reviews of Effects (DARE), The Cochrane Library, clinicaltrials.gov, The World Health Organisation International Trials Registry, ProQuest Dissertations & Theses, MEDLINE, EMBASE, CINAHL (Cumulative Index to Nursing and Allied Health Literature), PsycINFO, Web of Science, OpenGrey, LILACS, PubMed, and Google. The final search was performed in October 2015. SELECTION CRITERIA: Randomised controlled trials of women with breast cancer on adjuvant tamoxifen that compared endometrial surveillance alone (control condition) versus the LNG-IUS with endometrial surveillance (experimental condition) on the incidence of endometrial pathology. DATA COLLECTION AND ANALYSIS: Study selection, risk of bias assessment and data extraction were performed independently by two review authors. The primary outcome measure was endometrial pathology (including polyps, endometrial hyperplasia, or endometrial cancer) diagnosed at hysteroscopy or endometrial biopsy. Secondary outcome measures included fibroids, abnormal vaginal bleeding or spotting, breast cancer recurrence, and breast cancer-related deaths. The overall quality of evidence was rated using GRADE methods. MAIN RESULTS: Four randomised controlled trials involving 543 women were identified and are included in this review. In the included studies, the active treatment arm was the 20 µg/day levonorgestrel-releasing intrauterine system (LNG-IUS) plus endometrial surveillance; the control arm was endometrial surveillance alone. In tamoxifen users, the LNG-IUS led to a reduction in the incidence of endometrial polyps over both a 12-month period (Peto OR 0.22, 95% CI 0.08 to 0.64, 2 studies, n = 212, I² = 0%) and over a long-term follow-up period (24 to 60 months) (Peto OR 0.22, 95% CI 0.13 to 0.39, 4 studies, n = 417, I² = 0%, moderate quality evidence). Also the LNG-IUS led to a reduction in the incidence of endometrial hyperplasia over a long-term follow-up period (24 to 60 months) (Peto OR 0.13, 95% CI 0.03 to 0.67, four studies, n = 417, I² = 0%, moderate quality evidence). However, it should be noted that the number of events of endometrial hyperplasia was low (n = 6). None of the trials were sufficiently powered to detect whether LNG-IUS leads to significant changes in the incidence of endometrial cancer in tamoxifen users. At 12 months of follow-up abnormal vaginal bleeding or spotting was more common in the LNG-IUS treatment group (Peto OR 7.26, 95% CI 3.37 to 15.66, 3 studies, n = 376, I² = 0%, moderate quality evidence). By 24 months of follow-up, abnormal vaginal bleeding or spotting occurred less frequently compared to 12 months of follow-up in the LNG-IUS treatment group but was still more common than the control group (Peto OR 2.72, 95% CI 1.04 to 7.10, 2 studies, n = 233, I² = 0%, moderate quality evidence). By 60 months of follow-up, no cases of abnormal vaginal bleeding or spotting were reported in either group. The numbers of events for the following outcomes were low: fibroids (n = 13), breast cancer recurrence (n = 18), and breast cancer-related deaths (n = 16). There was no evidence of a difference between the LNG-IUS treatment group and controls for these outcomes. The quality of the evidence was judged as moderate, due to limited sample sizes and low event rates for the outcome comparisons. AUTHORS' CONCLUSIONS: The LNG-IUS reduces the incidence of benign endometrial polyps and endometrial hyperplasia in women with breast cancer taking tamoxifen. At 12 and 24 months of follow-up, the LNG-IUS increased abnormal vaginal bleeding or spotting among women in the treatment group compared to those in the control. There is no clear evidence from the available randomised controlled trials that the LNG-IUS prevents endometrial cancer in these women. There is no clear evidence from the available randomised controlled trials that the LNG-IUS affects the risk of breast cancer recurrence or breast cancer-related deaths. Larger studies are necessary to assess the effects of the LNG-IUS on the incidence of endometrial cancer, and to determine whether the LNG-IUS might have an impact on the risk of secondary breast cancer events.


Subject(s)
Breast Neoplasms/prevention & control , Endometrial Hyperplasia/prevention & control , Endometrial Neoplasms/prevention & control , Intrauterine Devices, Medicated , Levonorgestrel/administration & dosage , Adenocarcinoma/chemically induced , Adenocarcinoma/prevention & control , Antineoplastic Agents, Hormonal/adverse effects , Chemotherapy, Adjuvant , Contraceptive Agents, Female/administration & dosage , Endometrial Hyperplasia/chemically induced , Endometrial Neoplasms/chemically induced , Female , Humans , Neoplasm Recurrence, Local/prevention & control , Polyps/chemically induced , Polyps/prevention & control , Randomized Controlled Trials as Topic , Tamoxifen/adverse effects
18.
Ginekol Pol ; 86(4): 305-10, 2015 Apr.
Article in Polish | MEDLINE | ID: mdl-26117991

ABSTRACT

The use of hormone-releasing intrauterine devices has been on the increase for the last three decades. To date, evidence of their long-term efficiency is available. The aim of the present paper was to briefly review beneficial prophylactic effects of the levonorgestrel-releasing intrauterine system on the incidence of a variety of malignancies in women. Such an influence is of a particular importance in the light of the currently observed increased prevalence of endometrial and cervical adenocarcinomas. Low-dose releasing intrauterine systems are also available, but the hard evidence-based medical data have been derived primarily for Mirena® (Bayer) device, which topically releases from 20 to 14 pg of levonorgestrel daily. Consequently the risk of developing endometrial carcinoma in Mirena® users is lowered by as much as 50% compared with the general population risk To a lesser extent, the intrauterine system decreases the risk for cervical adenocarcinoma and squamous cell carcinoma, as well as ovarian, pancreas, and lung carcinomas. In one population-based study Mirena® increased the risk for breast carcinoma by approximately 20%, whereas a number of other studies failed to demonstrate such a hazard. In the recent decades of the increased predominance of insulin resistance and obesity and an occurrence of hormone-dependent carcinomas at earlier age, a broad application of levonorgestrel-releasing intrauterine systems may become a particularly important component of primary prevention of malignancies in women. Both obese and overweight patients seem perfect candidates for such a hormonal intervention.


Subject(s)
Carcinoma, Endometrioid/prevention & control , Contraceptive Agents, Female/adverse effects , Endometrial Neoplasms/prevention & control , Intrauterine Devices, Medicated , Levonorgestrel/administration & dosage , Adenocarcinoma/prevention & control , Endometrial Hyperplasia/prevention & control , Evidence-Based Medicine , Female , Humans
19.
Rev Med Chir Soc Med Nat Iasi ; 119(1): 154-61, 2015.
Article in English | MEDLINE | ID: mdl-25970959

ABSTRACT

AIM: Metaplasia is defined as a transformation of an adult epithelial or conjunctive cellular type into another adult cellular type. Endometrial hyperplasia and particularly complex atypical hyperplasia exposes to a high risk of development of the endometrial carcinoma, being considered a lesion precursory to the same. Endometrial hyperplasias are risk factors for the development of endometrial carcinoma and their prophylaxis implies their accurate diagnosis, taking into account that the adenocarcinomas diagnosed in an advanced stage, whose therapeutic context differs from the early stages, have a much lower chance of survival. MATERIAL AND METHODS: Our study aimed at highlighting objective criteria in establishing the morphological diagnosis and in evaluating the prognostic elements. The studied batch included 875 patients with endometrial hyperplasia and 263 patients with endometrial adenocarcinoma, who were admitted between 2003 and 2007, and the histopathologic diagnosis was obtained by processing the hysterectomy pieces. The presence of this tumour was at its highest level half-way through the study, which was in 2005. RESULTS: According to the study, there was a higher proportion of patients with endometrial carcinoma from the urban environment (58.2%) than the ones from the rural environment (only 41.8%). Depending on their age, most cases of endometrial adenocarcinoma were diagnosed in 53-year old patients, with an average age of 58.94 years. Our study, made of the two batches of endometrial adenocarcinomas, shows that between the endometrial and non-endometrial adenocarcinoma there are significant differences related to the patients' age, the morphological aspect of the carcinoma, the architectural degree, the nuclear degree of tumours and the invasion in the myometrium. CONCLUSIONS: Our study proves that endometrial hyperplasia is a frequent diagnosis in peri- and postmenopausal patients and is frequently identified following investigations for an abnormal uterine bleeding. The age of patients with endometrial carcinoma is an important prognostic factor independent of other parameters. The difference between complex hyperplasia with no atypias and complex hyperplasia with atypias is important, because atypical complex hyperplasia is considered the precursor of endometrial adenocarcinoma.


Subject(s)
Adenocarcinoma/epidemiology , Endometrial Hyperplasia/epidemiology , Endometrial Neoplasms/epidemiology , Rural Population/statistics & numerical data , Urban Population/statistics & numerical data , Adenocarcinoma/pathology , Adult , Aged , Endometrial Hyperplasia/pathology , Endometrial Hyperplasia/prevention & control , Endometrial Neoplasms/pathology , Female , Humans , Incidence , Middle Aged , Precancerous Conditions , Prognosis , Retrospective Studies , Risk Factors , Romania/epidemiology
20.
Endocr J ; 62(6): 531-42, 2015.
Article in English | MEDLINE | ID: mdl-25877295

ABSTRACT

Tissue-selective estrogen complex (TSEC), which combines a selective estrogen receptor modulator (SERM) with one or more estrogens, is a novel approach to menopausal therapy. It has been demonstrated that the phytoestrogen genistein (GEN) exhibits mixed estrogen receptor agonist and antagonist activity, suggesting that GEN may have potential for use as a natural SERM. We evaluated, for the first time, the effects of GEN, conjugated estrogens (CE), and their pairing effects as a TSEC treatment on estrogen-induced endometrial hyperplasia and metabolic dysfunction in ovariectomized (OVX) mice fed a high-fat diet. CE replacement prevented fat accumulation in the adipose tissue and liver, improved glucose homeostasis, and induced endometrial hyperplasia in OVX mice. GEN at 100 mg/kg showed CE mimetic effects in preventing ovariectomy-induced metabolic dysfunctions without endometrial stimulation. Combination treatments with CE and GEN prevented metabolic dysfunctions more strongly than CE alone, but at both low and high doses, GEN did not reverse CE-induced endometrial hyperplasia. In addition, we found that in a TSEC regimen, a typical SERM raloxifene maintains the metabolic benefits of CE while simultaneously protecting the endometrium in OVX mice. These findings indicate that GEN acts as an estrogen agonist in metabolic regulation, but has no SERM function in the uteri of OVX mice.


Subject(s)
Dietary Supplements , Endometrial Hyperplasia/prevention & control , Estrogen Replacement Therapy , Estrogens, Conjugated (USP)/therapeutic use , Genistein/therapeutic use , Glucose Intolerance/prevention & control , Phytoestrogens/therapeutic use , Adiposity/drug effects , Animals , Diet, High-Fat/adverse effects , Dietary Supplements/adverse effects , Endometrial Hyperplasia/chemically induced , Endometrial Hyperplasia/metabolism , Endometrial Hyperplasia/pathology , Endometrium/drug effects , Endometrium/metabolism , Endometrium/pathology , Estrogen Replacement Therapy/adverse effects , Estrogens/adverse effects , Estrogens/therapeutic use , Estrogens, Conjugated (USP)/adverse effects , Female , Genistein/administration & dosage , Genistein/adverse effects , Glucose Intolerance/chemically induced , Glucose Intolerance/metabolism , Glucose Intolerance/pathology , Liver/drug effects , Liver/metabolism , Liver/pathology , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/prevention & control , Ovariectomy/adverse effects , Overweight/etiology , Overweight/metabolism , Overweight/pathology , Overweight/prevention & control , Phytoestrogens/administration & dosage , Phytoestrogens/adverse effects , Raloxifene Hydrochloride/adverse effects , Raloxifene Hydrochloride/therapeutic use , Random Allocation , Selective Estrogen Receptor Modulators/adverse effects , Selective Estrogen Receptor Modulators/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL