Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35.184
Filter
1.
J Vis Exp ; (208)2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38975749

ABSTRACT

Embryo implantation is the first step in the establishment of a successful pregnancy. An in vitro model for embryo implantation is critical for basic biological research, drug development, and screening. This paper presents a simple, rapid, and highly efficient in vitro model for embryo implantation. In this protocol, we first introduce mouse blastocyst acquisition and human endometrial adenocarcinoma cells (Ishikawa) preparation for implantation, followed by the co-culture method for mouse embryos and Ishikawa cells. Finally, we conducted a study to assess the impact of varying concentrations of 17-ß-estradiol (E2) and progesterone (P4) on embryo adhesion rates based on this model. Our findings revealed that high concentrations of E2 significantly reduced embryo adhesion, whereas the addition of progesterone could restore the adhesion rate. This model offers a simple and fast platform for evaluating and screening molecules involved in the adhesion process, such as cytokines, drugs, and transcription factors controlling implantation and endometrial receptivity.


Subject(s)
Coculture Techniques , Embryo Implantation , Estradiol , Progesterone , Embryo Implantation/physiology , Embryo Implantation/drug effects , Female , Animals , Mice , Humans , Coculture Techniques/methods , Progesterone/pharmacology , Estradiol/pharmacology , Cell Line, Tumor , Blastocyst/cytology , Blastocyst/drug effects , Pregnancy , Endometrial Neoplasms/pathology
4.
Cells ; 13(13)2024 Jun 22.
Article in English | MEDLINE | ID: mdl-38994940

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.


Subject(s)
2-Methoxyestradiol , Apoptosis , Tumor Suppressor Protein p53 , 2-Methoxyestradiol/pharmacology , Tumor Suppressor Protein p53/metabolism , Apoptosis/drug effects , Cell Proliferation/drug effects , Animals , Oligodendrocyte Precursor Cells/metabolism , Oligodendrocyte Precursor Cells/drug effects , Signal Transduction/drug effects , Humans , Estradiol/pharmacology , Estradiol/analogs & derivatives , Oligodendroglia/metabolism , Oligodendroglia/drug effects , Antimitotic Agents/pharmacology , Cell Line
5.
Cells ; 13(13)2024 Jun 29.
Article in English | MEDLINE | ID: mdl-38994978

ABSTRACT

We report a three-pronged phenotypic evaluation of the bioprecursor prodrug 10ß,17ß-dihydroxyestra-1,4-dien-3-one (DHED) that selectively produces 17ß-estradiol (E2) in the retina after topical administration and halts glaucomatous neurodegeneration in a male rat model of the disease. Ocular hypertension (OHT) was induced by hyperosmotic saline injection into an episcleral vein of the eye. Animals received daily DHED eye drops for 12 weeks. Deterioration of visual acuity and contrast sensitivity by OHT in these animals were markedly prevented by the DHED-derived E2 with concomitant preservation of retinal ganglion cells and their axons. In addition, we utilized targeted retina proteomics and a previously established panel of proteins as preclinical biomarkers in the context of OHT-induced neurodegeneration as a characteristic process of the disease. The prodrug treatment provided retina-targeted remediation against the glaucomatous dysregulations of these surrogate endpoints without increasing circulating E2 levels. Collectively, the demonstrated significant neuroprotective effect by the DHED-derived E2 in the selected animal model of glaucoma supports the translational potential of our presented ocular neuroprotective approach owing to its inherent therapeutic safety and efficacy.


Subject(s)
Disease Models, Animal , Estradiol , Glaucoma , Prodrugs , Retinal Ganglion Cells , Animals , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Retinal Ganglion Cells/metabolism , Glaucoma/drug therapy , Glaucoma/pathology , Glaucoma/metabolism , Prodrugs/pharmacology , Estradiol/pharmacology , Male , Rats , Retina/drug effects , Retina/pathology , Retina/metabolism , Vision, Ocular/drug effects , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use
6.
Sci Rep ; 14(1): 15736, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38977751

ABSTRACT

Benign Prostatic Hyperplasia (BPH) is a complex condition leading to Lower Urinary Tract Symptoms in aging men, characterized by cellular proliferation, smooth muscle dysfunction, inflammation, and fibrosis. While BPH is known to involve heightened macrophage infiltration, the specific contribution of infiltrating monocytes/macrophages to the disease mechanism remains uncertain. This research explores the impact of reducing circulating monocytes and subsequently limiting their tissue infiltration by using Ccr2 knockout (Ccr2-KO) mice. Ccr2-KO and wild type mice were implanted with testosterone and estradiol (T + E2, 25 mg + 2.5 mg) pellets. Urinary function was assessed via weekly void spot assays over 12 weeks, and prostatic macrophage levels were visualized and quantified in tissue sections using an F4/80 antibody. Additionally, Ki-67 staining was used to evaluate cell proliferation, and picrosirius red staining to assess collagen accumulation. Increased voiding frequency which developed in T + E2 mice, was significantly ameliorated in Ccr2-KO mice, however, both Ccr2-KO and wild type (WT) mice showed increased bladder weights after three month, representing a hypertrophic response to bladder outlet obstruction. T + E2 substantially increased the density of macrophages in WT but not Ccr2-KO mouse prostate. Proliferation rate, as indicated by Ki-67 positivity, was elevated in the vental and anterior prostate lobes but was only marginally reduced in Ccr2-KO mice. Most importantly, a significant prostatic collagen accumulation was observed in WT mice that was markedly reduced by Ccr2 deficiency post T + E2 treatment. The absence of Ccr2 mitigates urinary dysfunction and alters prostatic macrophage levels and collagen accumulation in steroid hormone imbalance. These findings suggest a crucial role for monocyte infiltration, giving rise to macrophages or other cell derivatives, to drive fibrosis.


Subject(s)
Estradiol , Fibrosis , Macrophages , Mice, Knockout , Monocytes , Prostate , Receptors, CCR2 , Testosterone , Animals , Male , Receptors, CCR2/metabolism , Macrophages/metabolism , Mice , Monocytes/metabolism , Prostate/metabolism , Prostate/pathology , Testosterone/metabolism , Estradiol/metabolism , Estradiol/pharmacology , Prostatic Hyperplasia/metabolism , Prostatic Hyperplasia/pathology , Cell Proliferation , Mice, Inbred C57BL
7.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-39000231

ABSTRACT

Following metastatic spread, many hormone receptor positive (HR+) patients develop a more aggressive phenotype with an observed loss of the HRs estrogen receptor (ER) and progesterone receptor (PR). During metastasis, breast cancer cells are exposed to high magnitudes of fluid shear stress (FSS). Unfortunately, the role for FSS on the regulation of HR expression and function during metastasis is not fully understood. This study was designed to elucidate the impact of FSS on HR+ breast cancer. Utilizing a microfluidic platform capable of exposing breast cancer cells to FSS that mimics in situ conditions, we demonstrate the impact of FSS exposure on representative HR+ breast cancer cell lines through protein and gene expression analysis. Proteomics results demonstrated that 540 total proteins and 1473 phospho-proteins significantly changed due to FSS exposure and pathways of interest included early and late estrogen response. The impact of FSS on response to 17ß-estradiol (E2) was next evaluated and gene expression analysis revealed repression of ER and E2-mediated genes (PR and SDF1) following exposure to FSS. Western blot demonstrated enhanced phosphorylation of mTOR following exposure to FSS. Taken together, these studies provide initial insight into the effects of FSS on HR signaling in metastatic breast cancer.


Subject(s)
Breast Neoplasms , Gene Expression Regulation, Neoplastic , Receptors, Estrogen , Receptors, Progesterone , Stress, Mechanical , Humans , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Female , Receptors, Progesterone/metabolism , Receptors, Progesterone/genetics , Cell Line, Tumor , Receptors, Estrogen/metabolism , Receptors, Estrogen/genetics , Estradiol/pharmacology , Phosphorylation , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Proteomics/methods , MCF-7 Cells , Chemokine CXCL12/metabolism , Chemokine CXCL12/genetics
8.
Int J Mol Sci ; 25(13)2024 Jun 30.
Article in English | MEDLINE | ID: mdl-39000334

ABSTRACT

Systemic sclerosis (SSc) is characterized by dermal fibrosis with a female predominance, suggesting a hormonal influence. Patients with SSc have elevated interleukin (IL)-6 levels, and post-menopausal women and older men also have high estradiol (E2) levels. In the skin, IL-6 increases the enzymatic activity of aromatase, thereby amplifying the conversion of testosterone to E2. Therefore, we hypothesized that an interplay between E2 and IL-6 contributes to dermal fibrosis. We used primary dermal fibroblasts from healthy donors and patients with diffuse cutaneous (dc)SSc, and healthy donor skin tissues stimulated with recombinant IL-6 and its soluble receptor (sIL-6R) or E2. Primary human dermal fibroblasts and tissues from healthy donors stimulated with IL-6+sIL-6R produced E2, while E2-stimulated dermal tissues and fibroblasts produced IL-6. Primary dermal fibroblasts from healthy donors treated with IL-6+sIL-6R and the aromatase inhibitor anastrozole (ANA) and dcSSc fibroblasts treated with ANA produced less fibronectin (FN), type III collagen A1 (Col IIIA1), and type V collagen A1 (Col VA1). Finally, dcSSc dermal fibroblasts treated with the estrogen receptor inhibitor fulvestrant also generated less FN, Col IIIA1, and Col VA1. Our data show that IL-6 exerts its pro-fibrotic influence in human skin in part through E2 and establish a positive feedback loop between E2 and IL-6.


Subject(s)
Estradiol , Fibroblasts , Fibrosis , Interleukin-6 , Scleroderma, Systemic , Humans , Interleukin-6/metabolism , Estradiol/pharmacology , Estradiol/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Scleroderma, Systemic/metabolism , Scleroderma, Systemic/pathology , Female , Male , Skin/metabolism , Skin/pathology , Cells, Cultured , Feedback, Physiological , Middle Aged , Adult , Receptors, Interleukin-6/metabolism
9.
Pharmacol Res Perspect ; 12(4): e1234, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38961539

ABSTRACT

The association of hormonal contraception with increased risk of inflammatory bowel disease (IBD) observed in females suggests involvement of ovarian hormones, such as estradiol, and the estrogen receptors in the progression of intestinal inflammation. Here, we investigated the effects of prophylactic SERM2 and estradiol supplementation in dextran sulfate sodium-induced colitis using mice with intact ovaries and ovariectomized (OVX) female mice. We found that graded colitis score was threefold reduced in the OVX mice, compared to mice with intact ovaries. Estradiol supplementation, however, aggravated the colitis in OVX mice, increasing the colitis score to a similar level than what was observed in the intact mice. Further, we observed that immune infiltration and gene expression of inflammatory interleukins Il1b, Il6, and Il17a were up to 200-fold increased in estradiol supplemented OVX colitis mice, while a mild but consistent decrease was observed by SERM2 treatment in intact animals. Additionally, cyclo-oxygenase 2 induction was increased in the colon of colitis mice, in correlation with increased serum estradiol levels. Measured antagonist properties of SERM2, together with the other results presented here, indicates an exaggerating role of ERα signaling in colitis. Our results contribute to the knowledge of ovarian hormone effects in colitis and encourage further research on the potential use of ER antagonists in the colon, in order to alleviate inflammation.


Subject(s)
Colitis , Dextran Sulfate , Estradiol , Estrogen Receptor alpha , Ovariectomy , Animals , Female , Estrogen Receptor alpha/metabolism , Colitis/chemically induced , Colitis/metabolism , Colitis/drug therapy , Mice , Estradiol/pharmacology , Estradiol/blood , Mice, Inbred C57BL , Estrogens/pharmacology , Cyclooxygenase 2/metabolism , Disease Models, Animal , Interleukin-17/metabolism , Colon/pathology , Colon/drug effects , Colon/metabolism , Interleukin-6/metabolism , Interleukin-1beta/metabolism
10.
Zhongguo Dang Dai Er Ke Za Zhi ; 26(7): 757-764, 2024 Jul 15.
Article in Chinese | MEDLINE | ID: mdl-39014954

ABSTRACT

OBJECTIVES: To investigate the protective effects of 2-methoxyestradiol (2ME) against hypoxic pulmonary hypertension (HPH) in neonatal rats. METHODS: Ninety-six Wistar neonatal rats were randomly divided into a normoxia group, a hypoxia group, and a hypoxia + 2ME group, with each group further subdivided into 3-day, 7-day, 14-day, and 21-day subgroups, containing eight rats each. The hypoxia and hypoxia + 2ME groups received daily subcutaneous injections of saline and 2ME (240 µg/kg), respectively, while the normoxia group was raised in a normoxic environment with daily saline injections. Right ventricular systolic pressure (RVSP) was measured using the direct pressure method. Pulmonary vascular morphology was assessed using hematoxylin and eosin staining, with metrics including the percentage of medial thickness of small pulmonary arteries relative to the external diameter (MT%) and the cross-sectional area of the media of small pulmonary arteries relative to the total cross-sectional area (MA%). Immunohistochemistry was used to detect the expression levels of hypoxia-inducible factor-1α (HIF-1α) and proliferating cell nuclear antigen (PCNA) proteins, while real-time quantitative PCR was used to to assess HIF-1α and PCNA mRNA levels. RESULTS: Compared to the normoxia group, the hypoxia and hypoxia + 2ME groups showed increased RVSP and upregulated HIF-1α and PCNA protein and mRNA expression levels at 3, 7, 14, and 21 days after hypoxia (P<0.05). Furthermore, at 7, 14, and 21 days after hypoxia, the hypoxia group showed increased MT% and MA% (P<0.05). In comparison to the hypoxia group, the hypoxia + 2ME group exhibited reduced RVSP and downregulated HIF-1α and PCNA protein and mRNA expression levels, along with decreased MT% and MA% at 7, 14, and 21 days after hypoxia (P<0.05). CONCLUSIONS: 2ME may protect against HPH in neonatal rats by inhibiting the expression of HIF-1α and PCNA and reducing pulmonary vascular remodeling. Citation:Chinese Journal of Contemporary Pediatrics, 2024, 26(7): 757-764.


Subject(s)
2-Methoxyestradiol , Animals, Newborn , Hypertension, Pulmonary , Hypoxia-Inducible Factor 1, alpha Subunit , Hypoxia , Proliferating Cell Nuclear Antigen , Pulmonary Artery , Rats, Wistar , Animals , 2-Methoxyestradiol/pharmacology , Rats , Hypertension, Pulmonary/prevention & control , Hypertension, Pulmonary/drug therapy , Proliferating Cell Nuclear Antigen/analysis , Proliferating Cell Nuclear Antigen/genetics , Hypoxia/complications , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Pulmonary Artery/drug effects , Male , Female , Estradiol/pharmacology , Estradiol/analogs & derivatives , RNA, Messenger/analysis
12.
Methods Mol Biol ; 2811: 101-112, 2024.
Article in English | MEDLINE | ID: mdl-39037652

ABSTRACT

Here, we describe a clinically relevant xenograft model of hormone receptor-positive breast cancer that maintains estrogen receptor (ER) status without the need for exogenous supplementation of hormones. The naturally low 17-ß-estradiol levels in host mice recapitulate levels seen in post-menopausal women. By introducing breast cancer cells directly into their "natural" microenvironment of the milk ducts, these cells maintain hormone receptor status, model the clinical progression of the disease, and develop ER- metastatic lesions or dormant micrometastatic lesions in the case of ER+ BC. With the use of GFP/RFP:Luc2 reporters, we can monitor in vivo tumour growth and conduct ex vivo metastases assays to evaluate dormant metastatic cell harboring organs. Upon recovery of metastatic cells from ER+ breast cancer models, downstream analyses can be conducted to assess the relationship between epithelial plasticity and metastatic dormancy.


Subject(s)
Breast Neoplasms , Disease Models, Animal , Receptors, Estrogen , Animals , Mice , Female , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Humans , Receptors, Estrogen/metabolism , Neoplasm Metastasis , Cell Line, Tumor , Tumor Microenvironment , Estradiol/metabolism , Estradiol/pharmacology
13.
Biopharm Drug Dispos ; 45(3): 149-158, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38886878

ABSTRACT

Icaritin is a prenylflavonoid derivative of the genus Epimedium (Berberidaceae) and has a variety of pharmacological actions. Icaritin is approved by the National Medical Products Administration as an anticancer drug that exhibits efficacy and safety advantages in patients with hepatocellular carcinoma cells. This study aimed to evaluate the inhibitory effects of icaritin on UDP-glucuronosyltransferase (UGT) isoforms. 4-Methylumbelliferone (4-MU) was employed as a probe drug for all the tested UGT isoforms using in vitro human liver microsomes (HLM). The inhibition potentials of UGT1A1 and 1A9 in HLM were further tested by employing 17ß-estradiol (E2) and propofol (PRO) as probe substrates, respectively. The results showed that icaritin inhibits UGT1A1, 1A3, 1A4, 1A7, 1A8, 1A10, 2B7, and 2B15. Furthermore, icaritin exhibited a mixed inhibition of UGT1A1, 1A3, and 1A9, and the inhibition kinetic parameters (Ki) were calculated to be 3.538, 2.117, and 0.306 (µM), respectively. The inhibition of human liver microsomal UGT1A1 and 1A9 both followed mixed mechanism, with Ki values of 2.694 and 1.431 (µM). This study provides supporting information for understanding the drug-drug interaction (DDI) potential of the flavonoid icaritin and other UGT-metabolized drugs in clinical settings. In addition, the findings provide safety evidence for DDI when liver cancer patients receive a combination therapy including icaritin.


Subject(s)
Drug Interactions , Flavonoids , Glucuronosyltransferase , Microsomes, Liver , Glucuronosyltransferase/antagonists & inhibitors , Glucuronosyltransferase/metabolism , Humans , Flavonoids/pharmacology , Microsomes, Liver/metabolism , Estradiol/pharmacology , Hymecromone/pharmacology , Propofol/pharmacology , Enzyme Inhibitors/pharmacology
14.
Bone ; 186: 117143, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38866125

ABSTRACT

The effects of gender affirming hormone therapy (GAHT) on bone microarchitecture and fracture risk in adult transgender women is unclear. To investigate the concept that skeletal integrity and strength in trans women may be improved by treatment with a higher dose of GAHT than commonly prescribed, we treated adult male mice with a sustained, high dose of estradiol. Adult male mice at 16 weeks of age were administered ~1.3 mg estradiol by silastic implant, implanted intraperitoneally, for 12 weeks. Controls included vehicle treated intact females and males. High-dose estradiol treatment in males stimulated the endocortical deposition of bone at the femoral mid-diaphysis, increasing cortical thickness and bone area. This led to higher stiffness, maximum force, and the work required to fracture the bone compared to male controls, while post-yield displacement was unaffected. Assessment of the material properties of the bone showed an increase in both elastic modulus and ultimate stress in the estradiol treated males. Treatment of male mice with high dose estradiol was also anabolic for trabecular bone, markedly increasing trabecular bone volume, number and thickness in the distal metaphysis which was accompanied by an increase in the histomorphometric markers of bone remodelling, mineralizing surface/bone surface, bone formation rate and osteoclast number. In conclusion, a high dose of estradiol is anabolic for cortical and trabecular bone in a male to female transgender mouse model, increasing both stiffness and strength. These findings suggest that increasing the current dose of GAHT administered to trans women, while considering other potential adverse effects, may be beneficial to preserving their bone microstructure and strength.


Subject(s)
Estradiol , Animals , Male , Estradiol/pharmacology , Estradiol/blood , Female , Mice , Bone and Bones/drug effects , Bone and Bones/diagnostic imaging , Bone Density/drug effects , Anabolic Agents/pharmacology , Organ Size/drug effects , Mice, Inbred C57BL , Humans , Models, Animal , X-Ray Microtomography
15.
Reprod Domest Anim ; 59(6): e14617, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38837282

ABSTRACT

Sub-estrus buffaloes do not exhibit estrus signs despite being cyclic contributing to extended service periods and inter-calving intervals causing significant economic loss. The present study described the effect of synthetic prostaglandin (PGF2α) on estrus behaviour, follicular and luteal morphometry, and serum estradiol (E2) and progesterone (P4) profile in sub-estrus buffaloes during the non-breeding season. The incidence of sub-estrus was 38.4% during the non-breeding season. The sub-estrus buffaloes (n = 33) were divided into two groups, viz., Control (n = 16) and PGF2α treatment (Inj. Cloprostenol 500 µg, i.m., n = 17). Estrus induction response was significantly greater in the treatment (100 vs. 18.75%, p < .001), and a relatively greater proportion of animals conceived in the treatment group (29.41 vs. 6.25%, p = .08). The time elapsed to induction of estrus and insemination following treatment was significantly lower in the treatment group than control. A significant increment in the follicle diameter (9.72 ± 0.45 vs. 13.00 ± 0.45 mm, P < .0001) and serum estradiol (E2) concentration (66.01 ± 11.92 vs. 104.9 ± 13.21 pg/mL, p = .003) observed at the post-treatment period in the PGF2α treatment group. At the same time, CL diameter was reduced significantly at a higher regression rate in the PGF2α treated buffaloes than those of control. Of the responded buffaloes, only 30% showed high-intensity estrus attributed to the expulsion of cervico-vaginal mucus (CVM), uterine tonicity, micturition, and mounting response by a teaser bull. From this study, it can be concluded that the administration of PGF2α could induce estrus in the sub-estrus buffaloes during the non-breeding season. Behavioural changes, along with sonographic observation of POF, regressing CL, and serum E2 and P4 concentration would be useful to determine the right time of insemination in sub-estrus buffaloes during non-breeding season.


Subject(s)
Buffaloes , Dinoprost , Estradiol , Estrus Synchronization , Estrus , Ovarian Follicle , Progesterone , Animals , Female , Buffaloes/physiology , Estradiol/pharmacology , Estradiol/blood , Progesterone/blood , Progesterone/pharmacology , Estrus/drug effects , Ovarian Follicle/drug effects , Dinoprost/pharmacology , Dinoprost/administration & dosage , Pregnancy , Seasons , Cloprostenol/pharmacology , Cloprostenol/administration & dosage , Corpus Luteum/drug effects , Corpus Luteum/physiology , Insemination, Artificial/veterinary , Sexual Behavior, Animal/drug effects
16.
J Orthop Surg Res ; 19(1): 352, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38877549

ABSTRACT

PURPOSE: Facet joint degeneration (FJD) is a major cause of low back pain. Parathyroid hormone (PTH) (1-34) is commonly used to treat osteoporosis. However, little is known about its effects on FJD induced by estrogen deficiency. This study aims to investigate the effects of PTH (1-34) on FJD induced by estrogen deficiency and the underlying pathogenesis of the disease. METHODS: Forty 3-month-old female Sprague-Dawley rats were randomly divided into four groups: 30 received bilateral ovariectomy (OVX) followed by 12 weeks of treatment with normal saline, PTH (1-34) or 17ß-estradiol (E2), and 10 received sham surgery followed by administration of normal saline. Status and Wnt/ß-catenin signaling activity in the cartilage and subchondral bone of the L4-L5 FJs and serum biomarkers were analyzed. RESULTS: Administration of PTH (1-34) and E2 ameliorated cartilage lesions, and significantly decreased MMP-13 and caspase-3 levels and chondrocyte apoptosis. PTH (1-34) but not E2 significantly increased cartilage thickness, number of chondrocytes, and the expression of aggrecan. PTH (1-34) significantly improved microarchitecture parameters of subchondral bone, increased the expression of collagen I and osteocalcin, and decreased RANKL/OPG ratio. E2 treatment significantly increased the OPG level and decreased the RANKL/OPG ratio in the subchondral bone of ovariectomized rats, but it did not significantly improve the microarchitecture parameters of subchondral bone. Wnt3a and ß-catenin expression was significantly reduced in the articular cartilage and subchondral bone in OVX rats, but PTH (1-34) could increase the expression of these proteins. E2 significantly increased the activity of Wnt/ß-catenin pathway only in cartilage, but not in subchondral bone. The restoration of Wnt/ß-catenin signaling had an obvious correlation with the improvement of some parameters associated with the FJs status. CONCLUSION: Wnt/ß-catenin signaling may be a potential therapeutic target for FJD induced by estrogen deficiency. PTH (1-34) is effective in treating this disease with better efficacy than 17ß-estradiol, and the efficacy may be attributed to its restoration of Wnt/ß-catenin signaling.


Subject(s)
Lumbar Vertebrae , Ovariectomy , Parathyroid Hormone , Rats, Sprague-Dawley , Wnt Signaling Pathway , Zygapophyseal Joint , Animals , Female , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/physiology , Zygapophyseal Joint/drug effects , Rats , Estradiol/pharmacology , Estradiol/therapeutic use
17.
Reprod Domest Anim ; 59(6): e14642, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38894646

ABSTRACT

This study evaluated the effect of bovine somatotropin (bST) on pregnancy rate (PR) and size of the dominant follicle (DF) on the day of intravaginal progesterone (P4) removal in protocols for fixed-time artificial insemination (FTAI). Bos indicus (Nellore) females (n = 392) were distributed into three groups. The control group (CG; n = 92) received an intravaginal P4 device + estradiol benzoate on day (d)0; prostaglandin F2α on d7 (first application); removal of P4 + estradiol cypionate (EC) + PGF2α (second application) + ultrasound (US) of the DF on d9; the FTAI was performed on d11; and pregnancy diagnosis (PD) was performed on d45. The bST group (bSTG; n = 142) underwent the same protocol as the CG, except that the animals received 125 mg of bST on d7. The equine chorionic gonadotropin (eCG) group (eCGG; n = 158) underwent the same protocol as the CG, except that the animals received 300 IU of eCG on d9. The PRs of the bSTG, eCGG, and CG were 48%, 48%, and 35%, respectively (p < .05); the bSTG and eCGG showed greater PRs, with follicles 6-7.9 mm (p < .05) and 8-8.9 mm in diameter, respectively. The bSTG exhibited a greater dimension of the DF on d9 of the protocol (p < .05). The eCGG had higher PRs with a body condition score (BCS) of 2.5, and the bSTG had a BCS of 3.0 (p < .05). It was concluded that bST increased PR, bST showed better performance in smaller DF and larger follicular diameter on d9 of the protocol, eCG acted better on animals with lower BCSs, and bST can be used in FTAI.


Subject(s)
Growth Hormone , Insemination, Artificial , Pregnancy Rate , Progesterone , Animals , Female , Insemination, Artificial/veterinary , Insemination, Artificial/methods , Pregnancy , Cattle , Growth Hormone/pharmacology , Growth Hormone/administration & dosage , Progesterone/administration & dosage , Progesterone/pharmacology , Estradiol/administration & dosage , Estradiol/pharmacology , Estradiol/analogs & derivatives , Ovarian Follicle/drug effects , Dinoprost/administration & dosage , Dinoprost/pharmacology , Estrus Synchronization/methods , Administration, Intravaginal
18.
Biomed Pharmacother ; 176: 116852, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38834007

ABSTRACT

The incidence of breast cancer is increasing annually, making it a major health threat for women. Chemoprevention using natural, dietary, or synthetic products has emerged as a promising approach to address this growing burden. Atractylenolide-III (AT-III), a sesquiterpenoid present in various medicinal herbs, has demonstrated potential therapeutic effects against several diseases, including tumors, nonalcoholic fatty liver disease, and cerebral ischemic injury. However, its impact on breast cancer chemoprevention remains unexplored. In this study, we used an N-methyl-N-nitrosourea (NMU)-induced rat breast cancer model and 17ß-estradiol (E2)-treated MCF-10A cells to evaluate the chemopreventive potential of AT-III on mammary tumorigenesis. AT-III inhibited mammary tumor progression, evidenced by reduced tumor volume and multiplicity, prolonged tumor latency, and the reversal of NMU-induced weight loss. Furthermore, AT-III suppressed NMU-induced inflammation and oxidative stress through the Nrf2/ARE pathway in breast cancer tissues. In vitro, AT-III effectively suppressed E2-induced anchorage-independent growth and cell migration in MCF-10A cells. Nrf2 knockdown attenuated the protective effects of AT-III, highlighting the pivotal role of Nrf2 in AT-III-mediated suppression of tumorigenesis. The mechanism involves the induction of Nrf2 expression by AT-III through the autophagic degradation of Kelch-like ECH-associated protein 1 (Keap1). Overall, the results of this study indicate that AT-III is a promising candidate for breast cancer chemoprevention and provide valuable insights into its molecular interactions and signaling pathways.


Subject(s)
Autophagy , Kelch-Like ECH-Associated Protein 1 , Lactones , NF-E2-Related Factor 2 , Sesquiterpenes , Signal Transduction , Animals , NF-E2-Related Factor 2/metabolism , Sesquiterpenes/pharmacology , Female , Kelch-Like ECH-Associated Protein 1/metabolism , Lactones/pharmacology , Autophagy/drug effects , Signal Transduction/drug effects , Rats , Humans , Cell Line, Tumor , Rats, Sprague-Dawley , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/prevention & control , Mammary Neoplasms, Experimental/chemically induced , Oxidative Stress/drug effects , Methylnitrosourea/toxicity , Carcinogenesis/drug effects , Anticarcinogenic Agents/pharmacology , Estradiol/pharmacology
19.
Behav Brain Res ; 470: 115094, 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-38844057

ABSTRACT

Estrogen receptor (ER) activation by 17-ß estradiol (E2) can attenuate neuronal injury and behavioral impairments following global cerebral ischemia (GCI) in rodents. This study sought to further examine the discrete roles of ERs through characterization of the effects of selective ER activation on post-ischemic pro-inflammatory microglial activation, hippocampal neuronal injury, and anxiety-like behaviors. Forty-six ovariectomized (OVX) adult female Wistar rats received daily s.c injections (100 µg/kg/day) of propylpyrazole triol (PPT; ERα agonist), diarylpropionitrile (DPN; ERß agonist), G-1 (G-protein coupled ER agonist; GPER), E2 (activating all receptors), or vehicle solution (VEH) for 21 days. After final injection, rats underwent GCI via 4-vessel occlusion (n=8 per group) or sham surgery (n=6, vehicle injections). The Open Field Test (OFT), Elevated Plus Maze (EPM), and Hole Board Test (HBT) assessed anxiety-like behaviors. Microglial activation (Iba1, CD68, CD86) in the basolateral amygdala (BLA), CA1 of the hippocampus, and paraventricular nucleus of the hypothalamus (PVN) was determined 8 days post-ischemia. Compared to sham rats, Iba1 activation and CA1 neuronal injury were increased in all ischemic groups except DPN-treated rats, with PPT-treated ischemic rats also showing increased PVN Iba1-ir expression. Behaviorally, VEH ischemic rats showed slightly elevated anxiety in the EPM compared to sham counterparts, with no significant effects of agonists. While no changes were observed in the OFT, emotion regulation via grooming in the HBT was increased in G-1 rats compared to E2 rats. Our findings support selective ER activation to regulate post-ischemic microglial activation and coping strategies in the HBT, despite minimal impact on hippocampal injury.


Subject(s)
Anxiety , Brain Ischemia , CA1 Region, Hippocampal , Microglia , Phenols , Pyrazoles , Rats, Wistar , Animals , Female , Microglia/metabolism , Microglia/drug effects , Rats , Anxiety/metabolism , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/drug effects , Brain Ischemia/metabolism , Pyrazoles/pharmacology , Phenols/pharmacology , Ovariectomy , Neurons/metabolism , Neurons/drug effects , Propionates/pharmacology , Propionates/administration & dosage , Behavior, Animal/drug effects , Behavior, Animal/physiology , Estradiol/pharmacology , Disease Models, Animal , Receptors, Estrogen/metabolism , Nitriles/pharmacology
20.
Physiol Behav ; 283: 114609, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38851441

ABSTRACT

The neuropeptide kisspeptin (Kiss) is crucial in regulating the hypothalamic-pituitary-gonadal axis. It is produced by two main groups of neurons in the hypothalamus: the rostral periventricular region around the third ventricle and the arcuate nucleus. Kiss is the peptide product of the KiSS-1 gene and serves as the endogenous agonist for the GPR54 receptor. The Kiss/GPR54 system functions as a critical regulator of the reproductive system. Thus, we examined the effect of intracerebroventricular administration of 3 µg of Kiss to the right lateral ventricle of ovariectomized rats primed with a dose of 5 µg subcutaneous (sc) of estradiol benzoate (EB). Kiss treatment increased the lordosis quotient at all times tested. However, the lordosis reflex score was comparatively lower yet still significant compared to the control group. To investigate receptor specificity and downstream mechanisms on lordosis, we infused 10 µg of GPR54 receptor antagonist, Kiss-234, 5 µg of the progestin receptor antagonist, RU486, or 3 µg of antide, a gonadotropin-releasing hormone-1 (GnRH-1) receptor antagonist, to the right lateral ventricle 30 min before an infusion of 3 µg of Kiss. Results demonstrated a significant reduction in the facilitation of lordosis behavior by Kiss at 60 and 120 min when Kiss-234, RU486, or antide were administered. These findings suggest that Kiss stimulates lordosis expression by activating GPR54 receptors on GnRH neurons and that Kiss/GPR54 system is an essential intermediary by which progesterone activates GnRH.


Subject(s)
Estradiol , Kisspeptins , Receptors, LHRH , Receptors, Progesterone , Sexual Behavior, Animal , Animals , Kisspeptins/pharmacology , Kisspeptins/metabolism , Female , Sexual Behavior, Animal/drug effects , Sexual Behavior, Animal/physiology , Receptors, LHRH/antagonists & inhibitors , Receptors, LHRH/metabolism , Rats , Estradiol/pharmacology , Estradiol/analogs & derivatives , Receptors, Progesterone/metabolism , Receptors, Progesterone/drug effects , Receptors, Progesterone/antagonists & inhibitors , Ovariectomy , Rats, Wistar , Progesterone/pharmacology , Hormone Antagonists/pharmacology , Posture/physiology , Receptors, Kisspeptin-1/metabolism , Mifepristone/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...