Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35.197
Filter
1.
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
5.
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
6.
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
7.
Endocrinology ; 165(8)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38984714

ABSTRACT

The blood-brain barrier (BBB) is an extensive capillary network that protects the brain from environmental and metabolic toxins while limiting drug delivery to the central nervous system (CNS). The ATP-binding cassette transporter breast cancer resistance protein (Bcrp) reduces drug delivery across the BBB by actively transporting its clinical substrates back into peripheral circulation before their entry into the CNS compartment. 17ß-Estradiol (E2)-elicited changes in Bcrp transport activity and expression have been documented previously. We report a novel signaling mechanism by which E2 decreases Bcrp transport activity in mouse brain capillaries via rapid nongenomic signaling through estrogen receptor α. We extended this finding to investigate the effects of different endocrine-disrupting compounds (EDCs) and selective estrogen receptor modulators (SERMs) on Bcrp transport function. We also demonstrate sex-dependent expression of Bcrp and E2-sensitive Bcrp transport activity at the BBB ex vivo. This work establishes an explanted tissue-based model by which to interrogate EDCs and SERMs as modulators of nongenomic estrogenic signaling with implications for sex and hormonal regulation of therapeutic delivery into the CNS.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2 , Blood-Brain Barrier , Estradiol , Estrogen Receptor alpha , Signal Transduction , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Estrogen Receptor alpha/metabolism , Mice , Female , Signal Transduction/drug effects , Estradiol/pharmacology , Male , Biological Transport/drug effects , Mice, Inbred C57BL
8.
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
9.
Theriogenology ; 226: 343-349, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-38964033

ABSTRACT

Two experiments evaluated the effect of different hormonal treatments to synchronize follicle wave emergence on follicle dynamics and pregnancies per AI (P/AI) in estradiol (E2)/progesterone (P4) timed-AI (TAI) protocols in lactating dairy cows. In Experiment 1, lactating, primiparous Holstein cows (n = 36) received a P4 releasing device (Day 0) and were allocated at random to one of the following three treatment groups: Group EB received 2 mg E2 benzoate (EB) intramuscularly (i.m.), Group EB + GnRH received 2 mg EB+20 µg buserelin (GnRH) i.m., or Group EB + P4 received 2 mg EB + 100 mg of injectable P4 (iP4) in oil i.m. All cows received 0.150 mg D-Cloprostenol on Days 7 and 8 followed by P4 device removal, 400 IU eCG and 1 mg ECP on Day 8. Daily ultrasound examinations revealed that although the interval from P4 device removal to ovulation was not affected by treatment, cows that received EB + GnRH had an earlier (P < 0.05) emergence of the new follicular wave (Day 2.6 ± 0.2) than the other two treatment groups (Days 3.5 ± 0.3 and 6.1 ± 0.3, for EB and EB + P4, respectively). In Experiment 2, 808 lactating cows were assigned randomly to the three treatments evaluated in Experiment 1, and all the cows were TAI to determine P/AI. Cows in the EB + GnRH group had greater P/AI (57.4 %, P < 0.01) than those in the EB (44.6 %) or EB + P4 (45.7 %) groups. In conclusion, the administration of GnRH, but not iP4, on the day of insertion of a P4 device improves P/AI in lactating dairy cows synchronized for TAI with an estradiol/P4-based protocol.


Subject(s)
Estradiol , Estrus Synchronization , Gonadotropin-Releasing Hormone , Insemination, Artificial , Lactation , Ovarian Follicle , Progesterone , Animals , Cattle/physiology , Female , Insemination, Artificial/veterinary , Insemination, Artificial/methods , Lactation/drug effects , Ovarian Follicle/drug effects , Ovarian Follicle/physiology , Progesterone/administration & dosage , Progesterone/pharmacology , Estradiol/pharmacology , Estradiol/administration & dosage , Estradiol/analogs & derivatives , Estrus Synchronization/methods , Pregnancy , Gonadotropin-Releasing Hormone/pharmacology , Gonadotropin-Releasing Hormone/administration & dosage , Buserelin/pharmacology , Buserelin/administration & dosage
10.
Cytokine ; 181: 156686, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38991382

ABSTRACT

Liver injury, a major global health issue, stems from various causes such as alcohol consumption, nonalcoholic steatohepatitis, obesity, diabetes, metabolic syndrome, hepatitis, and certain medications. The liver's unique susceptibility to ischemia and hypoxia, coupled with the critical role of the gut-liver axis in inflammation, underscores the need for effective therapeutic interventions. The study highlights E2's interaction with estrogen receptors (ERs) and its modulation of the Toll-like receptor 4 (TLR4) signaling pathway as key mechanisms in mitigating liver injury. Activation of TLR4 leads to the release of pro-inflammatory cytokines and chemokines, exacerbating liver inflammation and injury. E2 down-regulates TLR4 expression, reduces oxidative stress, and inhibits pro-inflammatory cytokines, thereby protecting the liver. Both classic (ERα and ERß) and non-classic [G protein-coupled estrogen receptor (GPER)] receptors are influenced by E2. ERα is particularly crucial for liver regeneration, preventing liver failure by promoting hepatocyte proliferation. Furthermore, E2 exerts anti-inflammatory, antioxidant, and anti-apoptotic effects by inhibiting cytokines such as IL-6, IL-1ß, TNF-α, and IL-17, and by reducing lipid peroxidation and free radical damage. The article calls for further clinical research to validate these findings and to develop estrogen-based treatments for liver injuries. Overall, the research emphasizes the significant potential of E2 as a therapeutic agent for liver injuries. It advocates for extensive clinical studies to validate E2 hepatoprotective properties and develop effective estrogen-based treatments.


Subject(s)
Estradiol , Inflammation , Signal Transduction , Toll-Like Receptor 4 , Toll-Like Receptor 4/metabolism , Humans , Signal Transduction/drug effects , Animals , Estradiol/pharmacology , Inflammation/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Cytokines/metabolism , Oxidative Stress/drug effects , Protective Agents/pharmacology , Receptors, Estrogen/metabolism
11.
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
12.
Sci Rep ; 14(1): 15736, 2024 07 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
13.
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
14.
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
15.
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
16.
Anticancer Res ; 44(8): 3355-3364, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39060083

ABSTRACT

BACKGROUND/AIM: As an antagonist of bone morphogenetic protein (BMP), Noggin facilitates osteolytic bone metastases from breast cancer. The present study aimed to further dissect its role in oestrogen receptor (ER) positive breast cancer. MATERIALS AND METHODS: Noggin expression in ER positive breast cancer cell lines (MCF-7 and T-47D) was determined under conditions of oestrogen deprivation and treatment with 17-ß-oestradiol (E2). Activation of Smad1/5/8 in the oestrogen-regulated Noggin was examined using recombinant human BMP7 (rhBMP7) and a BMP receptor inhibitor (LDN-193189). The influence of Noggin on cellular functions was evaluated in MCF-7 and T-47D cell lines. Responses to tamoxifen and chemotherapy drugs were determined in MCF-7 and T-47D cells with Noggin over-expression using MTT assay. RESULTS: Noggin expression was negatively correlated with ERα in breast cancers. Noggin was up-regulated upon oestrogen deprivation, an effect that was eliminated by E2 Furthermore, increased levels of phosphorylated Smad1/5/8 were observed in the oestrogen-deprived MCF-7 and T-47D cells, which was prevented by E2 and LDN-193189, respectively. BMP7-induced Noggin expression and activation of Smad1/5/8 was also prevented by E2 and LDN-193189. Noggin over-expression resulted in an increase in the proliferation of both MCF-7 and T-47D cells. MCF-7 and T-47D cells over-expressing Noggin exhibited a good tolerance to tamoxifen (TAM), DTX, and 5-FU, but the percentage of viable cells was higher compared with the controls. CONCLUSION: Noggin expression can be repressed by oestrogen through inference with the BMP/Smad signalling. Over-expression of Noggin promotes the proliferation of MCF-7 and T-47D cells, contributing to drug resistance.


Subject(s)
Breast Neoplasms , Carrier Proteins , Estrogens , Signal Transduction , Smad Proteins , Tamoxifen , Humans , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Carrier Proteins/metabolism , Carrier Proteins/genetics , Female , Signal Transduction/drug effects , Smad Proteins/metabolism , Estrogens/pharmacology , Estrogens/metabolism , MCF-7 Cells , Tamoxifen/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Bone Morphogenetic Protein 7/metabolism , Bone Morphogenetic Protein 7/genetics , Gene Expression Regulation, Neoplastic/drug effects , Bone Morphogenetic Proteins/metabolism , Bone Morphogenetic Proteins/genetics , Estrogen Receptor alpha/metabolism , Estrogen Receptor alpha/genetics , Estradiol/pharmacology
17.
Steroids ; 209: 109469, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38992454

ABSTRACT

This is the first report on a unique hybrid molecule made of estradiol and testosterone (TS). This distinctive hybrid molecule (1) was designed to interact with both the estrogen receptor (ER) and the androgen receptor (AR) found in hormone-dependent female and male cancer cells, and was synthesized using ethynylestradiol (17EE) as the estrogenic component and 7α-(4-azido-but-2-enyl)-4-androsten-17ß-ol-3-one as the androgenic counterpart in a seven-step reaction with âˆ¼ 26 % overall yield. We reasoned that the dual receptor binding ability could allow 1 to act as an antihormone. This was tested on hormone-dependent and hormone-independent breast cancer (BCa) and prostate cancer (PCa) cells. The antiproliferative activity was also assessed on colon and skin cancer cells. We found that 1 was active against MCF7 (ER + ) BCa cells (IC50 of 4.9 µM), had lower inhibitory potency on LNCaP (AR + ) PCa cells (IC50 > 5 µM) and no effect on PC3 and DU145 (AR-) PCa cells. This suggests that the estrogenic component of 1 can interact with the ER on MCF7 cells more effectively than the androgenic component with the AR on LNCaP PCa cells, possibly due to a suboptimal spacer or linkage site(s). Nonetheless, the hybrid 1 was active against colon (HT-29) and melanoma (M21) cancer cells (IC50 of 3.5 µM and 2.3 µM, respectively), and had low cross-reactivity with the drug- and androgen-metabolizing cytochrome P450 3A4 (CYP3A4, IC50 ≫ 5 µM). These findings demonstrate the anticancer potential of 1 and warrant further explorations on this new type of hybrids.


Subject(s)
Antineoplastic Agents , Cell Proliferation , Estradiol , Testosterone , Humans , Testosterone/pharmacology , Testosterone/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Estradiol/pharmacology , Estradiol/chemistry , Estradiol/chemical synthesis , Drug Screening Assays, Antitumor , Cell Line, Tumor , Structure-Activity Relationship , Receptors, Estrogen/metabolism , Receptors, Androgen/metabolism , Molecular Structure , Male
18.
Int J Mol Sci ; 25(14)2024 Jul 10.
Article in English | MEDLINE | ID: mdl-39062809

ABSTRACT

The study evaluated the effects of Arthrospira maxima phycobiliproteins (PBPs), rosiglitazone (RSG), and 17ß-estradiol (E) on the differentiation process of 3T3-L1 cells and on their regulation of lipogenic and inflammatory gene expression at different stages of the process. The results showed that phycobiliproteins promoted cell proliferation after 24 h of treatment. Furthermore, for all three treatments, the regulation of the highest number of markers occurred on days 6 and 12 of differentiation, regardless of when the treatment was applied. Phycobiliproteins reduced lipid droplet accumulation on days 3, 6, 10, and 13 of the adipogenic process, while rosiglitazone showed no differences compared to the control. On day 6, both phycobiliproteins and rosiglitazone positively regulated Acc1 mRNA. Meanwhile, all three treatments negatively regulated Pparγ and C/ebpα. Phycobiliproteins and estradiol also negatively regulated Ucp1 and Glut4 mRNAs. Rosiglitazone and estradiol, on the other hand, negatively regulated Ppara and Il-6 mRNAs. By day 12, phycobiliproteins and rosiglitazone upregulated Pparγ mRNA and negatively regulated Tnfα and Il-1ß. Additionally, phycobiliproteins and estradiol positively regulated Il-6 and negatively regulated Ppara, Ucp2, Acc1, and Glut4. Rosiglitazone and estradiol upregulate C/ebpα and Ucp1 mRNAs. The regulation exerted by phycobiliproteins on the mRNA expression of the studied markers was dependent on the phase of cell differentiation. The results of this study highlight that phycobiliproteins have an anti-adipogenic and anti-inflammatory effect by reducing the expression of adipogenic, lipogenic, and inflammatory genes in 3T3-L1 cells at different stages of the differentiation process.


Subject(s)
3T3-L1 Cells , Adipocytes , Adipogenesis , Cell Differentiation , Estradiol , Phycobiliproteins , Rosiglitazone , Animals , Mice , Estradiol/pharmacology , Rosiglitazone/pharmacology , Adipocytes/drug effects , Adipocytes/metabolism , Adipocytes/cytology , Cell Differentiation/drug effects , Adipogenesis/drug effects , Adipogenesis/genetics , Phycobiliproteins/pharmacology , Phycobiliproteins/metabolism , Phycobiliproteins/genetics , Gene Expression Regulation/drug effects , Lipogenesis/drug effects , Lipogenesis/genetics , PPAR gamma/metabolism , PPAR gamma/genetics , Cell Proliferation/drug effects , Inflammation/metabolism , Inflammation/genetics , Spirulina
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL