Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 27
1.
J Endocrinol ; 255(3): 103-116, 2022 12 01.
Article En | MEDLINE | ID: mdl-36069766

Recent reports indicate an increase in Leydig cell tumor (LCT) incidence. Radical orchiectomy is the standard therapy in children and adults, although it entails physical and psychosocial side effects. Testis-sparing surgery can be a consideration for benign LCT of 2.5 cm or less in size. Malignant LCTs respond poorly to conventional chemotherapy, so new treatment modalities are needed. In this study, we observed increased histidine decarboxylase expression and pro-angiogenic potential in LCT surgically resected from pediatric patients (fetal to pubertal) vs control samples from patients without endocrine or metabolic disorders which were collected at necropsy. We, therefore, evaluated for the first time the antitumor efficacy of two histidine decarboxylase inhibitors (α-methyl-dl-histidine dihydrochloride (α-MHD) and epigallocatechin gallate (EGCG)), alone and combined with carboplatin, in two preclinical models of LCT. MA-10 and R2C Leydig tumor cells, representing two different LCT subtypes, were used to generate syngeneic and xenograft mouse LCT models, respectively. In the syngeneic model, monotherapy with α-MHD effectively reduced tumor growth and angiogenesis. In the xenografts, which showed co-expression of histidine decarboxylase and CYP19, the combination of EGCG plus carboplatin was the most effective therapy, leading to LCT growth arrest and undetectable levels of plasmatic estradiol. Testicular and body weights remained unaltered. On the basis of this study, histidine decarboxylase may emerge as a novel pharmacological target for LCT treatment.


Leydig Cell Tumor , Testicular Neoplasms , Animals , Aromatase , Carboplatin , Estradiol , Histidine , Histidine Decarboxylase/genetics , Humans , Leydig Cell Tumor/metabolism , Leydig Cell Tumor/pathology , Leydig Cell Tumor/surgery , Male , Mice , Testicular Neoplasms/metabolism , Testicular Neoplasms/pathology , Testicular Neoplasms/surgery
2.
Reproduction ; 161(1): R1-R11, 2021 01.
Article En | MEDLINE | ID: mdl-33112290

Although ovarian cancer mortality rates have slightly declined in the last 40 years, ovarian cancer continues to be the eighth cause of cancer death in women. Ovarian cancer is characterized by its high response to treatments but also by its high rate of recurrence. Although treatments are limited to cytoreductive surgery and platinum-based chemotherapy, other therapies using antiangiogenic agents and poly (ADP-ribose) polymerase inhibitors are being tested. Nevertheless, these therapeutic strategies have had poor results and new potential targets and approaches are thus needed. The present review focuses on the recent evidence on antiangiogenic strategies in ovarian cancer cells and on the mechanisms governed by Notch and ß-catenin proteins. It also describes the concept of 'vascular normalization' by using the platelet-derived growth factor, PDGFB, molecule as a tool to regulate ovarian tumor angiogenesis and thus improve ovarian tumor treatment. It has been reported that alterations in the Notch system components and changes in the canonical Wnt/ß-catenin signaling, the other pathway of our interest, are relevant to molecular events that contribute to ovarian cancer development. Thus, in this review, we consider these aspects of the ovarian tumor biology as potential new therapeutic strategies for the treatment of this disease.


Angiogenesis Inhibitors/therapeutic use , Carcinoma/drug therapy , Molecular Targeted Therapy , Ovarian Neoplasms/drug therapy , Animals , Carcinoma/metabolism , Female , Humans , Ovarian Neoplasms/metabolism , Receptors, Notch/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism
3.
Reprod Sci ; 27(2): 503-512, 2020 02.
Article En | MEDLINE | ID: mdl-32046463

The aim of this study was to investigate whether the Notch pathway is modulated in response to the downregulation of the Wnt/Β-catenin system in corpora lutea (CLs) from superovulated rats. To this end, we analyzed the effect of in vitro CL Wnt/Β-catenin inhibition on the expression of Notch members and on luteal function. Mechanically isolated rat CLs were cultured with ICG-001, a Wnt/B-catenin inhibitor. In this system, Wnt/B-catenin inhibition reduced progesterone production and decreased StAR protein levels. Besides, Wnt/B-catenin inhibition stimulated the Notch system, evidenced by an increase in Hes1 expression, and promoted the expression of selected Notch family members. At long incubation times, StAR levels and progesterone concentration reached the control values, effects probably mediated by the Notch pathway. These results provide the first evidence of a compensatory mechanism between Wnt/B-catenin signaling and the Notch system, which contributes to the homeostasis of luteal cells.


Corpus Luteum/metabolism , Receptors, Notch/metabolism , Wnt Signaling Pathway , Animals , Cyclin D1/metabolism , Down-Regulation , Female , Phosphoproteins/metabolism , Progesterone/metabolism , Rats, Sprague-Dawley , Transcription Factor HES-1/metabolism
4.
Mol Cell Endocrinol ; 499: 110591, 2020 01 01.
Article En | MEDLINE | ID: mdl-31546019

Metformin (MET) is the most widely prescribed hypoglycemic drug in type 2 diabetes and Polycystic Ovary Syndrome. Besides its effects on glucose metabolism, MET exerts beneficial effects on these patients' fertility. However, the exact mechanisms of action of MET on female fertility are still unclear. In this work, we analyzed a possible direct effect of MET on ovarian cells. We found expression of the organic cation transporters OCT1, OCT2 and OCT3, responsible for MET uptake into the cells, in rat granulosa cells and human cumulus cells. Furthermore, MET increased pAMPK and decreased VEGF levels both in vivo and in rat granulosa cells in culture. These last effects were reversed when OCTs were inhibited. Our results suggest that MET acts directly on ovarian cells regulating cell metabolism and VEGF expression. Our findings are relevant to optimize PCOS fertility treatment and to explore ovarian MET actions in other female pathologies.


Adenylate Kinase/metabolism , Cumulus Cells/cytology , Metformin/administration & dosage , Octamer Transcription Factors/metabolism , Vascular Endothelial Growth Factor A/metabolism , Adult , Animals , Cell Proliferation/drug effects , Cumulus Cells/drug effects , Cumulus Cells/metabolism , Female , Gene Expression Regulation/drug effects , Humans , Metformin/pharmacology , Models, Animal , Phosphorylation/drug effects , Rats
5.
J Cell Physiol ; 234(12): 22130-22143, 2019 12.
Article En | MEDLINE | ID: mdl-31087357

In the last 40 years ovarian cancer mortality rates have slightly declined and, consequently, it continues to be the fifth cause of cancer death in women. In the present study, we showed that ß-catenin signaling is involved in the functions of ovarian cancer cells and interacts with the Notch system. Wnt and Notch systems showed to be prosurvival for ovarian cancer cells and their inhibition impaired cell proliferation and migration. We also demonstrated that the inhibition of ß-catenin by means of two molecules, XAV939 and ICG-001, decreased the proliferation of the IGROV1 and SKOV3 ovarian cancer cell lines and that ICG-001 increased the percentage of IGROV1 cells undergoing apoptosis. The simultaneous inhibition of ß-catenin and Notch signaling, by using the DAPT inhibitor, decreased ovarian cancer cell proliferation to the same extent as targeting only the Wnt/ß-catenin pathway. A similar effect was observed in IGROV1 cell migration with ICG-001 and DAPT. ICG-001 increased the Notch target genes Hes-1 and Hey-1 and increased Jagged1 expression. However, no changes were observed in Dll4 or Notch 1 and 4 expressions. Our results suggest that Notch and ß-catenin signaling co-operate in ovarian cancer to ensure the proliferation and migration of cells and that this could be achieved, at least partly, by the upregulation of Notch Jagged1 ligand in the absence of Wnt signaling. We showed that the Wnt pathway crosstalks with Notch in ovarian cancer cell functions, which may have implications in ovarian cancer therapeutics.


Carcinoma, Ovarian Epithelial/pathology , Ovarian Neoplasms/pathology , Receptors, Notch/metabolism , Signal Transduction/physiology , Wnt Signaling Pathway/physiology , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Carcinoma, Ovarian Epithelial/metabolism , Cell Movement/drug effects , Cell Movement/physiology , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cell Survival , Diamines/pharmacology , Female , Humans , Mice , Mice, Nude , Ovarian Neoplasms/metabolism , Pyrimidinones/pharmacology , Thiazoles/pharmacology , Wnt Signaling Pathway/drug effects , Xenograft Model Antitumor Assays
6.
J Cell Physiol ; 233(8): 5949-5961, 2018 08.
Article En | MEDLINE | ID: mdl-29266203

Ovarian cancer is the fifth leading cause of cancer-related deaths in women. In the past 20 years, the canonical types of drugs used to treat ovarian cancer have not been replaced and the survival rates have not changed. These facts show the clear need to find new therapeutic strategies for this illness. Thus, the aim of the present study was to investigate the effect of a gamma-secretase inhibitor (DAPT) in combination with the Platelet-derived growth factor B (PDGFB) on an ovarian cancer xenograft model. To achieve this goal, we analyzed the effect of the administration of DAPT alone and the co-administration of DAPT and recombinant PDGFB on parameters associated with tumor growth and angiogenesis in an orthotopic experimental model of ovarian cancer. We observed that the dose of DAPT used was ineffective to reduce ovarian tumor growth, but showed anticancer activity when co-administered with recombinant PDGFB. The administration of PDGFB alone normalized tumor vasculature by increasing periendothelial coverage and vascular functionality. Interestingly, this effect exerted by PDGFB was also observed in the presence of DAPT. Our findings suggest that PDGFB is able to improve tumor vascularity and allows the anticancer action of DAPT in the tumor. We propose that this therapeutic strategy could be a new tool for ovarian cancer treatment and deserves further studies.


Amyloid Precursor Protein Secretases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Proto-Oncogene Proteins c-sis/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Diamines/pharmacology , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred BALB C , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Thiazoles/pharmacology
7.
Mol Reprod Dev ; 84(8): 719-730, 2017 Aug.
Article En | MEDLINE | ID: mdl-28628258

Tankyrases are physiological regulators of Axin, a protein involved in several cellular processes, including Wnt signaling. Here, we investigated the effect of a specific Tankyrase inhibitor (XAV939) in follicular-luteal dynamics, and its possible relationship with ovarian vascular development. Studies were designed to analyze the effect of intrabursa administration of XAV939 in gonadotropin-treated prepubertal rats. In particular, we examined follicle and corpus luteum development, steroidogenesis, angiogenic markers, and apoptotic parameters. We found that in vivo inhibition of Wnt signaling impaired corpus luteum development, with a decrease in the number of corpora lutea balanced by a high number of cysts; decreased circulating progesterone levels, likely due to a decrease in Steroidogenic acute regulatory protein content in the corpus luteum; and increased pro-apoptotic parameters. In addition, Extracellular signal-regulated kinase phosphorylation, Vascular endothelium growth factor 120 content, and endothelial cell area were diminished in corpora lutea of inhibitor-treated ovaries. Thus, Wnt/ß-catenin signaling appears to participate in the regulation of corpus luteum development and luteal cell function.


Corpus Luteum/metabolism , Progesterone/metabolism , Tankyrases/antagonists & inhibitors , Tankyrases/metabolism , Animals , Corpus Luteum/physiology , Female , Gonadotropins/metabolism , Rats , Signal Transduction/physiology , Wnt Proteins/metabolism , beta Catenin/metabolism
8.
Mol Hum Reprod ; 23(6): 417-427, 2017 06 01.
Article En | MEDLINE | ID: mdl-28379469

STUDY QUESTION: Can the bioactive lipid sphingosine-1 phosphate (S1P) act as an endothelial barrier-enhancing molecule and, in turn, restore the vascular integrity and homoeostasis in a rat model of ovarian hyperstimulation syndrome (OHSS). STUDY ANSWER: In vivo administration of S1P may prevent the early onset of OHSS and decrease its severity. WHAT IS KNOWN ALREADY: Although advances in the prediction and treatment of OHSS have been made, complete prevention has not been possible yet. S1P in follicular fluid from women at risk of developing OHSS are lower in comparison from women who are not at such risk and administration of S1P in an OHSS rat model decreases ovarian capillary permeability. STUDY DESIGN, SIZE, DURATION: We used an animal model that develops OHSS in immature Sprague-Dawley rats. The rats were randomly divided into three groups: the control group, which was injected with 10 IU of pregnant mare's serum gonadotropin (PMSG), and 10 IU of hCG 48 h later; the OHSS group, which was injected with excessive doses of PMSG (50 IU/day) for four consecutive days, followed by hCG; and the OHSS + S1P group, which was injected with the same doses of PMSG and hCG as the OHSS group and then treated with 5 µl S1P (1 mM) under the bursa of both ovaries, whereas the other groups of animals received the S1P vehicle. PARTICIPANTS /MATERIALS, SETTING, METHODS: Rats were killed by decapitation 48 h after the hCG injection for ovary, endometrium and blood collection. The ovaries were weighed and then used for subsequent assays, while the serum was used for hormone assays. One of the ovaries from each rat (n = 6) was used for Western immunoblot and the other for immunohistochemical analysis. Statistical comparisons between groups were carried out. MAIN RESULTS AND THE ROLE OF CHANCE: S1P administration reduced the ovarian weight (P < 0.05), and decreased the concentration of serum progesterone in the OHSS group compared to the OHSS group without treatment (P < 0.001). The percentage of antral follicles in the OHSS group was lower than that in the control group. S1P increased the percentage of antral follicles (P < 0.05) and decreased the percentage of corpora lutea (P < 0.01) and cystic structures in the OHSS group (P < 0.05). S1P had no effect on the expression levels of the enzymes 3ß-hydroxysteroid dehydrogenase (3ßHSD) or cholesterol side-chain cleavage enzyme (P450scc), but reduced the levels of steroidogenic acute regulatory protein (StAR) in OHSS rat ovaries (P < 0.05). S1P decreased the endothelial (P < 0.05) and periendothelial (P < 0.01) cell area in OHSS rat ovaries. S1P restored the levels of N-cadherin and VE-cadherin proteins to control values. Furthermore, S1P enhanced the levels of claudin-5, occludin (P < 0.05) and sphingosine-1-phosphate receptor 1 (S1PR1) in OHSS (P < 0.01). In addition, no histological differences were found in endometrium between OHSS and S1P-treated OHSS animals. LIMITATIONS REASONS FOR CAUTION: The results of this study were generated from an in vivo OHSS experimental model, which has been used by several authors and our group due to the similarity between the rat and human angiogenic systems. Further studies in patients will be needed to evaluate the effects of S1P in the pathogenesis of OHSS. WIDER IMPLICATIONS OF THE FINDINGS: These findings concern the pathophysiological importance of S1P in OHSS. More studies on the regulation of endothelial cell barrier function by S1P in reproductive pathological processes and its therapeutic application are required. LARGE SCALE DATA: N/A. STUDY FUNDING AND COMPETING INTEREST(S): This work was supported by grants from ANPCyT (PICT 2012-897), CONICET (PIP 5471), Roemmers and Baron Foundations, Argentina. The authors declare no conflicts of interest.


Capillary Permeability/drug effects , Corpus Luteum/drug effects , Lysophospholipids/pharmacology , Ovarian Follicle/drug effects , Ovarian Hyperstimulation Syndrome/drug therapy , Sphingosine/analogs & derivatives , 3-Hydroxysteroid Dehydrogenases/genetics , 3-Hydroxysteroid Dehydrogenases/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Cadherins/genetics , Cadherins/metabolism , Claudin-5/genetics , Claudin-5/metabolism , Corpus Luteum/metabolism , Corpus Luteum/pathology , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Disease Models, Animal , Female , Gene Expression Regulation/drug effects , Gonadotropins, Equine/pharmacology , Humans , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Occludin/genetics , Occludin/metabolism , Organ Size , Ovarian Follicle/metabolism , Ovarian Follicle/pathology , Ovarian Hyperstimulation Syndrome/genetics , Ovarian Hyperstimulation Syndrome/metabolism , Ovarian Hyperstimulation Syndrome/pathology , Phosphoproteins/genetics , Phosphoproteins/metabolism , Pregnancy , Progesterone/blood , Rats , Rats, Sprague-Dawley , Receptors, Lysosphingolipid/genetics , Receptors, Lysosphingolipid/metabolism , Sphingosine/pharmacology , Sphingosine-1-Phosphate Receptors
9.
Mol Cell Endocrinol ; 433: 47-55, 2016 09 15.
Article En | MEDLINE | ID: mdl-27256152

Alterations in ovarian angiogenesis are common features in Polycystic Ovary Syndrome (PCOS) patients; the most studied of these alterations is the increase in vascular endothelial growth factor (VEGF) production by ovarian cells. Platelet-derived growth factor B (PDGFB) and D (PDGFD) are decreased in follicular fluid of PCOS patients and in the ovaries of a rat model of PCOS. In the present study, we aimed to analyze the effects of local administration of PDGFB on ovarian angiogenesis, follicular development and ovulation in a DHEA-induced PCOS rat model. Ovarian PDGFB administration to PCOS rats partially restored follicular development, decreased the percentage of cysts, increased the percentage of corpora lutea, and decreased the production of anti-Müllerian hormone. In addition, PDGFB administration improved ovarian angiogenesis by reversing the increase in periendothelial cell area and restoring VEGF levels. Our results shed light into the mechanisms that lead to altered ovarian function in PCOS and provide new data for potential therapeutic strategies.


Follicular Fluid/drug effects , Neovascularization, Pathologic/drug therapy , Ovarian Follicle/drug effects , Polycystic Ovary Syndrome/drug therapy , Proto-Oncogene Proteins c-sis/administration & dosage , Animals , Anti-Mullerian Hormone , Female , Follicular Fluid/metabolism , Neovascularization, Pathologic/metabolism , Ovarian Follicle/metabolism , Ovarian Hyperstimulation Syndrome/drug therapy , Ovarian Hyperstimulation Syndrome/metabolism , Ovulation/drug effects , Polycystic Ovary Syndrome/metabolism , Rats , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor A/metabolism
10.
Reprod Fertil Dev ; 28(6): 690-9, 2016 Apr.
Article En | MEDLINE | ID: mdl-25485810

Ovarian hyperstimulation syndrome (OHSS) is a complication of ovarian stimulation with gonadotrophins following human chorionic gonadotrophin (hCG) administration. The relationship between hCG and OHSS is partly mediated via the production of angiogenic factors, such as vascular endothelial growth factor A (VEGFA) and angiopoietins (ANGPTs). Here, we investigated the effect of ANGPT1 inhibition on ovarian angiogenesis in follicular fluid (FF) from women at risk of OHSS, using the chorioallantoic membrane (CAM) of quail embryos as an experimental model. We also analysed cytoskeletal changes and endothelial junction protein expression induced by this FF in the presence or absence of an ANGPT1-neutralising antibody in endothelial cell cultures. The presence of this antibody restored the number of vascular branch points and integrin αvß3 levels in the CAMs to control values. ANGPT1 inhibition in FF from OHSS patients also restored the levels of claudin-5, vascular endothelial cadherin and phosphorylated ß-catenin and partially reversed actin redistribution in endothelial cells. Our findings suggest that ANGPT1 increases pathophysiological angiogenesis in patients at risk of OHSS by acting on tight and adherens junction proteins. Elucidating the mechanisms by which ANGPT1 regulates vascular development and cell-cell junctions in OHSS will contribute to identifying new therapeutic targets for the treatment of human diseases with aberrant vascular leakage.


Adherens Junctions/metabolism , Angiopoietin-1/metabolism , Endothelium, Vascular/metabolism , Neovascularization, Pathologic/etiology , Ovarian Follicle/metabolism , Ovarian Hyperstimulation Syndrome/physiopathology , Tight Junctions/metabolism , Adherens Junctions/drug effects , Adherens Junctions/pathology , Adult , Angiopoietin-1/antagonists & inhibitors , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Argentina/epidemiology , Biological Assay , Biomarkers/metabolism , Cell Line , Cells, Cultured , Chorioallantoic Membrane/blood supply , Chorioallantoic Membrane/drug effects , Chorioallantoic Membrane/metabolism , Coturnix , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Female , Follicular Fluid/cytology , Follicular Fluid/metabolism , Humans , Ovarian Follicle/blood supply , Ovarian Follicle/drug effects , Ovarian Follicle/pathology , Ovarian Hyperstimulation Syndrome/epidemiology , Ovarian Hyperstimulation Syndrome/metabolism , Ovarian Hyperstimulation Syndrome/pathology , Risk , Tight Junctions/drug effects , Tight Junctions/pathology
11.
Mol Cell Endocrinol ; 412: 148-58, 2015 Sep 05.
Article En | MEDLINE | ID: mdl-25937181

The platelet-derived growth factor (PDGF) system is crucial for blood vessel stability. In the present study, we evaluated whether PDGFs play a critical intraovarian survival role in gonadotropin-dependent folliculogenesis. We examined the effect of intrabursal administration of a selective platelet-derived growth factor receptor (PDGFR) inhibitor (AG1295) on follicular development, proliferation, apoptosis and blood vessel formation and stability in ovaries from rats treated with equine chorionic gonadotropin (eCG). The percentages of preantral follicles (PAFs) and early antral follicles (EAFs) were lower in AG1295-treated ovaries than in control ovaries (p < 0.01-0.05). The percentage of atretic follicles (AtrFs) increased in AG1295-treated ovaries compared to control (p < 0.05). The ovarian weight and estradiol concentrations were lower in AG1295-treated ovaries than in the control group (p < 0.01 and p < 0.05, respectively), whereas progesterone concentrations did not change. AG1295 decreased the proliferation index in EAFs (p < 0.05) and increased the percentage of nuclei positive for cleaved caspase-3 and apoptotic DNA fragmentation (p < 0.01-0.05). AG1295 increased the expression of Bax (p < 0.05) without changes in the expression of Bcl-2 protein. AG1295-treated ovaries increased the cleavage of caspase-8 (p < 0.05) and decreased AKT and BAD phosphorylation compared with control ovaries (p < 0.05). AG1295 caused a decrease not only in the endothelial cell area but also in the area of pericytes and vascular smooth muscle cells (VSMCs) in the ovary (p < 0.05). Our findings suggest that the local inhibition of PDGFs causes an increase in ovarian apoptosis through an imbalance in the ratio of antiapoptotic to proapoptotic proteins, thus leading a larger number of follicles to atresia. PDGFs could exert their mechanism of action through an autocrine/paracrine effect on granulosa and theca cells mediated by PDGFRs. In conclusion, these data clearly indicate that the PDGF system is necessary for follicular development induced by gonadotropins.


Apoptosis , Cell Proliferation , Chorionic Gonadotropin/pharmacology , Ovarian Follicle/physiology , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Animals , Female , Horses , Neovascularization, Physiologic , Ovarian Follicle/blood supply , Ovary/blood supply , Ovary/cytology , Platelet-Derived Growth Factor/physiology , Rats, Sprague-Dawley , Sexual Maturation , Tyrphostins/pharmacology
12.
Endocrinology ; 156(4): 1453-63, 2015 Apr.
Article En | MEDLINE | ID: mdl-25590243

Polycystic ovary syndrome (PCOS) is a frequent pathology that affects more than 5% of women of reproductive age. Among other heterogeneous symptoms, PCOS is characterized by abnormalities in angiogenesis. Metformin has been introduced in the treatment of PCOS to manage insulin resistance and hyperglycemia. Besides its metabolic effects, metformin has been shown to improve ovulation, pregnancy and live birth rates in PCOS patients. In the present study, we used a dehydroepiandrosterone-induced PCOS rat model to analyze the effect of metformin administration on ovarian angiogenesis. We found that metformin was able to restore the increased levels of vascular endothelial growth factor, angiopoietin (ANGPT)1, and ANGPT1/ANGPT2 ratio and the decreased levels of platelet-derived growth factor B and platelet-derived growth factor D observed in the dehydroepiandrosterone-treated rats. These effects could take place, at least in part, through a decrease in the levels of serum insulin. We also found an improvement in follicular development, with a lower percentage of small follicles and cysts and a higher percentage of antral follicles and corpora lutea after metformin administration. The improvement in ovarian angiogenesis is likely to restore the accumulation of small follicles observed in PCOS rats and to reduce cyst formation, thus improving follicular development and the percentage of corpora lutea. These results open new insights into the study of metformin action not only in glucose metabolism but also in ovarian dysfunction in PCOS women.


Angiogenesis Modulating Agents/pharmacology , Metformin/pharmacology , Neovascularization, Pathologic/drug therapy , Ovarian Follicle/drug effects , Polycystic Ovary Syndrome/drug therapy , Angiogenesis Modulating Agents/therapeutic use , Angiopoietin-1/blood , Angiopoietin-2/blood , Animals , Dehydroepiandrosterone , Female , Insulin/blood , Insulin Resistance , Metformin/therapeutic use , Neovascularization, Pathologic/blood , Neovascularization, Pathologic/physiopathology , Ovarian Follicle/physiopathology , Platelet-Derived Growth Factor/metabolism , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/physiopathology , Rats , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor A/blood
13.
J Steroid Biochem Mol Biol ; 144 Pt B: 392-401, 2014 Oct.
Article En | MEDLINE | ID: mdl-25151950

The relationship between human chorionic gonadotropin and ovarian hyperstimulation syndrome (OHSS) is partially mediated by vascular endothelial growth factor A (VEGF). The aim of this study was to investigate the effects of VEGF inhibition on the development of corpora lutea (CL) and cystic structures, steroidogenesis, apoptosis, cell proliferation, endothelial cell area, VEGF receptors (KDR and Flt-1), claudin-5 and occludin levels in ovaries from an OHSS rat model. The VEGF inhibitor used (VEGF receptor-1 (FLT-1)/Fc chimera, TRAP) decreased the concentrations of progesterone and estradiol as well as the percentage of CL and cystic structures in OHSS rats, and increased apoptosis in CL. Endothelial cell area in CL and KDR expression and its phosphorylation were increased, whereas claudin-5 and occludin levels were decreased in the OHSS compared to the control TRAP reversed these parameters. Our findings indicate that VEGF inhibition prevents the early onset of OHSS and decreases its severity in rats.


Angiogenesis Inhibitors/pharmacology , Ovarian Hyperstimulation Syndrome/metabolism , Receptors, Vascular Endothelial Growth Factor/pharmacology , Recombinant Fusion Proteins/pharmacology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Claudin-5/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Estradiol/blood , Female , Occludin/metabolism , Ovarian Hyperstimulation Syndrome/pathology , Ovary/drug effects , Ovary/metabolism , Ovary/pathology , Progesterone/blood , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
14.
Mol Reprod Dev ; 81(8): 748-56, 2014 Aug.
Article En | MEDLINE | ID: mdl-24889290

Polycystic ovary syndrome (PCOS) is the most common endocrinological pathology among women of reproductive age, and is characterized by abnormalities in ovarian angiogenesis, among other features. Consistent with this association, follicular fluid (FF) concentration and ovarian expression of vascular endothelial growth factor (VEGF) are increased in PCOS patients. In this study, we examined the protein levels of platelet-derived growth factor (PDGF) BB and DD (PDGFBB and PDGFDD), angiopoietin 1 and 2 (ANGPT1 and ANGPT2), and their soluble receptor sTIE2 in FF from PCOS and control patients undergoing assisted reproductive techniques. We also analyzed the effect of FF from PCOS and control patients on tight and adherens junction protein expression in an endothelial cell line. PDGFBB and PDGFDD were significantly lower whereas ANGPT1 concentration was significantly higher in FF from PCOS patients than from control patients. No changes were found in the concentration of ANGPT2 or sTIE2. Expression of claudin-5 was significantly increased in endothelial cells incubated for 24 hr in the presence of FF from PCOS versus from control patients, while vascular-endothelial cadherin, ß-catenin, and zonula occludens 1 expression were unchanged. The changes observed in the levels of PDGF isoforms and ANGPT1 may prevent VEGF-induced vascular permeability in the PCOS ovary by regulating endothelial-cell-junction protein levels. Restoring the levels of angiogenic factors may provide new insights into PCOS treatment and the prevention of ovarian hyperstimulation syndrome in affected women.


Angiopoietin-1/metabolism , Angiopoietin-2/metabolism , Follicular Fluid/metabolism , Ovary/metabolism , Polycystic Ovary Syndrome/metabolism , Proto-Oncogene Proteins c-sis/metabolism , Adherens Junctions/metabolism , Adult , Becaplermin , Blotting, Western , Claudin-5/metabolism , Endothelial Cells/metabolism , Estradiol/metabolism , Female , Humans , Ovarian Hyperstimulation Syndrome/prevention & control , Progesterone/metabolism , Radioimmunoassay , Reproductive Techniques, Assisted , Tight Junctions/metabolism , Vascular Endothelial Growth Factor A/metabolism
15.
J Cell Physiol ; 229(11): 1673-80, 2014 Nov.
Article En | MEDLINE | ID: mdl-24615682

Knockout models have shown that the coagulation system has a role in vascular development and angiogenesis. Herein, we report for the first time that zymogen FX and its active form (FXa) possess anti-angiogenic properties. Both the recombinant FX and FXa inhibit angiogenesis in vitro using endothelial EA.hy926 and human umbilical cord vascular endothelial cells (HUVEC). This effect is dependent on the Gla domain of FX. We demonstrate that FX and FXa use different mechanisms: the use of Rivaroxaban (RX) a specific inhibitor of FXa attenuated its anti-angiogenic properties but did not modify the anti-angiogenic effect of FX. Furthermore, only the anti-angiogenic activity of FXa is PAR-1dependent. Using in vivo models, we show that FX and FXa are anti-angiogenic in the zebrafish intersegmental vasculature (ISV) formation and in the chick embryo chorioallantoic membrane (CAM) assays. Our results provide further evidence for the non-hemostatic functions of FX and FXa and demonstrate for the first time a biological role for the zymogen FX.


Angiogenesis Inhibitors/pharmacology , Factor Xa/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Chick Embryo , Factor X/pharmacology , Factor X/therapeutic use , Factor Xa/therapeutic use , Helminth Proteins/pharmacology , Helminth Proteins/therapeutic use , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Neovascularization, Pathologic/drug therapy , Neovascularization, Physiologic/drug effects , Receptor, PAR-1/metabolism , Zebrafish
16.
Biol Reprod ; 89(1): 9, 2013 Jul.
Article En | MEDLINE | ID: mdl-23699387

Ovarian granulosa cell tumors (GCTs) represent 3%-5% of all ovarian malignancies. Treatments have limited proven efficacy and biologically targeted treatment is lacking. The aim of this study was to investigate the role of Notch signaling in the proliferation, steroidogenesis, apoptosis, and phosphatidylinositol 3-kinase (PI3K)/AKT pathway in a FOXL2-mutated granulosa tumor cell line (KGN) representative of the adult form of GCTs. When Notch signaling is initiated, the receptors expose a cleavage site in the extracellular domain to the metalloproteinase TACE and, following this cleavage, Notch undergoes another cleavage mediated by the presenilin-gamma-secretase complex. To achieve our goal, DAPT, an inhibitor of the gamma-secretase complex, was used to investigate the role of the Notch system in parameters associated with cell growth and death, using a human granulosa cell tumor line (KGN) as an experimental model. We observed that JAGGED1, DLL4, NOTCH1, and NOTCH4 were highly expressed in KGN cells as compared to granulosa-lutein cells obtained from assisted reproductive techniques patients. The proliferation and viability of KGN cells, as well as progesterone and estradiol production, decreased in the presence of 20 µM DAPT. Apoptotic parameters like PARP and caspase 8 cleavages, BAX, and BCLXs increased in KGN cells cultured with DAPT, whereas others such as BCL2, BCLXl, FAS, and FAS ligand did not change. AKT phosphorylation decreased and PTEN protein increased when Notch signaling was inhibited in KGN cells. We conclude that the Notch system acts as a survival pathway in KGN cells, and might be interacting with the PI3K/AKT pathway.


Amyloid Precursor Protein Secretases/antagonists & inhibitors , Apoptosis/drug effects , Cell Proliferation/drug effects , Forkhead Transcription Factors/genetics , Granulosa Cell Tumor/metabolism , Ovarian Neoplasms/metabolism , Receptors, Notch/metabolism , Adaptor Proteins, Signal Transducing , Amyloid Precursor Protein Secretases/metabolism , Calcium-Binding Proteins/metabolism , Cell Line, Tumor , Dipeptides , Female , Forkhead Box Protein L2 , Gonadal Steroid Hormones/biosynthesis , Granulosa Cell Tumor/genetics , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein , Membrane Proteins/metabolism , Mutation , Ovarian Neoplasms/genetics , Receptors, Notch/antagonists & inhibitors , Serrate-Jagged Proteins
17.
Endocrinology ; 153(7): 3446-56, 2012 Jul.
Article En | MEDLINE | ID: mdl-22577112

Polycystic ovary syndrome (PCOS) is the most common endocrinological pathology among women of reproductive age. It is characterized by anovulation, oligo- or amenorrhea, hyperandrogenism, obesity, and insulin resistance. PCOS patients present with elevated levels of vascular endothelial growth factor (VEGF) in serum and follicular fluid. In this study, we examined the ovarian expression of angiopoietins (ANGPT) and their receptor tyrosine kinase receptor (TIE2), involved in the stabilization of blood vessels, in a rat model of dehydroepiandrosterone-induced PCOS. We also analyzed the effect of ovarian VEGF inhibition on ANGPT/TIE2, follicular development, and vascular stability. VEGF levels were increased in the PCOS ovaries, whereas the levels of its receptor fetal liver kinase-1 were decreased. In addition, the periendothelial cell area and the ANGPT1 to ANGPT2 ratio in the ovary were increased in the PCOS group. Percentage of primary follicles was increased and the percentage of preantral follicles and corpora lutea was decreased in the PCOS group. VEGF inhibition decreased the percentage of primary follicles close to control values. Interestingly, despite the presence of cysts in the ovaries from VEGF inhibitor-treated PCOS rats, its percentage was lower than the PCOS group without treatment. In summary, this study describes an alteration not only in the VEGF/fetal liver kinase-1 system but also in the ANGPT/TIE2 system in a dehydroepiandrosterone-induced PCOS rat model. This leads to an increase in periendothelial cell recruitment. We also demonstrated that ovarian VEGF inhibition can partially restore the accumulation of small follicles in PCOS rats and reduces cyst formation, improving ovulation and follicular development. Therefore, the inhibition of VEGF could be considered, in addition to other currently applied treatments, as a new strategy to be studied in PCOS patients to restore ovarian function.


Angiopoietins/metabolism , Dehydroepiandrosterone/pharmacology , Gene Expression Regulation , Ovary/metabolism , Polycystic Ovary Syndrome/metabolism , Vascular Endothelial Growth Factor A/metabolism , Adjuvants, Immunologic/pharmacology , Animals , Disease Models, Animal , Endothelial Cells/cytology , Female , Immunohistochemistry/methods , Neovascularization, Pathologic , Ovulation , Rats , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor Receptor-2/biosynthesis
18.
Mol Cell Endocrinol ; 343(1-2): 79-87, 2011 Aug 22.
Article En | MEDLINE | ID: mdl-21704119

The aim of this study was to determine the effect of the local inhibition of ANGPT1 on steroid production, proliferation and apoptosis of ovarian follicular cells and on the PI3K/AKT pathway. We also examined the effect of ANGPTs on follicular cell apoptosis and proliferation in early antral follicles (EAFs) in culture. Follicular cells expressing PCNA decreased after ANGPT1 Ab treatment. Moreover, ANGPT1 inhibition increased the levels of active caspase 3 and androsterone, but decreased estradiol, AKT phosphorylation and the area of smooth muscle cell actin. In cultured EAFs from prepubertal rats treated with diethylstilbestrol (DES), ANGPT1 increased PCNA and decreased apoptosis while ANGPT2 reversed these effects. These results show that ANGPT1 alters steroidogenesis, reduces ovarian apoptosis, and stimulates cell proliferation in antral follicles. ANGPT1 may exert these roles by regulating ovarian vascular stability and/or by a direct effect on follicular cells, possibly involving the PI3K/AKT pathway.


Angiopoietin-1/metabolism , Apoptosis/physiology , Follicular Atresia/physiology , Ovarian Follicle/physiology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Angiopoietin-1/antagonists & inhibitors , Animals , Caspase 3/metabolism , Cell Proliferation , Cells, Cultured , Female , Humans , Ovarian Follicle/cytology , Ovarian Follicle/pathology , Rats , Rats, Sprague-Dawley , Steroids/biosynthesis
19.
Biol Reprod ; 84(5): 859-65, 2011 May.
Article En | MEDLINE | ID: mdl-21209419

We investigated the expression and cell localization of NOTCH1, NOTCH4, and the delta-like ligand DLL4 in corpus luteum (CL) from pregnant rats during prostaglandin F2alpha (PGF2alpha)-induced luteolysis. We also examined serum progesterone (P(4)) and CL proteins related to apoptosis after local administration of the notch inhibitor N-[N-(3,5-difluorophenacetyl-l-alanyl)]-S-phenylglycine t-butyl ester (DAPT). Specific staining for NOTCH1 and NOTCH4 receptors was detected predominantly in large and small luteal cells. Furthermore, in line with the fact that the notch intracellular domain is translocated to the nucleus, where it regulates gene expression, staining was evident in the nuclei of luteal cells. In addition, we detected diffuse cytoplasmic immunostaining for DLL4 in small and large luteal cells, in accordance with the fact that DLL4 undergoes proteolytic degradation after receptor binding. The mRNA expression of Notch1, Notch4, and Dll4 in CL isolated on Day 19 of pregnancy decreased significantly after administration of PGF2alpha. Consistent with the mRNA results, administration of PGF2alpha to pregnant rats on Day 19 of pregnancy decreased the protein fragment corresponding to the cleaved forms of NOTCH1/4 CL receptors. In contrast, no significant changes were detected in protein levels for the ligand DLL4. The local intrabursal administration of DAPT decreased serum P(4) levels and increased luteal levels of active caspase 3 and the BAX:BCL2 ratio 24 h after the treatment. These results support a luteotropic role for notch signaling to promote luteal cell viability and steroidogenesis, and they suggest that the luteolytic hormone PGF2alpha may act in part by reducing the expression of some notch system members.


Corpus Luteum/metabolism , Dinoprost/metabolism , Luteolysis/metabolism , Membrane Proteins/metabolism , Pregnancy Proteins/metabolism , Receptor, Notch1/metabolism , Receptors, Notch/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Cell Nucleus/metabolism , Corpus Luteum/drug effects , Corpus Luteum/ultrastructure , Female , Gene Expression Regulation , Intracellular Signaling Peptides and Proteins , Luteolysis/blood , Membrane Proteins/genetics , Peptide Fragments/metabolism , Pregnancy , Pregnancy Proteins/antagonists & inhibitors , Pregnancy Proteins/genetics , Progesterone/blood , Protease Inhibitors/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/genetics , Receptor, Notch4 , Receptors, Notch/antagonists & inhibitors , Receptors, Notch/genetics , Signal Transduction/drug effects
20.
Mol Cell Endocrinol ; 335(2): 116-25, 2011 Mar 30.
Article En | MEDLINE | ID: mdl-21238536

Ovarian hyperstimulation syndrome (OHSS) is a complication of ovarian stimulation with gonadotropins followed by the administration of human chorionic gonadotropin (hCG) to trigger the final steps of oocyte maturation. Gonadotropin-releasing hormone (GnRH) analogs are thought to be effective in preventing this complication and a clinical trial has found a lower incidence of OHSS in patients treated with these molecules. Our aim was to analyze the in vivo effect of a GnRH-I agonist on corpus luteum development and regression, ANGPT-1, ANGPT-2 and Tie-2 protein expression and luteal blood vessel stabilization, the expression of the steroidogenic acute regulatory protein (StAR) and the cytochrome P450 side-chain cleavage enzyme (P450scc) and cell proliferation, in ovaries from an OHSS rat model. To this end immature female Sprague-Dawley rats were hyperstimulated and treated with a GnRH-I agonist from the start of pregnant mare serum gonadotropin (PMSG) administration until the day of hCG injection for 5 consecutive days. Blood and tissue samples were collected 48h after hCG injection. Vascular endothelial growth factor VEGF levels were evaluated in the peritoneal fluid by ELISA. Serum progesterone and estradiol were measured by RIA. Histological features of sectioned ovaries were assessed in hematoxylin and eosin (H&E) stained slides. Luteal blood vessel stability, cell proliferation and apoptosis were assessed by immunohistochemistry for SMCA, PCNA, and TUNEL, respectively. P450scc, StAR, FLK-1, ANGPT-1, ANGPT-2, Tie-2 and PCNA protein levels were evaluated by Western blot from dissected corpora lutea (CL). The treatment with the GnRH-I agonist significantly decreased serum progesterone and estradiol levels as well as P450scc and StAR protein expression in the untreated OHSS group. In addition, the agonist significantly decreased the number of CL in the OHSS group, as compared with the untreated OHSS group. In the OHSS group, the area of periendothelial cells in the CL was larger than that of the control group. However, the treatment with the GnRH-I agonist significantly reduced the area of periendothelial cells in the CL in the OHSS group. The luteal levels of ANGPT-1 and its receptor Tie-2 significantly increased in the OHSS group when compared with the control group. Conversely, the administration of the GnRH-I agonist significantly decreased the levels of these factors in the CL from the OHSS group, as compared with the untreated OHSS group. In addition, the treatment with the GnRH-I agonist reduced the diameter of CL and decreased CL cell proliferation as compared with that observed in the untreated OHSS group. Finally, the GnRH-I agonist increased apoptosis in the CL from the OHSS group. In conclusion, these results show that GnRH-I agonist exerts diverse actions on the CL from a rat OHSS model. The decrease in P450scc, StAR, ANGPT-1 and Tie-2 expression, blood vessel stability and luteal proliferation leads to CL regression in the ovaries from OHSS rats. Moreover, our results suggest that the downregulation of ANGPT-1 and its receptor is a possible mechanism whereby GnRH-I agonists could prevent early OHSS.


Apoptosis/drug effects , Blood Vessels/drug effects , Corpus Luteum/drug effects , Fertility Agents, Female/adverse effects , Gonadotropins, Equine/adverse effects , Leuprolide/adverse effects , Ovarian Hyperstimulation Syndrome/chemically induced , Angiopoietin-1/metabolism , Angiopoietin-2/metabolism , Animals , Blood Vessels/pathology , Cell Proliferation/drug effects , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Corpus Luteum/blood supply , Corpus Luteum/growth & development , Corpus Luteum/pathology , Female , Gonadal Steroid Hormones/blood , Ovarian Hyperstimulation Syndrome/pathology , Ovary/drug effects , Ovary/metabolism , Ovary/pathology , Phosphoproteins/metabolism , Rats , Rats, Sprague-Dawley , Receptor, TIE-2/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
...