Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 782
Filtrar
1.
Gynecol Endocrinol ; 40(1): 2351525, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38726683

RESUMO

OBJECTIVE: Stable luteal cell function is an important prerequisite for reproductive ability and embryonic development. However, luteal insufficiency seriously harms couples who have the desire to have a pregnancy, and the most important thing is that there is no complete solution. In addition, Vaspin has been shown to have regulatory effects on luteal cells, but the complex mechanisms involved have not been fully elucidated. Therefore, this study aimed to explore the effect of Vaspin on rat luteal cells and its mechanism. METHODS: Granulosa lutein cells separated from the ovary of female rats were incubated for 24h with gradient concentrations of Vaspin, and granulosa lutein cells incubated with 0.5% bovine serum albumin were used as controls. The proliferation, apoptosis, angiogenesis, progesterone (P4) and estradiol (E2) were detected by CCK-8, Anneixn-FITC/PI staining, angiogenesis experiment and ELISA. Western blot was applied to observe the expression levels of proteins related to cell proliferation, apoptosis, angiogenesis and MEK/MAPK signaling pathway. RESULTS: Compared with the Control group, Vaspin could significantly up-regulate the proliferation of granulosa lutein cells and reduce the apoptosis. Moreover, Vaspin promoted the angiogenesis of granulosa lutein cells and the production of P4 and E2 in a concentration-dependent manner. Furthermore, Vaspin up-regulated the CyclinD1, CyclinB1, Bcl2, VEGFA and FGF-2 expression in granulosa lutein cells, and down-regulated the level of Bax. Also, Vaspin increased the p-MEK1 and p-p38 levels. CONCLUSION: Vaspin can up-regulate the proliferation and steroidogenesis of rat luteal cells and reduce apoptosis, which may be related to the influence of MEK/MAPK activity.


Assuntos
Apoptose , Proliferação de Células , Células Lúteas , Progesterona , Serpinas , Animais , Feminino , Proliferação de Células/efeitos dos fármacos , Serpinas/metabolismo , Serpinas/farmacologia , Ratos , Células Lúteas/efeitos dos fármacos , Células Lúteas/metabolismo , Apoptose/efeitos dos fármacos , Progesterona/farmacologia , Estradiol/farmacologia , Células Cultivadas , Ratos Sprague-Dawley , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos
2.
Theriogenology ; 223: 98-107, 2024 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-38697014

RESUMO

The ALDH1A1 gene encodes a cytoplasmic member of the aldehyde dehydrogenase 1 family, which plays an important role in regulating animal reproductive performance, including estrus cycle and embryonic development. The aim of this study was to characterize ALDH1A1 activity in ovaries of 3-5 year-old yaks and to determine its effects on cell proliferation, apoptosis, and progesterone secretion in luteal cells (LCs). The coding sequence (CDS) of the ALDH1A1 gene was cloned by reverse transcription-PCR and immunohistochemical analysis was used to confirm localization of the ALDH1A1 protein in the ovary. To assess the activity of ALDH1A1 in regulating progesterone secretion, si-ALDH1A1 was transfected into LCs in vitro and progesterone levels in LC supernatants were measured by ELISA. The interference efficiency was assessed by real-time quantitative PCR (RT-qPCR) and immunofluorescence staining, and cell proliferation and apoptosis were evaluated by EdU and TUNEL staining, respectively. The cloned ALDH1A1 sequence contained 1462 bp, encoding 487 amino acids. Immunohistochemical analysis showed that ALDH1A1 protein expression, which was significantly higher in LCs, was mainly found in antral follicles and the corpus luteum (CL). The expression of ALDH1A1 mRNA in LCs was effectively inhibited by si-ALDH1A1transfection, and progesterone secretion was markedly decreased along with the significant down-regulation of progesterone pathway-related genes, STAR, CYP11A1, CYP19A1, CYP17A1, 3ß-HSD, and HSD17B1. Knockdown of ALDH1A1 mRNA expression decreased cell proliferation and increased apoptosis in LCs. The mRNA expression of the proliferation-related genes, PCNA, CCND1, CCNB1 and CDC25A, was significantly down-regulated, while expression of the apoptosis-promoting CASP3 gene was significantly increased. In summary, we characterized the yak ALDH1A1 gene and revealed that ALDH1A1 knockdown promoted apoptosis, repressed cell proliferation, and decreased progesterone secretion by yak LCs, potentially by regulating the mRNA expression of genes related to proliferation, apoptosis, and progesterone synthesis and secretion.


Assuntos
Família Aldeído Desidrogenase 1 , Células Lúteas , Retinal Desidrogenase , Animais , Feminino , Bovinos/genética , Células Lúteas/metabolismo , Retinal Desidrogenase/genética , Retinal Desidrogenase/metabolismo , Família Aldeído Desidrogenase 1/genética , Família Aldeído Desidrogenase 1/metabolismo , Apoptose , Progesterona/metabolismo , Aldeído Desidrogenase/genética , Aldeído Desidrogenase/metabolismo , Proliferação de Células , Regulação da Expressão Gênica/fisiologia
3.
J Ovarian Res ; 17(1): 97, 2024 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-38720330

RESUMO

The epidermal growth factor (EGF)-like factors, comprising amphiregulin (AREG), betacellulin (BTC), and epiregulin (EREG), play a critical role in regulating the ovulatory process. Pentraxin 3 (PTX3), an essential ovulatory protein, is necessary for maintaining extracellular matrix (ECM) stability during cumulus expansion. The aim of this study was to investigate the impact of EGF-like factors, AREG, BTC, and EREG on the expression and production of PTX3 in human granulosa-lutein (hGL) cells and the molecular mechanisms involved. Our results demonstrated that AREG, BTC, and EREG could regulate follicular function by upregulating the expression and increasing the production of PTX3 in both primary (obtained from 20 consenting patients undergoing IVF treatment) and immortalized hGL cells. The upregulation of PTX3 expression was primarily facilitated by the activation of the extracellular signal-regulated kinase 1 and 2 (ERK1/2) signaling pathway, induced by these EGF-like factors. In addition, we found that the upregulation of PTX3 expression triggered by the EGF-like factors was completely reversed by either pretreatment with the epidermal growth factor receptor (EGFR) inhibitor, AG1478, or knockdown of EGFR, suggesting that EGFR is crucial for activating the ERK1/2 signaling pathway in hGL cells. Overall, our findings indicate that AREG, BTC, and EREG may modulate human cumulus expansion during the periovulatory stage through the upregulation of PTX3.


Assuntos
Anfirregulina , Betacelulina , Proteína C-Reativa , Epirregulina , Células Lúteas , Componente Amiloide P Sérico , Regulação para Cima , Feminino , Humanos , Anfirregulina/metabolismo , Anfirregulina/genética , Betacelulina/metabolismo , Proteína C-Reativa/metabolismo , Proteína C-Reativa/genética , Fator de Crescimento Epidérmico/metabolismo , Fator de Crescimento Epidérmico/farmacologia , Epirregulina/metabolismo , Epirregulina/genética , Receptores ErbB/metabolismo , Células Lúteas/metabolismo , Sistema de Sinalização das MAP Quinases , Componente Amiloide P Sérico/metabolismo , Componente Amiloide P Sérico/genética
4.
Biol Reprod ; 110(1): 154-168, 2024 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-37815939

RESUMO

Phoenixin is a neuropeptide with a well-established role in the central regulation of reproductive processes; however, knowledge regarding its role in the ovary is limited. One of the main active phoenixin isoforms is phoenixin-14, which acts through G protein-coupled receptor 173. Our research hypothesis was that phoenixin-14 is expressed in porcine corpus luteum and exerts luteotropic action by affecting the endocrine function of luteal cells through G protein-coupled receptor 173 and protein kinase signaling. Luteal cells were cultured to investigate the effect of phoenixin-14 (1-1000 nM) on endocrine function. We showed that phoenixin-14 and G protein-coupled receptor 173 are produced locally in porcine corpus luteum and their levels change during the estrous cycle. We detected phoenixin-14 immunostaining in the cytoplasm and G protein-coupled receptor 173 in the cell membrane. Plasma phoenixin levels were highest during the early luteal phase. Interestingly, insulin, luteinizing hormone, progesterone, and prostaglandins decreased phoenixin-14 levels in luteal cells. Phoenixin-14 increased progesterone, estradiol, and prostaglandin E2 secretion, but decreased prostaglandin F2α, upregulated the expression of steroidogenic enzymes, and downregulated receptors for luteinizing hormone and prostaglandin. Also, phoenixin-14 increased the expression of G protein-coupled receptor 173 and the phosphorylation of extracellular signal-regulated kinase 1/2, protein kinase B, inhibited the phosphorylation of protein kinase A, and had mixed effect on AMP-activated protein kinase alpha and protein kinase C. G protein-coupled receptor 173 and extracellular signal-regulated kinase 1/2 mediated the effect of phoenixin-14 on endocrine function of luteal cells. Our results suggest that phoenixin is produced by porcine luteal cells and can be a new regulator of their function.


Assuntos
Células Lúteas , Feminino , Animais , Suínos , Células Lúteas/metabolismo , Progesterona/farmacologia , Corpo Lúteo/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Hormônio Luteinizante/farmacologia , Hormônio Luteinizante/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
5.
Mol Cell Endocrinol ; 582: 112126, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38109991

RESUMO

Ovarian steroidogenesis mediated by granulosa cells is pivotal in maintaining normal female reproductive function. The steroidogenic acute regulatory protein (StAR) regulates the rate-limiting step in steroidogenesis. Bone morphogenetic protein-9 (BMP-9), also known as growth differentiation factor-2 (GDF-2), is a member of the transforming growth factor-beta (TGF-ß) superfamily. BMP-9 induces epithelial-mesenchymal transition (EMT) that contributes to cancer progression. However, the function of BMP-9 in the female reproductive system remains largely unknown. It has been recently shown that BMP-9 is expressed in human follicular fluid and can downregulate StAR expression in human ovarian granulosa cells. However, the underlying molecular mechanisms warrant investigation. Our results show that treatment of primary granulosa-lutein (hGL) cells with BMP-9 downregulates StAR expression. In addition, two EMT-related transcription factors, Snail and Slug, are upregulated by the treatment of BMP-9. Using pharmacological inhibitors and a siRNA-mediated knockdown approach, we show that BMP-9 upregulates Snail and Slug expression by activating SMAD1/5/8 signaling. We also examine the effects of BMP-9 on SMAD-independent signaling pathways, including ERK1/2, p38, JNK, AKT, and CREB. However, none of them is affected by the BMP-9. Moreover, we use gain- and loss-of-function approaches to reveal that only Snail, not Slug, is required for the BMP-9-induced downregulation of StAR expression in hGL cells. This study increases the understanding of the physiology function of BMP-9 in hGL cells and provides important insights into the regulation of StAR expression.


Assuntos
Células Lúteas , Feminino , Humanos , Proteína Morfogenética Óssea 15/metabolismo , Proteína Morfogenética Óssea 15/farmacologia , Células Cultivadas , Células da Granulosa/metabolismo , Fator 2 de Diferenciação de Crescimento/metabolismo , Fator 2 de Diferenciação de Crescimento/farmacologia , Células Lúteas/metabolismo , Fosfoproteínas/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/farmacologia , Fator de Crescimento Transformador beta/metabolismo
6.
Reprod Biol ; 23(3): 100787, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37467532

RESUMO

Polycystic ovary syndrome (PCOS) is a complex heterogeneous disorder with reproductive and metabolic consequences whose aetiology is still elusive. To understand the cellular mechanisms that potentially govern follicular defect in women with PCOS, we performed transcriptomic profiles of granulosa-lutein cells (GLCs) by RNA-Seq analysis. We found differential expression of 876 genes in GLCs between PCOS and controls that belonged to various processes such as cell cycle, extracellular matrix organization, angiogenesis, oxidative stress, metabolism, etc. that support folliculogenesis, oocyte development, and maturation. The cross-talk between oocyte and GLCs is a fundamental cornerstone in determining oocyte quality and highly interlinked pathways of metabolism and redox homeostasis may influence this. We found several genes involved in the metabolism of carbohydrates, nucleotides, cholesterol, and lipids were dysregulated, which may impair the supply of metabolites to the growing oocyte, affecting oocyte development and competence. Additionally, high metabolic activity during folliculogenesis may augment oxidative damage to cells and macromolecules if not counter-balanced. We observed dysregulation of redox homeostasis and AGE-RAGE signalling in the follicular environment. Among the validated genes, prokineticin-1 and growth differentiation factor-15 were found to be negatively regulated, while, S100, calcium-binding protein A9 and angiomotin-like-2 were positively regulated in GLCs of women with PCOS. Comparing our data with previously published relevant transcriptomic studies showed metabolic, cytokine-cytokine receptor interaction, IL-17, and chemokine signalling pathways were most commonly affected in PCOS. Overall, this data can provide insights into mechanisms contributing to PCOS pathophysiology and can be explored as potential indicators for oocyte/embryo quality in IVF settings.


Assuntos
Células Lúteas , Síndrome do Ovário Policístico , Transcriptoma , Feminino , Humanos , Células Lúteas/metabolismo , Oócitos/metabolismo , Oogênese , Síndrome do Ovário Policístico/genética , Síndrome do Ovário Policístico/metabolismo , RNA-Seq
7.
J Ovarian Res ; 16(1): 103, 2023 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-37231448

RESUMO

BACKGROUND: The gap junction protein, connexin 43 (Cx43) is highly expressed in human granulosa-lutein (hGL) cells. The phosphorylation of certain amino acid residues in the Cx43 protein has been shown to be related to a decline in gap junction intercellular communication (GJIC), which subsequently affects oocyte meiotic resumption. As a member of the epidermal growth factor (EGF) family, betacellulin (BTC) mediates luteinizing hormone (LH)-induced oocyte maturation and cumulus cell expansion in mammalian follicles. Whether BTC can regulate Cx43 phosphorylation, which further reduces Cx43-coupled GJIC activity in hGL cells remains to be determined. METHODS: Immortalized human granulosa cells (SVOG cells) and primary human granulosa-lutein cells obtained from women undergoing in vitro fertilization in an academic research center were used as the study models. The expression levels of Cx43 and phosphorylated Cx43 were examined following cell incubation with BTC at different time points. Several kinase inhibitors (sotrastaurin, AG1478, and U0126) and small interfering RNAs targeting EGF receptor (EGFR) and receptor tyrosine-protein kinase 4 (ErbB4) were used to verify the specificity of the effects and to investigate the molecular mechanisms. Real-time-quantitative PCR and western blot analysis were used to detect the specific mRNA and protein levels, respectively. GJIC between SVOG cells were evaluated using a scrape loading and dye transfer assay. Results were analyzed by one-way analysis of variance. RESULTS: The results showed that BTC induced the rapid phosphorylation of Cx43 at serine368 without altering the expression of Cx43 in primary and immortalized hGL cells. Additionally, using a dual inhibition approach (kinase inhibitors and siRNA-based expression knockdown), we demonstrated that this effect was mainly mediated by the EGFR but not the ErbB4 receptor. Furthermore, using a protein kinase C (PKC) kinase assay and a scrape-loading and dye transfer assay, we revealed that PKC signaling is the downstream signaling pathway that mediates the increase in Cx43 phosphorylation and subsequent decrease in GJIC activity in response to BTC treatment in hGL cells. CONCLUSIONS: BTC promptly induced the phosphorylation of connexin 43 at Ser368, leading to decreased GJIC activity in hGL cells. The BTC-induced cellular activities were most likely driven by the EGFR-mediated PKC-dependent signaling pathway. Our findings shed light on the detailed molecular mechanisms by which BTC regulates the process of oocyte meiotic resumption.


Assuntos
Conexina 43 , Células Lúteas , Feminino , Humanos , Betacelulina/metabolismo , Betacelulina/farmacologia , Comunicação Celular , Conexina 43/genética , Conexina 43/metabolismo , Receptores ErbB/metabolismo , Junções Comunicantes/metabolismo , Células Lúteas/metabolismo , Mamíferos/metabolismo , Fosforilação
8.
Theriogenology ; 206: 40-48, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37178673

RESUMO

Evidence has shown that microRNA-665 (miR-665) is highly expressed in the mid-luteal phase compared with the early and end-luteal phase of the corpus luteum (CL) life cycle. However, whether miR-665 is a positive regulator of the life span of the CL is still unknown. The objective of this study is to explore the effect of miR-665 on the structural luteolysis in the ovarian CL. In this study, the targeting relationship between miR-665 and hematopoietic prostaglandin synthase (HPGDS) was firstly verified by dual luciferase reporter assay. Then, quantitative real-time PCR (qRT-PCR) was used to detect the expression of miR-665 and HPGDS in luteal cells. Following miR-665 overexpression, the apoptosis rate of the luteal cells was determined using flow cytometry; B-cell lymphoma-2 (BCL-2) and caspase-3 mRNA and protein were measured using qRT-PCR and Western blot (WB) analysis. Finally, the DP1 and CRTH2 receptors of PGD2, a synthetic product of HPGDS, were localized using immunofluorescence. Results confirmed that HPGDS was a direct target gene of miR-665, and miR-665 expression was negatively correlated with HPGDS mRNA expression in luteal cells. Meanwhile, after miR-665 was overexpressed, the apoptotic rate of the luteal cells showed a significant decrease (P < 0.05) and this was accompanied by elevated expression levels of anti-apoptotic factor BCL-2 mRNA and protein and decreased expression levels of apoptotic factor caspase-3 mRNA and protein (P < 0.01). Moreover, the immune fluorescence staining results showed that the DP1 receptor was also significantly decreased (P < 0.05), but the CRTH2 receptor was significantly increased (P < 0.05) in luteal cells. Overall, these results indicate that miR-665 reduces the apoptosis of luteal cells via inhibiting caspase-3 expression and promoting BCL-2 expression, and the biological function of miR-665 may be attributed to its target gene HPGDS which regulates the balance of DP1 and CRTH2 receptors expression in luteal cells. As a consequence, this study suggests that miR-665 might be a positive regulator of the life span of the CL rather than destroy the integrity of CL in small ruminants.


Assuntos
Células Lúteas , MicroRNAs , Feminino , Animais , Células Lúteas/metabolismo , Prostaglandina-Endoperóxido Sintases/metabolismo , Caspase 3/genética , Caspase 3/metabolismo , Corpo Lúteo/fisiologia , Apoptose/fisiologia , Ruminantes , RNA Mensageiro/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , MicroRNAs/metabolismo
9.
Curr Pharm Des ; 29(12): 947-956, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37013424

RESUMO

INTRODUCTION: This paper aims to reveal the molecular mechanism of resveratrol against oxidative stress and cell injury. The ovarian granulosa-lutein cell injury and apoptosis induced by oxidative stress may be responsible for female luteal phase deficiency. The antioxidant function of resveratrol has been confirmed; however, its effect on the expression of antioxidant enzymes and regulatory mechanisms in ovarian granulosa-lutein cells remains unclear. OBJECTIVE: This study aimed to investigate the role of the SIRT1/Nrf2/ARE signaling pathway in the effect of resveratrol on the hydrogen peroxide-induced injury of rat ovarian granulosa-lutein cells. METHODS: In this study, ovarian granulosa-lutein cells extracted from 3-week female SD rats were treated with 200 µM H2O2 in the presence or absence of 20 µM resveratrol. siRNA-SIRT1 and siRNA-Nrf2 were used to inhibit the expression of SIRT1 and Nrf2, respectively. Cell counting kit 8 (CCK-8), cellular morphology, progesterone secretion, and estradiol were used to evaluate cell injury. Hoechst 33258 staining was used to measure cell apoptosis. DHE staining, DCFH-DA staining, malondialdehyde content, protein carbonyl content, total antioxidant capacity and SOD viability were used to estimate the levels of oxidative stress. Western blot analysis was used to detect the levels of apoptosis-related proteins, and SIRT1/Nrf2/ARE signaling pathway-related proteins. RESULTS: The H2O2 treatment-induced rat ovarian granulosa-lutein cells injury was shown as decreased cell viability, impaired cellular morphology, and decreased levels of progesterone and estradiol. The H2O2 treatment also exacerbated cell apoptosis demonstrated as more apoptotic cells stained by Hoechst staining, decreased level of anti-apoptosis protein Bcl-2 and increased level of pro-apoptosis protein Bax. These effects of cell injury and apoptosis induced by H2O2 can be ameliorated by resveratrol. Resveratrol also alleviated oxidative stress induced by H2O2, supported by decreased superoxide anion and cellular total ROS, decreased malondialdehyde and protein carbonyl levels, and increased total antioxidant capacity and SOD viability. Western blot results demonstrated resveratrol reversed the H2O2-induced decrease in levels of antioxidant enzymes containing ARE sequences and activated SIRT1/Nrf2 pathway. Further treatment by siRNA-Nrf2 suggested resveratrol could not activate the expression of antioxidant enzymes under a condition of inhibition of Nrf2. CONCLUSION: This study demonstrates that resveratrol attenuated oxidative stress to protect H2O2-induced rat ovarian granulosa-lutein cell injury and apoptosis via SIRT1/Nrf2/ARE signaling pathway.


Assuntos
Antioxidantes , Células Lúteas , Ratos , Feminino , Animais , Resveratrol/farmacologia , Antioxidantes/farmacologia , Antioxidantes/metabolismo , Peróxido de Hidrogênio/farmacologia , Fator 2 Relacionado a NF-E2/metabolismo , Células Lúteas/metabolismo , Progesterona/metabolismo , Progesterona/farmacologia , Sirtuína 1/metabolismo , Carbonilação Proteica , Ratos Sprague-Dawley , Estresse Oxidativo , Transdução de Sinais , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas Reguladoras de Apoptose/farmacologia , RNA Interferente Pequeno/farmacologia , Estradiol/farmacologia , Superóxido Dismutase/metabolismo , Superóxido Dismutase/farmacologia , Malondialdeído/metabolismo , Malondialdeído/farmacologia , Espécies Reativas de Oxigênio/metabolismo
10.
Biol Reprod ; 108(5): 720-730, 2023 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-36881661

RESUMO

Sirtuin 1 (SIRT1) is a nicotinamide adenine dinucleotide-dependent deacetylase that modifies gene expression through histone deacetylation. It also deacetylates nonhistone substrates, e.g., tumor suppressor p53, NOS3, HIF1A, NFKB, FOXO3a, PGC-1α, and PPARγ. Consequently, it regulates a wide range of physiological functions including cell cycle control, energy expenditure, oxidative stress response, apoptosis, and aging. SIRT1 is expressed in ovarian granulosa cells (GCs) of various species including humans at different stages of the reproductive cycle. The importance of SIRT1 in female reproduction is supported by the findings that SIRT1-knockout mice exhibit defects in reproductive tissue development. These mice were found to have a thin-walled uterus, small ovaries, with follicles present but no corpora lutea. This review aims to provide state-of-the-art information on SIRT1's mode of action and its roles in human granulosa-lutein cells and GCs from other species where data are available. It also discusses the overlapping actions of SIRT1 and human chorionic gonadotropin on the production of critical GC-borne factors.


Assuntos
Células Lúteas , Sirtuína 1 , Animais , Feminino , Humanos , Camundongos , Gonadotropina Coriônica/farmacologia , Gonadotropina Coriônica/metabolismo , Células da Granulosa/metabolismo , Células Lúteas/metabolismo , Estresse Oxidativo , Sirtuína 1/genética , Sirtuína 1/metabolismo
11.
Sci Rep ; 13(1): 5085, 2023 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-36991037

RESUMO

Prokineticin 1 (PROK1) is a pleiotropic factor secreted by endocrine glands; however, its role has not been studied in the corpus luteum (CL) during pregnancy in any species. The present study aimed to investigate the contribution of PROK1 in regulating processes related to porcine CL function and regression: steroidogenesis, luteal cell apoptosis and viability, and angiogenesis. The luteal expression of PROK1 was greater on Days 12 and 14 of pregnancy compared to Day 9. PROK1 protein expression during pregnancy increased gradually and peaked on Day 14, when it was also significantly higher than that on Day 14 of the estrous cycle. Prokineticin receptor 1 (PROKR1) mRNA abundance increased on Days 12 and 14 of pregnancy, whereas PROKR2 elevated on Day 14 of the estrous cycle. PROK1, acting via PROKR1, stimulated the expression of genes involved in progesterone synthesis, as well as progesterone secretion by luteal tissue. PROK1-PROKR1 signaling reduced apoptosis and increased the viability of luteal cells. PROK1 acting through PROKR1 stimulated angiogenesis by increasing capillary-like structure formation by luteal endothelial cells and elevating angiogenin gene expression and VEGFA secretion by luteal tissue. Our results indicate that PROK1 regulates processes vital for maintaining luteal function during early pregnancy and the mid-luteal phase.


Assuntos
Células Lúteas , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina , Gravidez , Feminino , Animais , Suínos , Progesterona/metabolismo , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/genética , Células Endoteliais/metabolismo , Corpo Lúteo/metabolismo , Células Lúteas/metabolismo
12.
Mol Cell Endocrinol ; 566-567: 111891, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-36801432

RESUMO

A multitude of cytokines have been reported to participate in the folliculogenesis process in female. Interleukin-1 (IL-1), belonging to interleukin family, is originally identified as an important immune factor involved in inflammation response. Besides the immunity system, IL-1 is also expressed in reproductive system. However, the role of IL-1 in regulating ovarian follicle function remains to be elucidated. In the current study, using the primary human granulosa-lutein (hGL) and immortalized human granulosa-like tumor cell line (KGN) models, we demonstrated that both IL-1α and IL-1ß increased prostaglandin E2 (PGE2) production via upregulating its cyclooxygenase (COX) enzyme COX-2 expression in human granulosa cells. Mechanistically, IL-1α and IL-1ß treatment activated nuclear factor kappa B (NF-κB) signaling pathway. Using the specific siRNA to knock down endogenous gene expression, we found that the inhibition of p65 expression abolished IL-1α and IL-1ß-induced upregulation of COX-2 expression whereas knockdown of p50 and p52 had no effect. Moreover, our results also showed that IL-1α and IL-1ß promoted the nuclear translocation of p65. ChIP assay demonstrated the transcriptional regulation of p65 on COX-2 expression. Additionally, we also found that IL-1α and IL-1ß could activate the extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathway. The inhibition of ERK1/2 signaling pathway activation reversed IL-1α and IL-1ß-induced upregulation of COX-2 expression. Our findings shed light on the cellular and molecular mechanisms by which IL-1 modulates the COX-2 expression through NF-κB/P65 and ERK1/2 signaling pathways in human granulosa cells.


Assuntos
Células Lúteas , NF-kappa B , Humanos , Feminino , NF-kappa B/metabolismo , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Células Lúteas/metabolismo , Transdução de Sinais
13.
Theriogenology ; 198: 292-304, 2023 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-36634443

RESUMO

This study aimed to investigate the role of NR4A1 in forskolin (FSK)-induced granulosa cell (GC) differentiation and PGF2α-induced granulosa-lutein cell (GLC) regression. For experiment 1, primary porcine GCs were pre-cultured for 6 d before induced-differentiation by FSK with or without siNR4A1, and changes in GC proliferation, lipid droplets (LDs), and P4 level were detected. For experiment 2, the GLC model was established by FSK as in experiment 1, and then PGF2α was utilized to induce GLC regression with or without siNR4A1, changes in P4 secretion, apoptosis proteins, and associated signaling pathway members were detected. Results showed that in experiment 1, FSK up-regulated NR4A1 expression during GC differentiation and decreased GC proliferation activity, which was reversed by siNR4A1. siNR4A1 inhibited the FSK-induced decreases in Cyclin B1/D1 and CDK1/2 mRNA abundances, and increases in P21/P27 mRNA abundances, and FSK-induced LD accumulation. FSK up-regulated P4 secretion and StAR, CYP11A1 and HSD3B expression, decreased CYP19A1 expression, which were reversed by siNR4A1 except for StAR expression. In experiment 2, PGF2α induced NR4A1 expression and reduced GLC viability, which were reversed by siNR4A1. Compared with PGF2α group, the levels of P4 secretion and StAR expression were higher in PGF2α+siNR4A1 group, while CYP11A1 and HSD3B expressions held at low levels. siNR4A1 inhibited PGF2α-induced expression of apoptosis proteins (caspase3, Bax, Fas, TNFa), ATF3, and phosphorylated MAPKs (ERK1/2, P38, JNK). In summary, NR4A1 is involved in regulating porcine GC differentiation and GLC regression as well as the changes in cell proliferation, apoptosis, steroidogenesis, and MAPK pathways, which provide a theoretical basis for further understanding of the mechanism of porcine luteal formation and regression.


Assuntos
Células Lúteas , Animais , Feminino , Diferenciação Celular , Enzima de Clivagem da Cadeia Lateral do Colesterol/metabolismo , Células da Granulosa/metabolismo , Células Lúteas/metabolismo , Progesterona/metabolismo , RNA Mensageiro/metabolismo , Suínos , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo
14.
Hum Reprod ; 38(1): 103-112, 2023 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-36367827

RESUMO

STUDY QUESTION: Does LH addition to FSH in vitro recover the human primary granulosa lutein cell (hGLC) sub/poor-response? SUMMARY ANSWER: A picomolar concentration of LH may recover the FSH-induced cAMP and progesterone production of hGLC from sub/poor-responder women. WHAT IS KNOWN ALREADY: Clinical studies suggested that FSH and LH co-treatment may be beneficial for the ovarian response of sub/poor-responders undergoing ovarian stimulation during ART. STUDY DESIGN, SIZE, DURATION: hGLC samples from 286 anonymous women undergoing oocyte retrieval for ART were collected from October 2017 to February 2021. PARTICIPANTS/MATERIALS, SETTING, METHODS: hGLCs from women undergoing ovarian stimulation during ART were blindly purified, cultured, genotyped and treated in vitro by increasing concentrations of FSH (nM) ±0.5 nM LH. cAMP and progesterone levels produced after 3 and 24 h, respectively, were measured. In vitro data were stratified a posteriori, according to the donors' ovarian response, into normo-, sub- and poor-responder groups and statistically compared. The effects of LH addition to FSH were compared with those obtained by FSH alone in all the groups as well. MAIN RESULTS AND THE ROLE OF CHANCE: hGLCs from normo-responders were shown to have higher sensitivity to FSH treatment than sub-/poor-responders in vitro. Equimolar FSH concentrations induced higher cAMP (about 2.5- to 4.2-fold), and progesterone plateau levels (1.2- to 2.1-fold), in cells from normo-responder women than those from sub-/poor-responders (ANOVA; P < 0.05). The addition of LH to the cell treatment significantly increased overall FSH efficacy, indicated by cAMP and progesterone levels, within all groups (P > 0.05). Interestingly, these in vitro endpoints, collected from the normo-responder group treated with FSH alone, were similar to those obtained in the sub-/poor-responder group under FSH + LH treatment. No different allele frequencies and FSH receptor (FSHR) gene expression levels between groups were found, excluding genetics of gonadotropin and their receptors as a factor linked to the normo-, sub- and poor-response. In conclusion, FSH elicits phenotype-specific ovarian lutein cell response. Most importantly, LH addition may fill the gap between cAMP and steroid production patterns between normo- and sub/poor-responders. LIMITATIONS, REASONS FOR CAUTION: Although the number of experimental replicates is overall high for an in vitro study, clinical trials are required to demonstrate if the endpoints evaluated herein reflect parameters of successful ART. hGLC retrieved after ovarian stimulation may not fully reproduce the response to hormones of granulosa cells from the antral follicular stage. WIDER IMPLICATIONS OF THE FINDINGS: This in vitro assay may describe the individual response to personalize ART stimulation protocol, according to the normo-, sub- and poor-responder status. Moreover, this in vitro study supports the need to conduct optimally designed, randomized clinical trials exploring the personalized use of LH in assisted reproduction. STUDY FUNDING/COMPETING INTEREST(S): This study was supported by Merck KGaA. M.L. and C.C. are employees of Merck KGaA or of the affiliate Merck Serono SpA. Other authors have no competing interests to declare. TRIAL REGISTRATION NUMBER: N/A.


Assuntos
Hormônio Foliculoestimulante , Células Lúteas , Humanos , Feminino , Hormônio Foliculoestimulante/uso terapêutico , Células Lúteas/metabolismo , Progesterona , Gonadotropinas , Reprodução , Indução da Ovulação/métodos , Fertilização in vitro/métodos
15.
Int J Biol Macromol ; 224: 1118-1128, 2023 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-36302480

RESUMO

Cholesterol is a precursor to steroid hormones and can be obtained from serum LDL or de novo synthesis in steroidogenic cells. Before luteinizing hormone (LH) surge-induced ovulation, follicles remain avascular, and cholesterol required for progesterone production in granulosa cells (GCs) is derived from de novo biosynthesis. Previous studies have verified that the intrafollicular TGF-ß1 plays inhibitory roles in GCs luteinization, vascularization, and progesterone production. Nevertheless, the regulatory function of TGF-ß1 on de novo cholesterol synthesis in granulosa-lutein (GL) cells remains largely unknown. We aim to investigate this aspect in this study using in vivo cultured human GL cells. Our results suggested that TGF-ß1 significantly suppresses intracellular cholesterol levels and down-regulates the expression of the final step enzyme, DHCR24, that catalyzes de novo cholesterol synthesis. We used specific inhibitors and siRNA-mediated knockdown approaches demonstrate that TGF-ß1 suppression of DHCR24 expression in GL cells is mediated by the GSK-3ß/EZH2/H3K27me3 signaling pathway. Further ChIP assays revealed that elevated H3K27me3 levels in the promoter region of DHCR24 play a vital role in TGF-ß1-induced DHCR24 down-regulation, and RNA-sequencing results confirmed these findings. Notably, our study provides a novel insight into the molecular mechanisms by which TGF-ß1 suppresses de novo cholesterol biosynthesis in GL cells.


Assuntos
Células Lúteas , Oxirredutases atuantes sobre Doadores de Grupo CH-CH , Feminino , Humanos , Fator de Crescimento Transformador beta1/metabolismo , Células Lúteas/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Histonas/metabolismo , Progesterona , Células Cultivadas , Transdução de Sinais , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/metabolismo
16.
Anim Biotechnol ; 34(7): 2183-2196, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35678291

RESUMO

Luteal steroidogenesis is critical to implantation and pregnancy maintenance in mammals. The role of androgen receptors (AR) in the progesterone (P4) producing luteal cells of porcine corpus luteum (CL) remains unexplored. The aim of the present study was to establish AR gene knock out (KO) porcine luteal cell culture system model by CRISPR/Cas9 genome editing technology and to study the downstream effects of AR gene deficiency on steroidogenic potential and viability of luteal cells. For this purpose, genomic cleavage detection assay, microscopy, RT-qPCR, ELISA, annexin, MTT, and viability assay complemented by bioinformatics analysis were employed. There was significant downregulation (p < 0.05) in the relative mRNA expression of steroidogenic marker genes STAR, CYP11A1, HSD3B1 in AR KO luteal cells as compared to the control group, which was further validated by the significant (p < 0.05) decrease in the P4 production. Significant decrease (p < 0.05) in relative viability on third passage were also observed. The relative mRNA expression of hypoxia related gene HIF1A was significantly (p < 0.05) downregulated in AR KO luteal cells. Protein-protein interaction analysis mapped AR to signaling pathways associated with luteal cell functionality. These findings suggests that AR gene functionality is critical to luteal cell steroidogenesis in porcine.


Assuntos
Células Lúteas , Gravidez , Feminino , Suínos , Animais , Células Lúteas/química , Células Lúteas/metabolismo , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Corpo Lúteo/metabolismo , Progesterona/metabolismo , Progesterona/farmacologia , RNA Mensageiro/metabolismo , Mamíferos/metabolismo
17.
Domest Anim Endocrinol ; 82: 106763, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36166950

RESUMO

Studies have shown that ghrelin played direct actions in ovarian function, but the direct role of ghrelin in corpus luteum (CL) of pregnant sows has remained obscure. The study aimed to examine the expressions of ghrelin and its functional receptor (GHSR-1a) in the CL of sows during pregnancy, and evaluate the role of ghrelin in CL function of pregnant sows. Immunohistochemistry analysis showed that ghrelin and GHSR-1a are both predominantly localized in the luteal cells of pregnant sows CL. Strong immunoreactivity for ghrelin and GHSR-1a is detected at days 20 (early) and 50 (middle), but weak immunoreactivity is observed at days 90 (late) post mating. Similarly, there is a significant effect of pregnant phase on the expression (mRNA and protein) of ghrelin and GHSR-1a in the CL, with higher levels at days 20 (early) and 50 (middle), and lower values at 90 (late) post mating. In vitro, treatments of luteal cells with ghrelin (from 0.01 to 10 ng/mL) are promoted cell viability and P4 secretion in a dose-dependent manner. Ghrelin is also accelerated the LH-induced P4 secretion in luteal cells. Moreover, ghrelin is induced the release and mRNA expression of LH, and increased the release of prostaglandin (PG)E2, but reduced the secretion of PGF2α in luteal cells. In conclusion, the presences of ghrelin and GHSR-1a in the porcine CL during pregnancy, and the stimulatory effect of ghrelin on luteal cells suggest positive regulation by ghrelin in CL function of pregnant sows.


Assuntos
Grelina , Células Lúteas , Gravidez , Suínos , Feminino , Animais , Grelina/farmacologia , Corpo Lúteo/fisiologia , Receptores de Grelina/genética , Células Lúteas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
18.
Cell Commun Signal ; 20(1): 166, 2022 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-36284301

RESUMO

BACKGROUND: Heparin-binding epidermal growth factor-like growth factor (HB-EGF) belongs to the epidermal growth factor (EGF) family of growth factors. HB-EGF and its receptors, epidermal growth factor receptor (EGFR) and HER4, are expressed in the human corpus luteum. HB-EGF has been shown to regulate luteal function by preventing cell apoptosis. Steroidogenesis is the primary function of the human corpus luteum. Steroidogenic acute regulatory protein (StAR) plays a critical role in steroidogenesis. StAR expression and progesterone (P4) production in human granulosa-lutein (hGL) cells have been shown to be upregulated by a ligand of EGFR, amphiregulin. However, whether HB-EGF can achieve the same effects remains unknown. METHODS: A steroidogenic human ovarian granulosa-like tumor cell line, KGN, and primary culture of hGL cells obtained from patients undergoing in vitro fertilization treatment were used as experimental models. The underlying molecular mechanisms mediating the effects of HB-EGF on StAR expression and P4 production were explored by a series of in vitro experiments. RESULTS: Western blot showed that EGFR, HER2, and HER4 were expressed in both KGN and hGL cells. Treatment with HB-EGF for 24 h induced StAR expression but did not affect the expression of steroidogenesis-related enzymes, P450 side chain cleavage enzyme, 3ß-hydroxysteroid dehydrogenase, and aromatase. Using pharmacological inhibitors and a siRNA-mediated knockdown approach, we showed that EGFR, HER4, but not HER2, were required for HB-EGF-stimulated StAR expression and P4 production. In addition, HB-EGF-induced upregulations of StAR expression and P4 production were mediated by the activation of the ERK1/2 signaling pathway. CONCLUSION: This study increases the understanding of the physiological role of HB-EGF in human luteal functions. Video Abstract.


Assuntos
Células Lúteas , Feminino , Humanos , Células Lúteas/metabolismo , Progesterona/metabolismo , Aromatase/metabolismo , Aromatase/farmacologia , Anfirregulina/metabolismo , Anfirregulina/farmacologia , Fator de Crescimento Epidérmico/farmacologia , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/metabolismo , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/farmacologia , Sistema de Sinalização das MAP Quinases , RNA Interferente Pequeno/metabolismo , Ligantes , Luteína/metabolismo , Luteína/farmacologia , Fosfoproteínas/metabolismo , Transdução de Sinais , Receptores ErbB/metabolismo , Hidroxiesteroide Desidrogenases/metabolismo , Hidroxiesteroide Desidrogenases/farmacologia , Heparina/metabolismo , Heparina/farmacologia , Células da Granulosa/metabolismo , Células Cultivadas
19.
Eur J Obstet Gynecol Reprod Biol ; 276: 125-133, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35882072

RESUMO

Excessive production of reactive oxygen species (ROS) in granulosa cells (GCs) plays a role in pathogenesis of polycystic ovarian syndrome (PCOS) by developing oxidative stress (OS). It was shown that Sulforaphane (SFN), with known antioxidant properties, can have protective effects in different diseases through affecting the nuclear factor (erythroid-derived 2)-like 2 (NRF2) signaling pathway. Thus, the purpose of the current work was to examine the protective impact of SFN through the activation of the AMPK/AKT/NRF2 pathway against OS produced by H2O2 in granulosa-lutein cells (GLCs). Individuals' GLCs were obtained during ovum retrieval in intracytoplasmic sperm injection (ICSI) cycles. First, the induced OS model was created in GLCs using H2O2 exposure. To examine the protective effect of SFN against OS, the cells were cultured for 24 h in presence or absence of SFN. Eventually, the levels of intracellular ROS and apoptosis were measured by flow cytometry, and genes and proteins expression levels of AMPK, AKT, and NRF2 were evaluated using qRT-PCR and western blotting. Compared to the control group, the levels of intracellular ROS and apoptosis rose dramatically in GLCs with enhanced OS. SFN therapy decreased ROS and apoptosis levels and increased the overexpression of AMPK, AKT, and NRF2 genes and proteins. This study's results revealed that SFN exposure results in the alleviation of ROS and apoptosis levels possibly through activating the overexpression of genes and proteins of AMPK, AKT, and NRF2, and exerts its protective effects against OS in GLCs.


Assuntos
Peróxido de Hidrogênio , Células Lúteas , Proteínas Quinases Ativadas por AMP/metabolismo , Proteínas Quinases Ativadas por AMP/farmacologia , Apoptose , Feminino , Humanos , Peróxido de Hidrogênio/metabolismo , Peróxido de Hidrogênio/farmacologia , Isotiocianatos , Células Lúteas/metabolismo , Masculino , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-akt/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Sêmen/metabolismo , Sulfóxidos
20.
J Anim Sci ; 100(7)2022 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-35772754

RESUMO

Development of the corpus luteum (CL) requires the growth of a new capillary network from preexisting vasculature, a process known as angiogenesis. Successful building of this capillary network occurs through a sequence of cellular events-differentiation, proliferation, migration, and adhesion-which are regulated by a suite of angiogenic proteins that includes cellular communication network factor 1 (CCN1). We previously reported that the expression of CCN1 was highest in luteal tissue obtained from the early-cycle, 4-d-old bovine CL (i.e., corpus hemorrhagicum) compared to the mid- and late-cycle CL. In the present study, we treated steroidogenic bovine luteal cells from early-cycle CL with luteinizing hormone (LH), but it had no effect on CCN1 expression. Direct stimulation of the canonical LH pathway with forskolin and dibutyryl-cyclic adenosine monophosphate (cAMP), however, inhibited CCN1 mRNA expression. In endothelial cells, stimulation of Ras homolog family member A (RhoA) induces CCN1 expression, whereas RhoA inactivation inhibits it. Yet, it is unknown if regulation of CCN1 in steroidogenic luteal cells works likewise. We hypothesized that a similar mechanism of CCN1 regulation exists in bovine luteal cells and that thrombin, a known RhoA activator, may be a physiologic trigger for this mechanism in the early-cycle CL. To test this hypothesis, ovaries were collected from lactating dairy cows on days 3 or 4 of the estrous cycle, and corpora lutea were dissected and dissociated. Steroidogenic luteal cells were suspended in defined Ham's F12 medium, supplemented with insulin/transferrin/selenium and gentamicin, and seeded into 6-well plates. After 24 h, spent medium was replaced with fresh Ham's F12, and the cells were cultured for 24 to 48 h. Cells were treated for 2 h with defined medium, 10% fetal bovine serum (FBS), thrombin (1, 5, 10 U/mL), or Rho Activator II (0.25, 1, 2 µg/mL). Cells were then lysed for RNA extraction, followed by cDNA generation, and quantitative polymerase chain reaction (qPCR). Thrombin (1, 5, 10 U/mL; n = 3) and Rho Activator II (0.25, 1, 2 µg/mL; n = 6) increased (P < 0.05) CCN1 mRNA expression. In summary, CCN1 in bovine steroidogenic luteal cells was induced by thrombin and appeared to be regulated in a Rho-dependent manner. Future work will elucidate the signaling partners downstream of Rho which leads to CCN1 gene expression.


The corpus luteum (CL) is a transient ovarian endocrine gland that secretes progesterone, the hormone of pregnancy. Development of an optimally functioning CL requires the creation of a dense capillary bed through growth of new blood vessels, which is an intricate process called angiogenesis. A myriad of factors regulates angiogenesis, including the angiogenic inducer protein, cellular communication network factor 1 (CCN1). Although it is highly expressed in the early-cycle bovine CL, the mechanisms of CCN1 regulation have not been fully elucidated. In the present study, we showed that CCN1 expression in steroidogenic luteal cells from the early-cycle bovine CL was induced by Ras homolog family member A (RhoA) and by thrombin, but not by luteinizing hormone (LH). To the best of our knowledge, the involvement of thrombin and its signaling partner, RhoA, in regulating CCN1 in bovine steroidogenic luteal cells has not been previously reported. These findings will inform our future work to determine how RhoA activation by thrombin leads to increased expression of CCN1.


Assuntos
Células Lúteas , Animais , Bovinos , Corpo Lúteo , Células Endoteliais/metabolismo , Feminino , Lactação , Células Lúteas/metabolismo , Hormônio Luteinizante/metabolismo , Progesterona/metabolismo , RNA Mensageiro/metabolismo , Trombina/metabolismo , Trombina/farmacologia , Proteína rhoA de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA