Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 489
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.
Mol Cell Endocrinol ; 540: 111518, 2022 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-34808277

RESUMO

Glucose uptake increases in canine luteal cells under insulin treatment. We hypothesize that insulin also increases luteal cell steroidogenesis. Dogs underwent elective ovariohysterectomy from days 10-60 post ovulation and their corpora lutea (CL) and blood samples were collected. Deep RNA sequencing determined differentially expressed genes in CL; those related to insulin signaling and steroidogenesis were validated in vivo by qPCR and their respective proteins by Western blotting and immunofluorescence. Next, luteal cell cultures were stimulated with insulin with or without inhibition of MAPK14, MAP2K1 and PI3K. Studied proteins except P450 aromatase showed the same expression pattern of coding genes in vivo. The expression of HSD3B and CYP19A1 was higher in insulin-treated cells (P < 0.005). Following respective pathway blockades, the culture medium had decreased concentrations of progesterone (P4) and 17b-estradiol (E2) (P < 0.01). Our results indicate that insulin increases HSD3B and CYP19A1 expression via MAPK and PI3K, and contributes to the regulation of P4 and E2 production in canine luteal cells.


Assuntos
Insulina/farmacologia , Células Lúteas/efeitos dos fármacos , Esteroides/biossíntese , Animais , Células Cultivadas , Corpo Lúteo/efeitos dos fármacos , Corpo Lúteo/metabolismo , Cães , Estradiol/metabolismo , Feminino , Glucose/metabolismo , Células Lúteas/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Progesterona/metabolismo , Transdução de Sinais/efeitos dos fármacos
3.
Reprod Biol Endocrinol ; 19(1): 173, 2021 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-34838049

RESUMO

BACKGROUND: Bone morphogenetic protein 2 (BMP2), growth differentiation factor 8 (GDF8) and their functional receptors are expressed in human ovarian follicles, and these two intrafollicular factors play essential roles in regulating follicle development and luteal function. As BMP antagonists, gremlin1 (GREM1) and gremlin2 (GREM2) suppress BMP signaling through blockage of ligand-receptor binding. However, whether BMP2 regulates the expression of GREM1 and GREM2 in follicular development remains to be determined. METHODS: In the present study, we investigated the effect of BMP2 on the expression of GREM1 and GREM2 and the underlying mechanisms in human granulosa-lutein (hGL) cells. An established immortalized human granulosa cell line (SVOG) and primary hGL cells were used as study models. The expression of GREM1 and GREM2 were examined following cell incubation with BMP2 at different concentrations and time courses. The TGF-ß type I inhibitors (dorsomorphin, DMH-1 and SB431542) and small interfering RNAs targeting ALK2, ALK3, SMAD2/3, SMAD1/5/8 and SMAD4 were used to investigate the involvement of the SMAD-dependent pathway. RESULTS: Our results showed that BMP2 significantly increased the expression of GREM2 (but not GREM1) in a dose- and time-dependent manner. Using a dual inhibition approach combining kinase inhibitors and siRNA-mediated knockdown, we found that the BMP2-induced upregulation of GREM2 expression was mediated by the ALK2/3-SMAD1/5-SMAD4 signaling pathway. Moreover, we demonstrated that BMP2 pretreatment significantly attenuated the GDF8-induced phosphorylation of SMAD2 and SMAD3, and this suppressive effect was reversed by knocking down GREM2 expression. CONCLUSIONS: Our findings provide new insight into the molecular mechanisms by which BMP2 modulates the cellular activity induced by GDF8 through the upregulated expression of their antagonist (GREM2).


Assuntos
Proteína Morfogenética Óssea 2/farmacologia , Citocinas/biossíntese , Células Lúteas/metabolismo , Miostatina/antagonistas & inibidores , Transdução de Sinais/fisiologia , Regulação para Cima/efeitos dos fármacos , Linhagem Celular Transformada , Citocinas/genética , Relação Dose-Resposta a Droga , Feminino , Expressão Gênica , Humanos , Células Lúteas/efeitos dos fármacos , Proteínas Recombinantes/farmacologia , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/fisiologia
4.
Int J Biol Sci ; 17(9): 2338-2347, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34239360

RESUMO

Rationale: Growth differentiation factor-8 (GDF-8), also known as myostatin, belongs to the transforming growth factor-beta (TGF-ß) superfamily. GDF-8 is expressed in the ovary and regulates various ovarian functions. Ovarian hyperstimulation syndrome (OHSS) is one of the most serious disorders during in vitro fertilization treatment. Aromatase, encoded by the CYP19A1 gene, is the enzyme that catalyzes the final step in estradiol (E2) biosynthesis. It has been demonstrated that high serum E2 levels are associated with the development of OHSS. However, the effects of GDF-8 on aromatase expression and its roles in the pathogenesis of OHSS remain unclear. Methods: The effect of GDF-8 on aromatase expression and the underlying mechanisms were explored by a series of in vitro experiments in primary human granulosa-lutein (hGL) and KGN cells. Rat OHSS model and human follicular fluid samples were used to examine the roles of the GDF-8 system in the pathogenesis of OHSS. Results: We demonstrate that GDF-8 stimulates aromatase expression and E2 production in hGL and KGN cells. In addition, TGF-ß type I receptor ALK5-mediated SMAD2/3 signaling is required for GDF-8-induced aromatase expression and E2 production. Using a rat OHSS model, we show that the aromatase and GDF-8 levels are upregulated in the ovaries of OHSS rats. Blocking the function of ALK5 by the administration of its inhibitor, SB431542, alleviates OHSS symptoms and the upregulation of aromatase. Clinical results reveal that the protein levels of GDF-8 are upregulated in the follicular fluid of OHSS patients. Moreover, the expression of GDF-8 is increased in hGL cells of OHSS patients. Conclusions: This study helps to elucidate the mechanisms mediating the expression of aromatase in human granulosa cells, which may lead to the development of alternative therapeutic approaches for OHSS.


Assuntos
Aromatase/metabolismo , Estradiol/biossíntese , Miostatina/metabolismo , Síndrome de Hiperestimulação Ovariana/sangue , Síndrome de Hiperestimulação Ovariana/enzimologia , Animais , Benzamidas/farmacologia , Dioxóis/farmacologia , Feminino , Líquido Folicular/metabolismo , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/metabolismo , Humanos , Células Lúteas/efeitos dos fármacos , Células Lúteas/metabolismo , Ratos , Ratos Wistar , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína Smad2/metabolismo , Regulação para Cima
5.
J Assist Reprod Genet ; 38(9): 2301-2305, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34031766

RESUMO

OBJECTIVES: To evaluates the effect of different modes of final follicular maturation triggering on the degree of apoptosis of granulosa cells (GCs) and the potential effect on progesterone secretion. METHODS: Thirty patients undergoing controlled ovarian hyperstimulation for IVF who received hCG, GnRH agonist, or dual trigger for final follicular maturation were included in the study. Granulosa cells were obtained at the time of oocyte retrieval. The proportion of apoptotic cells was evaluated via TUNEL and immunohistochemistry. RESULTS: The proportion of apoptotic cells was significantly higher in the GnRH agonist-alone group compared to hCG-alone and the dual trigger groups (13.5 ± 1.5% vs. 7.8% ± 1.8 vs. 10.1% ± 2, respectively, P < 0.01). Moreover, the expression of active-caspase-3 was also significantly increased in the GnRH agonist-alone group compared with the hCG-alone and the dual trigger groups (15.5% ± 2.9 vs. 8.4% ± 1.6 vs. 12.7% ± 2.6, respectively, P < 0.01). The progesterone levels measured in the granulosa-luteal cell culture medium after 24 h of incubation were similar between the three groups. CONCLUSIONS: The levels of apoptosis are increased after GnRH agonist/dual trigger. The increased apoptosis might be one of the culprit of the subsequent premature demise of the corpus luteum post GnRH agonist trigger.


Assuntos
Apoptose , Gonadotropina Coriônica/farmacologia , Hormônio Liberador de Gonadotropina/agonistas , Infertilidade Masculina/fisiopatologia , Células Lúteas/patologia , Luteólise , Indução da Ovulação/métodos , Adulto , Feminino , Fertilização in vitro/métodos , Humanos , Células Lúteas/efeitos dos fármacos , Masculino , Recuperação de Oócitos , Gravidez , Substâncias para o Controle da Reprodução/farmacologia
6.
Biol Reprod ; 105(1): 137-147, 2021 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-33864060

RESUMO

In the present study, we investigated the effect of the synthetic analog of prostaglandin F2α (PGF2α)-cloprostenol-on cultured steroidogenic luteal cells of selected felid species over a 2-day culture period. The changes induced by cloprostenol were measured based on progesterone concentration and mRNA expression analysis of selected genes. Cloprostenol significantly reduced concentration of progesterone in cell culture medium of small luteal cells isolated from domestic cat corpora lutea (CL) at the development/maintenance stage (P < 0.05), but did not influence progesterone production in cultured cells from the regression stage. A decrease or complete silencing of progesterone production was also measured in cultured luteal cells of African lion (formation stage) and Javan leopard (development/maintenance stage). Gene-expression analysis by real-time PCR revealed that treatment with cloprostenol did not have an influence on expression of selected genes coding for enzymes of steroidogenesis (StAR, HSD3B, CYP11A1) or prostaglandin synthesis (PTGS2, PGES), nor did it effect hormone receptors (AR, ESR1, PGR, PTGER2), an anti-oxidative enzyme (SOD1) or factors of cell apoptosis (FAS, CASP3, TNFRSF1B, BCL2) over the studied period. Significant changes were measured only for expressions of luteinizing hormone (P < 0.05), prolactin (P < 0.05) and PGF2α receptors (P < 0.005) (LHCGR, PRLR, and PTGFR). The obtained results confirm that PGF2α/cloprostenol is a luteolytic agent in CL of felids and its impact on progesterone production depends on the developmental stage of the CL. Cloprostenol short-term treatment on luteal cells was associated only with functional but not structural changes related to luteal regression.


Assuntos
Gatos/fisiologia , Cloprostenol/farmacologia , Leões/fisiologia , Células Lúteas/efeitos dos fármacos , Luteólise/psicologia , Luteolíticos/farmacologia , Panthera/fisiologia , Animais , Células Cultivadas , Feminino
7.
Int J Mol Sci ; 22(7)2021 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-33808393

RESUMO

Regulation of oxidative stress (OS) is important to prevent damage to female reproductive physiology. While normal OS levels may have a regulatory role, high OS levels may negatively affect vital processes such as folliculogenesis or embryogenesis. The aim of this work was to study OS induced by glucose, a reactive oxygen species generator, or peroxynitrite, a reactive nitrogen species generator, in cultured human granulosa-lutein (hGL) cells from oocyte donors, analyzing expression of genes involved in oocyte maturation (FSHR, PAPP, and CYP19A1) and OS damage response (ALDH3A2). We also evaluated the effect of celastrol as an antioxidant. Our results showed that although both glucose and peroxynitrite produce OS increments in hGL cells, only peroxynitrite treatment increases ALDH3A2 and PAPP gene expression levels and decreases FSHR gene expression levels. Celastrol pre-treatment prevents this effect of peroxynitrite. Interestingly, when celastrol alone was added, we observed a reduction of the expression of all genes studied, which was independent of both OS inductors. In conclusion, regulation of OS imbalance by antioxidant substances such as celastrol may prevent negative effects of OS in female fertility. In addition to the antioxidant activity, celastrol may well have an independent role on regulation of gene expression in hGL cells.


Assuntos
Células da Granulosa/metabolismo , Células Lúteas/metabolismo , Triterpenos Pentacíclicos/farmacologia , Adulto , Aromatase/genética , Células Cultivadas , Feminino , Expressão Gênica/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Células da Granulosa/efeitos dos fármacos , Humanos , Células Lúteas/efeitos dos fármacos , Oócitos/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Triterpenos Pentacíclicos/metabolismo , Proteína Plasmática A Associada à Gravidez/genética , Cultura Primária de Células , Receptores do FSH/genética
8.
J Chin Med Assoc ; 84(4): 389-399, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33784266

RESUMO

BACKGROUND: Kisspeptin (KISS1) and kisspeptin receptor (KISS1R) are essential gatekeepers of the reproductive system. The functions of KISS1 and KISS1R in corpus luteal cells remain ambiguous. The objective was to observe normal physiologic functions of corpus luteal cells in vivo and clarify the functions of KISS1 in vitro. METHODS: We conducted an in vivo observation of cellular patterns as well as the levels of steroidogenic enzymes and KISS1/KISS1R in corpus luteal cells obtained from female crossbred Taiwan native goats in the estrous cycle; the observation was performed using hematoxylin and eosin and immunohistochemistry staining. Subsequently, we used kisspeptin-10 (Kp-10) to stimulate temperature sensitive-caprine luteal cell line (ts-CLC-D) cells to investigate the progesterone (P4) levels, steroidogenic messenger RNA (mRNA)/protein levels, cell survival rate, intracellular Ca2+ concentration, and cell proliferation-related mRNA/protein levels in the mitogen-activated protein kinase pathway in vitro by applying immunofluorescence staining, Western blotting, 2,3-bis (2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide assay, and real-time polymerase chain reaction. RESULTS: We observed the presence of proteins and mRNAs for STAR, CYP11A1, HSD3B, KISS1, and KISS1R in the corpus luteal cells from goats in vivo. In vitro, the addition of Kp-10 reduced the P4 levels (p < 0.01) and increased cell proliferation (p < 0.05) of the ts-CLC-D cells. Furthermore, we found that the levels of proteins and mRNA for STAR, CYP11A1, and HSD3B decreased significantly when Kp-10 was added (p < 0.05). However, adding Kp-10 did not affect the mRNA levels for PLCG2, DAG1, PRKCA, KRAS, RAF1, MAP2K1, MAP2K2, MAPK3, MAPK1, and MAPK14. CONCLUSION: We determined that KISS1 could affect the P4 levels, steroidogenesis, and cell proliferation in luteal cells. However, further research is required to clarify how KISS1 regulates proliferation and steroid production in luteal cells.


Assuntos
Proliferação de Células/efeitos dos fármacos , Kisspeptinas/farmacologia , Células Lúteas/efeitos dos fármacos , Animais , Sobrevivência Celular , Feminino , Expressão Gênica/genética , Cabras , Reação em Cadeia da Polimerase , RNA Mensageiro , Taiwan
9.
Reprod Domest Anim ; 56(5): 744-753, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33560544

RESUMO

The fate of the corpus luteum, a transient endocrine gland formed and degraded during an oestrous cycle, is decided by various physiological factors, such as luteinizing hormone (LH). As a stimulator of progesterone, LH is known to maintain corpus luteum functional and structural integrity by inhibiting apoptosis, a programmed cell death. Therefore, we aim to investigate its action during the mid-luteal phase hypothesized that LH suppresses the death mechanism of bovine luteal steroidogenic cells (LSC) by analysing the expression of proteins involved. Cultured bovine LSC obtained from corpus luteum were treated for 24 hr with recombinant TNF and IFNG in the presence or absence of LH. The result showed that LH proved to have a protective effect by increased cell viability (p < .05) and prevented DNA fragmentation (p < .05), as demonstrated by the WST-1 colorimetric assay and TUNEL assay. Expression analysis of mRNA and protein levels showed that LH altered the expression of BCL2 (p < .05), CASP3 (p < .05), FAS (p < .05), and BAX (p < .05) to support cell survival. In conclusion, our study suggests that LH prolongs the corpus luteum life span through the anti-apoptotic mechanism by increasing cell viability and suppressing apoptosis-related genes and protein expression.


Assuntos
Apoptose/efeitos dos fármacos , Células Lúteas/efeitos dos fármacos , Hormônio Luteinizante/farmacologia , Animais , Apoptose/genética , Bovinos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Fragmentação do DNA/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Interferon gama , Fator de Necrose Tumoral alfa
10.
Biol Reprod ; 104(2): 430-444, 2021 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-33571374

RESUMO

Corpus luteum (CL) plays a critical role in mammalian reproductive physiology. Its dysfunction will lead to infertility or habitual abortion. In the current study, by use of melatonin specific membrane receptor 2 (MT2) knocking out (KO) mice model combined with RNA-Seq, immunohistochemistry, and immunofluorescence analyses, the genes of melatonin synthetic enzyme arylalkylamine N-acetyltransferase (AANAT) and MT2 were identified to strongly express in the CL of sows and mice. KO MT2 significantly impaired the reproductive performance in mice indicated by the reduced litter sizes. Melatonin treatment elevated the progesterone production in sows suggesting the improved CL function. Mechanistic analysis showed that melatonin upregulated a set of progesterone synthesis-related genes including cytochrome P450 family 11 subfamily A member 1 (Cyp11a1), aldo-keto reductase family 1, member C18 (Akr1c18), isopentenyl-diphosphate delta isomerase 1 (Idi1), and luteinizing hormone/choriogonadotropin receptor (Lhcgr). The upregulation of these genes directly related to the increased progesterone production. The regulatory effects of melatonin on these gene expressions were mediated by MT2 and MT2KO diminished the effects of melatonin in this respect. Thus, the presence of melatonergic system of AANAT, melatonin, and its receptor MT2 in CL is essential for reproductive success in mammals.


Assuntos
Arilalquilamina N-Acetiltransferase/metabolismo , Transtornos de Estresse por Calor/veterinária , Melatonina/metabolismo , Melatonina/farmacologia , Receptores de Melatonina/metabolismo , Ração Animal , Animais , Arilalquilamina N-Acetiltransferase/genética , Células Cultivadas , Relação Dose-Resposta a Droga , Feminino , Fertilidade , Regulação da Expressão Gênica/efeitos dos fármacos , Transtornos de Estresse por Calor/metabolismo , Células Lúteas/efeitos dos fármacos , Células Lúteas/metabolismo , Melatonina/administração & dosagem , Camundongos , Camundongos Knockout , Receptores de Melatonina/genética , Suínos
11.
Biol Reprod ; 104(2): 387-398, 2021 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-33112382

RESUMO

Endothelin-2 (EDN2) expression in granulosa cells was previously shown to be highly dependent on the hypoxic mediator, hypoxia inducible factor 1 alpha (HIF1A). Here, we investigated whether sirtuin-1 (SIRT1), by deacetylating HIF1A and class III histones, modulates EDN2 in human granulosa-lutein cells (hGLCs). We found that HIF1A was markedly suppressed in the presence of resveratrol or a specific SIRT1 activator, SRT2104. In turn, hypoxia reduced SIRT1 levels, implying a mutually inhibitory interaction between hypoxia (HIF1A) and SIRT1. Consistent with reduced HIF1A transcriptional activity, SIRT1 activators, resveratrol, SRT2104, and metformin, each acting via different mechanisms, significantly inhibited EDN2. In support, knockdown of SIRT1 with siRNA markedly elevated EDN2, whereas adding SRT2104 to SIRT1-silenced cells abolished the stimulatory effect of siSIRT1 on EDN2 levels further demonstrating that EDN2 is negatively correlated with SIRT1. Next, we investigated whether SIRT1 can also mediate the repression of the EDN2 promoter via histone modification. Chromatin immunoprecipitation (ChIP) analysis revealed that SIRT1 is indeed bound to the EDN2 promoter and that elevated SIRT1 induced a 40% decrease in the acetylation of histone H3, suggesting that SIRT1 inhibits EDN2 promoter activity by inducing a repressive histone configuration. Importantly, SIRT1 activation, using SRT2104 or resveratrol, decreased the viable numbers of hGLC, and silencing SIRT1 enhanced hGLC viability. This effect may be mediated by reducing HIF1A and EDN2 levels, shown to promote cell survival. Taken together, these findings propose novel, physiologically relevant roles for SIRT1 in downregulating EDN2 and survival of hGLCs.


Assuntos
Endotelina-2/metabolismo , Células da Granulosa/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Células Lúteas/metabolismo , Sirtuína 1/metabolismo , Antioxidantes/farmacologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Endotelina-2/genética , Epigênese Genética , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Células da Granulosa/efeitos dos fármacos , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Células Lúteas/efeitos dos fármacos , Oxigênio , RNA Interferente Pequeno , Resveratrol/farmacologia , Sirtuína 1/genética
12.
J Endocrinol ; 247(3): 283-294, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33108345

RESUMO

Vaspin, visceral adipose tissue-derived serine protease inhibitor, plays important roles in inflammation, obesity, and glucose metabolism. Our recent research has shown the expression and role of vaspin in the function of ovarian follicles. However, whether vaspin regulates steroidogenesis and luteolysis in the corpus luteum (CL) is still unknown. The aim of this study was first to determine the expression of vaspin and its receptor GRP78 in porcine CL at the early, middle, and late stages of the luteal phase. Next, we investigated the hormonal regulation of vaspin levels in luteal cells in response to luteinizing hormone (LH), progesterone (P4), and prostaglandin PGE2 and PGF2α. Finally, we determined vaspin's direct impact on luteal cells steroidogenesis, luteolysis and kinases phosphorylation. Our results are the first to show higher vaspin/GRP78 expression in middle and late vs early stages; immunohistochemistry showed cytoplasmic vaspin/GRP78 localization in small and large luteal cells. In vitro, we found that LH, P4, PGE2, and PGF2α significantly decreased vaspin levels. Furthermore, vaspin stimulated steroidogenesis by the activation of the GRP78 receptor and protein kinase A (PKA). Also, vaspin increased the ratio of luteotropic PGE2 to luteolytic PGF2α secretion via GRP78 and mitogen-activated kinase (MAP3/1). Moreover, vaspin, in a dose-dependent manner, decreased GRP78 expression, while it, in a time-dependent manner, increased kinases PKA and MAPK3/1 phosphorylation. Taken together, we found that vaspin/GRP78 expression depends on the luteal phase stage and vaspin affects luteal cells endocrinology, indicating that vaspin is a new regulator of luteal cells steroidogenesis and CL formation.


Assuntos
Corpo Lúteo/metabolismo , Células Lúteas/metabolismo , Serpinas/metabolismo , Animais , Corpo Lúteo/efeitos dos fármacos , Dinoprosta/farmacologia , Dinoprostona/farmacologia , Feminino , Proteínas de Choque Térmico/metabolismo , Células Lúteas/efeitos dos fármacos , Hormônio Luteinizante/farmacologia , Luteólise/efeitos dos fármacos , Progesterona/farmacologia , Serpinas/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Suínos
13.
Int J Mol Sci ; 21(18)2020 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-32957618

RESUMO

Formation and limited lifespan of corpus luteum (CL) are important for proper ovarian periodicity and fertility. Failed vascularization, imbalance between proliferation and apoptosis leads to luteal phase deficiency and infertility. The aim of this study was to examine the effect of vaspin on angiogenesis, apoptosis and proliferation as well as the involvement of 78-kDa glucose-regulated protein receptor (GRP78) and mitogen-activated kinase (MAP3/1) in these processes. Porcine luteal cells were incubated with vaspin (0.1-10 ng/mL) for 24 h to 72 h and then mRNA and protein expression of angiogenesis: vascular endothelial growth factor (VEGFA), fibroblast growth factor 2 (FGF2), angiopoietin 1 (ANGPT1), VEGFA receptors (VEGFR1, VEGFR2), apoptosis: caspase 3, bcl-2-like protein 4 (BAX), B-cell lymphoma (BCL2), and proliferation: proliferating cells nuclear antigen (PCNA), cyclin A factors as well as secretion of VEGFA, FGF2, ANGT1 were measured by real-time polymerase chain reaction (PCR), immunoblotting and enzyme-linked immunosorbent assay (ELISA), respectively. Moreover, apoptosis was assessed by caspase activity using the Caspase-Glo 3/7 assay, while proliferation was by alamarBlue. We found that vaspin enhanced luteal cell angiogenesis, proliferation, and significantly decreased apoptosis. Additionally, using GRP78 siRNA and the pharmacological inhibitor of MAP3/1 (PD98059), we observed that the effect of vaspin was reversed to the control level in all investigated processes. Taken together, our results suggest that vaspin is a new regulator of female fertility by direct regulation of CL formation and maintenance of luteal cell function.


Assuntos
Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Corpo Lúteo/efeitos dos fármacos , Proteínas de Choque Térmico/metabolismo , Células Lúteas/efeitos dos fármacos , MAP Quinase Quinase Quinase 1/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Serpinas/farmacologia , Angiopoietina-1/metabolismo , Animais , Apoptose/genética , Proliferação de Células/genética , Corpo Lúteo/citologia , Corpo Lúteo/metabolismo , Chaperona BiP do Retículo Endoplasmático , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Proteínas de Choque Térmico/genética , Células Lúteas/metabolismo , MAP Quinase Quinase Quinase 1/genética , Neovascularização Fisiológica/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Suínos , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
14.
FASEB J ; 34(11): 15591-15604, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32996643

RESUMO

Type I collagen, which is mainly composed of collagen type I alpha 1 chain (COL1A1), is the most abundant extracellular matrix (ECM) protein in the mammalian ovary; and the cyclical remodeling of the ECM plays an essential role in the regulation of corpus luteum formation. Our previous studies have demonstrated that TGF-ß1 is a potent inhibitor of luteinization in human granulosa-lutein (hGL) cells. Whether TGF-ß1 can regulate the expression of COL1A1 during the luteal phase remains to be elucidated. The aim of this study was to investigate the effect of TGF-ß1 on the regulation of COL1A1 expression and the underlying molecular mechanisms using an immortalized hGL cell line (SVOG cells) and primary hGL cells (obtained from 20 consenting patients undergoing IVF treatment). The results showed that TGF-ß1 significantly upregulated the expression of COL1A1. Using inhibition approaches, including pharmacological inhibition (a specific p38 inhibitor, SB203580, and a specific ERK1/2 inhibitor, U0126) and specific siRNA-mediated knockdown inhibition, we demonstrated that TGF-ß1 promoted the expression and production of COL1A1 in hGL cells, most likely via the ALK5-mediated p38 signaling pathway. Our findings provide insights into the molecular mechanisms by which TGF-ß1 promotes the deposition of type I collagen during the late follicular phase in humans.


Assuntos
Colágeno Tipo I/metabolismo , Células da Granulosa/metabolismo , Células Lúteas/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Células Cultivadas , Colágeno Tipo I/genética , Cadeia alfa 1 do Colágeno Tipo I , Feminino , Células da Granulosa/citologia , Células da Granulosa/efeitos dos fármacos , Humanos , Células Lúteas/citologia , Células Lúteas/efeitos dos fármacos , Receptor do Fator de Crescimento Transformador beta Tipo I/genética , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/genética
15.
Exp Mol Med ; 52(8): 1341-1350, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32855437

RESUMO

Ovarian hyperstimulation syndrome (OHSS) is one of the most life-threatening and potentially fatal complications associated with controlled ovarian hyperstimulation (COH) during in vitro fertilization (IVF) treatment. Although the pathogenesis of OHSS remains unclear, elevated serum estradiol (E2) levels before human chorionic gonadotropin (hCG) administration are associated with the risk of OHSS. The pineal hormone melatonin and its receptors are expressed in human granulosa cells and have been shown to stimulate E2 production. However, the effect of melatonin on the expression of aromatase, an enzyme responsible for a key step in the biosynthesis of E2, in human granulosa cells remains to be determined. Here, we demonstrate that melatonin upregulates aromatase expression in primary cultured human granulosa-lutein (hGL) cells through the melatonin receptor-mediated PKA-CREB pathway. Using a mouse model of OHSS, we demonstrate that administration of the melatonin receptor inhibitor luzindole inhibits the development of OHSS. In addition, the expression of ovarian aromatase and serum E2 levels are upregulated in OHSS mice compared to control mice, but this upregulation is attenuated by inhibition of the function of melatonin. Moreover, clinical results reveal that aromatase expression levels are upregulated in hGL cells from OHSS patients. Melatonin and E2 levels in the follicular fluid are significantly higher in OHSS patients than in non-OHSS patients. Furthermore, melatonin levels are positively correlated with E2 levels in follicular fluid. This study helps to elucidate the mechanisms mediating the expression of aromatase in hGL cells and provides a potential mechanism explaining the high E2 levels in patients with OHSS.


Assuntos
Aromatase/metabolismo , Estradiol/biossíntese , Células da Granulosa/metabolismo , Células Lúteas/metabolismo , Melatonina/farmacologia , Síndrome de Hiperestimulação Ovariana/sangue , Síndrome de Hiperestimulação Ovariana/enzimologia , Animais , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Feminino , Líquido Folicular/metabolismo , Células da Granulosa/efeitos dos fármacos , Humanos , Células Lúteas/efeitos dos fármacos , Camundongos Endogâmicos ICR , Receptores de Melatonina/metabolismo , Transdução de Sinais/efeitos dos fármacos
16.
Mol Cell Endocrinol ; 518: 110981, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32791190

RESUMO

Melatonin can be synthesized and secreted not only by the pineal gland but also by many extra-pineal tissues. It has been shown that many ovarian functions are regulated by melatonin locally. Ovarian hyperstimulation syndrome (OHSS) is a serious complication during ovulation induction of the in vitro fertilization treatment. To date, the etiology of OHSS is not fully understood. However, vascular endothelial growth factor (VEGF) is recognized as a critical mediator for the pathogenesis of OHSS. High expression of melatonin has been detected in the follicular fluid of OHSS patients. However, whether VEGF expression can be regulated by melatonin in human granulosa cells and further contributes to the pathogenesis of OHSS remain unknown. In this study, we show that melatonin stimulates VEGF expression in human granulosa-lutein (hGL) cells. Our results reveal that the MT2 receptor and activation of AKT are involved in melatonin-induced VEGF expression. Using a rat OHSS model, we report that the VEGF levels are up-regulated in the ovaries of OHSS rats. Blocking the melatonin system by administrating MT2 receptor antagonist, 4-P-PDOT, alleviates OHSS symptoms by decreasing the expression of VEGF. In addition, the expression levels of melatonin and VEGF in the follicular fluid of OHSS patients are up-regulated and positively correlated. This study demonstrates an important role for melatonin in regulating the pathogenesis of OHSS.


Assuntos
Células Lúteas/citologia , Melatonina/farmacologia , Síndrome de Hiperestimulação Ovariana/metabolismo , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Células Cultivadas , Modelos Animais de Doenças , Feminino , Humanos , Células Lúteas/efeitos dos fármacos , Células Lúteas/metabolismo , Síndrome de Hiperestimulação Ovariana/tratamento farmacológico , Síndrome de Hiperestimulação Ovariana/genética , Cultura Primária de Células , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Receptor MT2 de Melatonina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tetra-Hidronaftalenos/administração & dosagem , Tetra-Hidronaftalenos/farmacologia
17.
Reprod Biol ; 20(3): 273-281, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32741720

RESUMO

Sirtuin-1 (SIRT1), a NAD+-dependent deacetylase, is present in the ovarian granulosa cells (GCs) of various species. This study examined the regulation of SIRT1 expression in human granulosa-lutein cells (hGLCs). Two different, structurally unrelated SIRT1 activators, SRT2104 and resveratrol, dose- and time-dependently enhanced SIRT1 (∼2- and 1.5-fold increase at 50 µmol/L for mRNA and protein levels, respectively), whereas EX-527, an inhibitor of SIRT1 deacetylase activity, significantly suppressed SIRT1 protein induced by these activators. Transfecting cells with SIRT1 siRNA molecules efficiently silenced SIRT1 (∼70 % decrease in 48 h post-transfection). Furthermore, the stimulatory effects of SRT2104 on SIRT1 expression observed in non-transfected or in scrambled siRNA-transfected cells were diminished with SIRT1 silencing. The findings described above imply that SIRT1 autoregulates its own expression. Interestingly, SRT2104 elevated cAMP accumulation (1.4-fold) in the culture media of hGLCs which was further augmented in the presence of hCG (2.2-fold); these effects were evident after 12 h of incubation. This additive effect of hCG and SRT2104 on cAMP accumulation may explain the incremental outcome observed on SIRT1 expression (∼3-fold increase from basal level and ∼1.6-fold stimulation for each compound alone) with these two compounds. SIRT1 knockdown diminished SIRT1 induced by forskolin, providing additional evidence that cAMP promotes SIRT1. These findings imply that by activating adenylyl cyclase (hCG or forskolin) and inhibiting phosphodiesterases (SIRT1 activators), these two signals converge to produce an incremental, positive feedback loop on SIRT1 expression. Such a mechanism highlights the importance of maintaining high SIRT1 levels in human luteinized GCs.


Assuntos
AMP Cíclico/metabolismo , Células da Granulosa/metabolismo , Células Lúteas/metabolismo , Sirtuína 1/metabolismo , Adulto , Carbazóis/farmacologia , Linhagem Celular , Colforsina/farmacologia , Relação Dose-Resposta a Droga , Ativadores de Enzimas/farmacologia , Feminino , Células da Granulosa/efeitos dos fármacos , Compostos Heterocíclicos com 2 Anéis/farmacologia , Humanos , Células Lúteas/efeitos dos fármacos , RNA Interferente Pequeno , Resveratrol/farmacologia , Transdução de Sinais/efeitos dos fármacos , Sirtuína 1/genética
18.
Reproduction ; 160(4): 603-612, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32698154

RESUMO

Pentraxin 3 (PTX3), a multimeric glycoprotein, is implicated in various biological functions. PTX3 was shown to be elevated in the corpus luteum (CL) of early pregnant ewes; however, its role in sheep or other ruminants' CL during this reproductive stage or how it is regulated remain unknown. Here we explored the role of PTX3 and its relationship with interferon-tau (IFNT; the pregnancy recognition signaling molecule during early pregnancy in domestic ruminants) in bovine luteinized granulosa cells (LGCs). IFNT robustly elevated PTX3 expression in bovine LGCs, and significantly stimulated its expression in luteal endothelial cells, along with CL slices; yet, LGCs were the most responsive and sensitive among these luteal models. ALK2/ALK3/ALK6 kinase inhibitor, dorsomorphin, dose-dependently inhibited basal and IFNT-elevated PTX3 expression in LGCs. In contrast, ALK4/5/7 inhibitor, SB431542, did not alter basal and TGFB1-induced PTX3. We found that recombinant human PTX3 itself moderately but significantly increases LGC numbers. Because PTX3 is highly expressed in bovine LGCs, we next examined the impact of lowering endogenous PTX3 levels with siRNA. PTX3 silencing decreased the viable cell numbers and reversed IFNT actions on cell viability, percentage of proliferating cells, and on two key survival/death genes: BIRC5 encoding surviving protein, and FASL - a death-inducing signal. Interestingly, thrombospondin-1, a known luteal proapoptotic factor, was inversely related to PTX3 in LGCs. Together, these findings suggest a novel role for PTX3 during early pregnancy, as mediator of IFNT prosurvival actions supporting CL maintenance during this reproductive stage.


Assuntos
Proteína C-Reativa/metabolismo , Corpo Lúteo/citologia , Regulação da Expressão Gênica/efeitos dos fármacos , Interferon Tipo I/farmacologia , Células Lúteas/citologia , Proteínas da Gravidez/farmacologia , Componente Amiloide P Sérico/metabolismo , Animais , Proteína C-Reativa/genética , Bovinos , Corpo Lúteo/efeitos dos fármacos , Corpo Lúteo/metabolismo , Feminino , Células Lúteas/efeitos dos fármacos , Células Lúteas/metabolismo , Gravidez , Componente Amiloide P Sérico/genética
19.
Reprod Domest Anim ; 55(6): 683-690, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32125030

RESUMO

Bisphenol A (BPA), diethylhexyl phthalate (DEHP) and pentabrominated diphenyl ether 99 (PBDE 99) are environmental toxicants belonging to the endocrine disrupting compounds (EDCs). They exert adverse effects on the various physiological systems, especially the reproductive system of humans and animals. The aim of this study was to investigate the effects of BPA, DEHP and PBDE 99 on progesterone (P4) synthesis in cultured bovine luteal cells. The bovine luteal cells isolated from the mid-luteal corpora lutea were exposed to different concentrations of BPA (1, 3, 10 and 30 µM), DEHP (1, 3, 10 and 30 µM) and PBDE 99 (0.1, 0.3, 1 and 3 µM) in a serum-free culture media for 48 and 96 hr. At 48 hr, the P4 level in the luteal cells decreased after treatment with all concentrations of BPA; 3, 10 and 30 µM of DEHP; and 3 µM of PBDE 99 compared to the control (p < .05). Treatment of cells with 3-30 µM of BPA, 1-30 µM of DEHP and 1-3 µM of PBDE 99 for 96 hr resulted in reduction in P4 synthesis (p < .05). However, lower concentrations of PBDE 99 (0.1 and 0.3 µM) increased P4 levels at 48 and 96 hr. Synthesis of P4 was lower at 96 hr compared to the 48 hr in the groups treated with BPA (30 µM), DEHP (1-30 µM), PBDE 99 (0.3-3 µM) and control group. Our results showed that BPA, DEHP and PBDE 99 are able to alter luteal steroidogenesis in bovine cells and can disrupt hormonal balance in the ovary. However, it is necessary to evaluate the exact mechanism underlying these effects in future studies.


Assuntos
Disruptores Endócrinos/toxicidade , Células Lúteas/efeitos dos fármacos , Progesterona/metabolismo , Animais , Compostos Benzidrílicos/toxicidade , Bovinos , Células Cultivadas , Dietilexilftalato/toxicidade , Feminino , Éteres Difenil Halogenados/toxicidade , Células Lúteas/metabolismo , Fenóis/toxicidade
20.
J Endocrinol ; 245(1): 79-92, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31999623

RESUMO

Reproductive tract inflammatory disease (RTID) commonly occurs after the traumatic events of parturition and adversely affects follicular function. This study is the first to describe the cellular and steroidogenic characteristics of corpora lutea from cattle with RTID and the effects of pathogen-associated molecular patterns (PAMPs) on luteal angiogenesis and function in vitro. Luteal weight (P < 0.05) and progesterone content (P < 0.05) were reduced (1.2-fold) in cows with RTID, accompanied by reduced CYP11A (P < 0.05), HSD3B (P < 0.01) and STAR (P < 0.01) protein expression. Immunohistochemistry revealed that luteal vascularity (VWF) and pericyte (ACTA2) coverage were >3-fold lower in RTID cows (P < 0.05). To link these observations to bacterial infection and determine specificity of action, a physiologically relevant luteal angiogenesis culture system examined the effects of PAMPs on endothelial cell (EC) network formation and progesterone production, in the presence of pro-angiogenic factors. Luteal EC networks were reduced ≤95% (P < 0.05) by lipopolysaccharide (LPS, toll-like receptor (TLR) 4 agonist) but not by TLR2 agonists lipoteichoic acid or peptidoglycan. Conversely, progesterone production and steroidogenic protein expression were unaffected by PAMPs (P > 0.05). Moreover, the adverse effect of LPS on luteal EC networks was dose-dependent and effective from 1 ng/mL (P < 0.05), while few EC networks were present above 10 ng/mL LPS (P < 0.001). LPS reduced proliferation (P < 0.05) and increased apoptosis of EC (P < 0.001). The specific TLR4 inhibitor TAK242 reversed the effects of LPS on EC networks. In conclusion, luteal vasculature is adversely sensitive to LPS acting via TLR4, therefore ovarian exposure to LPS from any Gram-negative bacterial infection will profoundly influence subsequent reproductive potential.


Assuntos
Corpo Lúteo/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Células Lúteas/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Doenças Uterinas/metabolismo , Animais , Apoptose/efeitos dos fármacos , Bovinos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Corpo Lúteo/irrigação sanguínea , Corpo Lúteo/metabolismo , Família 11 do Citocromo P450/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Feminino , Humanos , Células Lúteas/metabolismo , Moléculas com Motivos Associados a Patógenos/metabolismo , Pericitos/efeitos dos fármacos , Pericitos/metabolismo , Fosfoproteínas/metabolismo , Progesterona/metabolismo , Receptor 4 Toll-Like/metabolismo , Doenças Uterinas/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...