Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
J Immunol ; 2022 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-36426987

RESUMO

The process of parturition is associated with inflammation within the uterine tissues, and IL-1ß is a key proinflammatory cytokine involved. Autophagy is emerging as an important pathway to remove redundant cellular components. However, it is not known whether IL-1ß employs the autophagy pathway to degrade collagen, thereby participating in membrane rupture at parturition. In this study, we investigated this issue in human amnion. Results showed that IL-1ß levels were significantly increased in human amnion obtained from deliveries with spontaneous labor and membrane rupture, which was accompanied by decreased abundance of COL1A1 and COL1A2 protein but not their mRNA, the two components of collagen I. Consistently, IL-1ß treatment of cultured primary human amnion fibroblasts reduced COL1A1 and COL1A2 protein but not their mRNA abundance along with increased abundance of autophagy activation markers, including the microtubule-associated protein L chain 3ß II/I ratio and autophagy-related 7 (ATG7) in the cells. The reduction in COL1A1 and COL1A2 protein abundance induced by IL-1ß could be blocked by the lysosome inhibitor chloroquine or small interfering RNA-mediated knockdown of ATG7 or ER-phagy receptor FAM134C, suggesting that FAM134C-mediated ER-phagy was involved in IL-1ß-induced reduction in COL1A1 and COL1A2 protein in amnion fibroblasts. Consistently, levels of L chain 3ß II/I ratio, ATG7, and FAM134C were significantly increased in human amnion obtained from deliveries with spontaneous labor and membrane rupture. Conclusively, increased IL-1ß abundance in human amnion may stimulate ER-phagy-mediated COL1A1 and COL1A2 protein degradation in amnion fibroblasts, thereby participating in membrane rupture at parturition.

2.
J Immunol ; 2022 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-36288908

RESUMO

The process of parturition is associated with inflammation within the uterine tissues, and IL-1ß is a key proinflammatory cytokine involved. Autophagy is emerging as an important pathway to remove redundant cellular components. However, it is not known whether IL-1ß employs the autophagy pathway to degrade collagen, thereby participating in membrane rupture at parturition. In this study, we investigated this issue in human amnion. Results showed that IL-1ß levels were significantly increased in human amnion obtained from deliveries with spontaneous labor and membrane rupture, which was accompanied by decreased abundance of COL1A1 and COL1A2 protein but not their mRNA, the two components of collagen I. Consistently, IL-1ß treatment of cultured primary human amnion fibroblasts reduced COL1A1 and COL1A2 protein but not their mRNA abundance along with increased abundance of autophagy activation markers, including the microtubule-associated protein L chain 3ß II/I ratio and autophagy-related 7 (ATG7) in the cells. The reduction in COL1A1 and COL1A2 protein abundance induced by IL-1ß could be blocked by the lysosome inhibitor chloroquine or small interfering RNA-mediated knockdown of ATG7 or ER-phagy receptor FAM134C, suggesting that FAM134C-mediated ER-phagy was involved in IL-1ß-induced reduction in COL1A1 and COL1A2 protein in amnion fibroblasts. Consistently, levels of L chain 3ß II/I ratio, ATG7, and FAM134C were significantly increased in human amnion obtained from deliveries with spontaneous labor and membrane rupture. Conclusively, increased IL-1ß abundance in human amnion may stimulate ER-phagy-mediated COL1A1 and COL1A2 protein degradation in amnion fibroblasts, thereby participating in membrane rupture at parturition.

3.
Mol Med ; 29(1): 88, 2023 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-37403020

RESUMO

BACKGROUND: Inflammation of the fetal membranes is an indispensable event of labor onset at both term and preterm birth. Interleukin-33 (IL-33) is known to participate in inflammation via ST2 (suppression of tumorigenicity 2) receptor as an inflammatory cytokine. However, it remains unknown whether IL-33/ST2 axis exists in human fetal membranes to promote inflammatory reactions in parturition. METHODS: The presence of IL-33 and ST2 and their changes at parturition were examined with transcriptomic sequencing, quantitative real-time polymerase chain reaction, Western blotting or immunohistochemistry in human amnion obtained from term and preterm birth with or without labor. Cultured primary human amnion fibroblasts were utilized to investigate the regulation and the role of IL-33/ST2 axis in the inflammation reactions. A mouse model was used to further study the role of IL-33 in parturition. RESULTS: Although IL-33 and ST2 expression were detected in both epithelial and fibroblast cells of human amnion, they are more abundant in amnion fibroblasts. Their abundance increased significantly in the amnion at both term and preterm birth with labor. Lipopolysaccharide, serum amyloid A1 and IL-1ß, the inflammatory mediators pertinent to labor onset, could all induce IL-33 expression through NF-κB activation in human amnion fibroblasts. In turn, via ST2 receptor, IL-33 induced the production of IL-1ß, IL-6 and PGE2 in human amnion fibroblasts via the MAPKs-NF-κB pathway. Moreover, IL-33 administration induced preterm birth in mice. CONCLUSION: IL-33/ST2 axis is present in human amnion fibroblasts, which is activated in both term and preterm labor. Activation of this axis leads to increased production of inflammatory factors pertinent to parturition, and results in preterm birth. Targeting the IL-33/ST2 axis may have potential value in the treatment of preterm birth.


Assuntos
Âmnio , Nascimento Prematuro , Animais , Feminino , Humanos , Recém-Nascido , Camundongos , Gravidez , Âmnio/metabolismo , Fibroblastos/metabolismo , Inflamação/metabolismo , Proteína 1 Semelhante a Receptor de Interleucina-1/genética , Interleucina-33 , NF-kappa B/metabolismo , Parto/metabolismo , Nascimento Prematuro/metabolismo
4.
Inflamm Res ; 72(4): 797-812, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36879064

RESUMO

OBJECTIVES: Sterile inflammation of fetal membranes is an indispensable event of normal parturition. However, triggers of sterile inflammation are not fully resolved. Serum amyloid A1 (SAA1) is an acute phase protein produced primarily by the liver. Fetal membranes can also synthesize SAA1 but its functions are not well defined. Given the role of SAA1 in the acute phase response to inflammation, we postulated that SAA1 synthesized in the fetal membranes may be a trigger of local inflammation at parturition. METHODS: The changes of SAA1 abundance in parturition were studied in the amnion of human fetal membranes. The role of SAA1 in chemokine expression and leukocyte chemotaxis was examined in cultured human amnion tissue explants as well as primary human amnion fibroblasts. The effects of SAA1 on monocytes, macrophages and dendritic cells were investigated in cells derived from a human leukemia monocytic cell line (THP-1). RESULTS: SAA1 synthesis increased significantly in human amnion at parturition. SAA1 evoked multiple chemotaxis pathways in human amnion fibroblasts along with upregulation of a series of chemokines via both toll-like receptor 4 (TLR4) and formyl peptide receptor 2 (FPR2). Moreover, SAA1-conditioned medium of cultured amnion fibroblasts was capable of chemoattracting virtually all types of mononuclear leukocytes, particularly monocytes and dendritic cells, which reconciled with the chemotactic activity of conditioned medium of cultured amnion tissue explants collected from spontaneous labor. Furthermore, SAA1 could induce the expression of genes associated with inflammation and extracellular matrix remodeling in monocytes, macrophages and dendritic cells derived from THP-1. CONCLUSIONS: SAA1 is a trigger of sterile inflammation of the fetal membranes at parturition.


Assuntos
Âmnio , Parto , Gravidez , Feminino , Humanos , Âmnio/metabolismo , Meios de Cultivo Condicionados/metabolismo , Meios de Cultivo Condicionados/farmacologia , Parto/genética , Parto/metabolismo , Membranas Extraembrionárias/metabolismo , Quimiocinas/metabolismo , Inflamação/metabolismo , Proteína Amiloide A Sérica
5.
Int J Mol Sci ; 24(13)2023 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-37446059

RESUMO

Inflammation of the fetal membranes is an indispensable event of parturition, with increasing prostaglandin E2 (PGE2) synthesis as one of the ultimate products that prime labor onset. In addition to PGE2, the fetal membranes also boast a large capacity for cortisol regeneration. It is intriguing how increased PGE2 synthesis is achieved in the presence of increasing amounts of classical anti-inflammatory glucocorticoids in the fetal membranes at parturition. 15(S)-hydroxyeicosatetraenoic acid (15(S)-HETE) synthesized by lipoxygenase 15/15B (ALOX15/15B) has been shown to enhance inflammation-induced PGE2 synthesis in amnion fibroblasts. Here, we examined whether glucocorticoids could induce ALOX15/15B expression and 15(S)-HETE production to promote PGE2 synthesis in amnion fibroblasts at parturition. We found that cortisol and 15(S)-HETE abundance increased parallelly in the amnion at parturition. Cortisol induced ALOX15/15B expression and 15(S)-HETE production paradoxically in amnion fibroblasts. Mechanism study revealed that this paradoxical induction was mediated by p300-mediated histone acetylation and interaction of glucocorticoid receptor with transcription factors CREB and STAT3. Conclusively, cortisol regenerated in the fetal membranes can paradoxically induce ALOX15/15B expression and 15(S)-HETE production in human amnion fibroblasts, which may further assist in the induction of PGE2 synthesis in the inflammatory responses of the fetal membranes for parturition.


Assuntos
Âmnio , Hidrocortisona , Gravidez , Feminino , Humanos , Hidrocortisona/metabolismo , Âmnio/metabolismo , Glucocorticoides/metabolismo , Dinoprostona/metabolismo , Parto , Membranas Extraembrionárias/metabolismo , Fibroblastos/metabolismo , Inflamação/metabolismo , Araquidonato 15-Lipoxigenase/metabolismo
6.
J Lipid Res ; 63(11): 100294, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36206855

RESUMO

Human parturition is associated with massive arachidonic acid (AA) mobilization in the amnion, indicating that large amounts of AA-derived eicosanoids are required for parturition. Prostaglandin E2 (PGE2) synthesized from the cyclooxygenase (COX) pathway is the best characterized AA-derived eicosanoid in the amnion which plays a pivotal role in parturition. The existence of any other pivotal AA-derived eicosanoids involved in parturition remains elusive. Here, we screened such eicosanoids in human amnion tissue with AA-targeted metabolomics and studied their role and synthesis in parturition by using human amnion fibroblasts and a mouse model. We found that lipoxygenase (ALOX) pathway-derived 15(S)-hydroxyeicosatetraenoic acid (15(S)-HETE) and its synthetic enzymes ALOX15 and ALOX15B were significantly increased in human amnion at parturition. Although 15(S)-HETE is ineffective on its own, it potently potentiated the activation of NF-κB by inflammatory mediators including lipopolysaccharide, interleukin-1ß, and serum amyloid A1, resulting in the amplification of COX-2 expression and PGE2 production in amnion fibroblasts. In turn, we determined that PGE2 induced ALOX15/15B expression and 15(S)-HETE production through its EP2 receptor-coupled PKA pathway, thereby forming a feed-forward loop between 15(S)-HETE and PGE2 production in the amnion at parturition. Our studies in pregnant mice showed that 15(S)-HETE injection induced preterm birth with increased COX-2 and PGE2 abundance in the fetal membranes and placenta. Conclusively, 15(S)-HETE is identified as another crucial parturition-pertinent AA-derived eicosanoid in the amnion, which may form a feed-forward loop with PGE2 in parturition. Interruption of this feed-forward loop may be of therapeutic value for the treatment of preterm birth.


Assuntos
Âmnio , Dinoprostona , Ácidos Hidroxieicosatetraenoicos , Nascimento Prematuro , Animais , Feminino , Humanos , Camundongos , Gravidez , Âmnio/metabolismo , Ciclo-Oxigenase 2/metabolismo , Ácidos Hidroxieicosatetraenoicos/farmacologia , Parto/metabolismo , Nascimento Prematuro/metabolismo
7.
BMC Med ; 20(1): 189, 2022 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-35610640

RESUMO

BACKGROUND: Enhancer of zeste homolog 2 (EZH2)-mediated histone 3 lysine 27 trimethylation (H3K27me3) is a transcription silencing mark, which is indispensable for cell lineage specification at the early blastocyst stage. This epigenetic repression is maintained in placental cytotrophoblasts but is lifted when cytotrophoblasts differentiate into syncytiotrophoblasts. However, the physiological impact of this lift remains elusive. Here, we investigated whether lifting EZH2-mediated H3K27me3 during syncytialization upregulates the expression of a short secretory isoform of a disintegrin and metalloprotease 12 (ADAM12-S), a well-recognized placenta-derived protease that cleaves insulin-like growth factor binding protein 3 to increase insulin-like growth factor (IGF) bioavailability for the stimulation of fetoplacental growth. The transcription factor and the upstream signal involved were also explored. METHODS: Human placenta tissue and cultured primary human placental cytotrophoblasts were utilized to investigate the role of EZH2-mediated H3K27me3 in ADAM12-S expression and the associated transcription factor and upstream signal during syncytialization. A mouse model was used to examine whether inhibition of EZH2-mediated H3K27me3 regulates placental ADAM12-S expression and fetoplacental growth. RESULTS: EZH2 and ADAM12 are distributed primarily in villous cytotrophoblasts and syncytiotrophoblasts, respectively. Increased ADAM12-S expression, decreased EZH2 expression, and decreased EZH2/H3K27me3 enrichment at the ADAM12 promoter were observed during syncytialization. Knock-down of EZH2 further increased ADAM12-S expression in trophoblasts. Syncytialization was also accompanied by increased STAT5B expression and phosphorylation as well as its enrichment at the ADAM12 promoter. Knock-down of STAT5B attenuated ADAM12-S expression during syncytialization. Epidermal growth factor (EGF) was capable of inducing ADAM12-S expression via stimulation of STAT5B expression and phosphorylation during syncytialization. Mouse studies revealed that administration of an EZH2 inhibitor significantly increased ADAM12-S levels in maternal blood and fetoplacental weights along with decreased H3K27me3 abundance and increased ADAM12-S expression in the placenta. CONCLUSIONS: Lifting EZH2-mediated H3K27me3 increases ADAM12-S expression during syncytialization with the participation of EGF-activated STAT5B, which may lead to elevation of ADAM12-S level in maternal blood resulting in increased IGF bioavailability for the stimulation of fetoplacental growth in pregnancy. Our studies suggest that the role of EZH2-mediated H3K27me3 may switch from cell lineage specification at the early blastocyst stage to regulation of fetoplacental growth in later gestation.


Assuntos
Proteína ADAM12 , Proteína Potenciadora do Homólogo 2 de Zeste , Histonas , Placenta , Proteína ADAM12/biossíntese , Proteína ADAM12/genética , Proteína ADAM12/metabolismo , Animais , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Fator de Crescimento Epidérmico/metabolismo , Feminino , Desenvolvimento Fetal , Histonas/metabolismo , Camundongos , Placenta/metabolismo , Placentação , Gravidez , Transdução de Sinais
8.
Reprod Biol Endocrinol ; 20(1): 4, 2022 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-34980155

RESUMO

BACKGROUND: Insulin resistance (IR) contributes to ovarian dysfunctions in polycystic ovarian syndrome (PCOS) patients. Serum amyloid A1 (SAA1) is an acute phase protein produced primarily by the liver in response to inflammation. In addition to its role in inflammation, SAA1 may participate in IR development in peripheral tissues. Yet, expressional regulation of SAA1 in the ovary and its role in the pathogenesis of ovarian IR in PCOS remain elusive. METHODS: Follicular fluid, granulosa cells and peripheral venous blood were collected from PCOS and non-PCOS patients with and without IR to measure SAA1 abundance for analysis of its correlation with IR status. The effects of SAA1 on its own expression and insulin signaling pathway were investigated in cultured primary granulosa cells. RESULTS: Ovarian granulosa cells were capable of producing SAA1, which could be induced by SAA1 per se. Moreover, the abundance of SAA1 significantly increased in granulosa cells and follicular fluid in PCOS patients with IR. SAA1 treatment significantly attenuated insulin-stimulated membrane translocation of glucose transporter 4 and glucose uptake in granulosa cells through induction of phosphatase and tensin homolog deleted on chromosome 10 (PTEN) expression with subsequent inhibition of Akt phosphorylation. These effects of SAA1 could be blocked by inhibitors for toll-like receptors 2/4 (TLR 2/4) and nuclear factor kappa light chain enhancer of activated B (NF-κB). CONCLUSIONS: Human granulosa cells are capable of feedforward production of SAA1, which significantly increased in PCOS patients with IR. Excessive SAA1 reduces insulin sensitivity in granulosa cells via induction of PTEN and subsequent inhibition of Akt phosphorylation upon activation of TLR2/4 and NF-κB pathway. These findings highlight that elevation of SAA1 in the ovary promotes the development of IR in granulosa cells of PCOS patients.


Assuntos
Células da Granulosa/metabolismo , Resistência à Insulina/genética , Síndrome do Ovário Policístico/genética , Proteína Amiloide A Sérica/fisiologia , Adulto , Estudos de Casos e Controles , Células Cultivadas , Feminino , Líquido Folicular/química , Líquido Folicular/metabolismo , Células da Granulosa/efeitos dos fármacos , Humanos , Ovário/efeitos dos fármacos , Ovário/metabolismo , Síndrome do Ovário Policístico/sangue , Síndrome do Ovário Policístico/metabolismo , Proteína Amiloide A Sérica/genética , Proteína Amiloide A Sérica/metabolismo , Proteína Amiloide A Sérica/farmacologia
9.
Mol Ther ; 29(3): 1279-1293, 2021 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-33212300

RESUMO

Polycystic ovary syndrome (PCOS) is an endocrine-related disease and global cause of infertility that is associated with abnormal folliculogenesis. Inhibited granulosa cell (GC) proliferation is recognized as a key factor that underlies aberrant follicle maturation. Many epigenetic landscape modifications have been characterized in PCOS patients. However, the epigenetic regulation pathways in follicular dysplasia are not completely understood. In this study, we reported a novel mechanism of DNA hypomethylation induced by long non-coding RNAs (lncRNAs) and its function in cell cycle progression. We observed that lnc-MAP3K13-7:1 was highly expressed in GCs from patients with PCOS, with concomitant global DNA hypomethylation, decreased DNA methyltransferase 1 (DNMT1) expression, and increased cyclin-dependent kinase inhibitor 1A (CDKN1A, p21) expression. In KGN cells, lnc-MAP3K13-7:1 overexpression resulted in cell cycle arrest in the G0/G1 phase, as well as the molecular inhibition and genetic silencing of DNMT1. Mechanistically, lnc-MAP3K13-7:1 inhibited DNMT1 expression by acting as a protein-binding scaffold and inducing ubiquitin-mediated DNMT1 protein degradation. Moreover, DNMT1-dependent CDKN1A promoter hypomethylation increased CDKN1A transcription, resulting in attenuated GC growth. Our work uncovered a novel and essential mechanism through which lnc-MAP3K13-7:1-dependent DNMT1 inhibition regulates CDKN1A/p21 expression and inhibits GC proliferation.


Assuntos
Inibidor de Quinase Dependente de Ciclina p21/metabolismo , DNA (Citosina-5-)-Metiltransferase 1/metabolismo , Metilação de DNA , Células da Granulosa/patologia , Ovário/patologia , Síndrome do Ovário Policístico/patologia , RNA Longo não Codificante/genética , Apoptose , Biomarcadores/metabolismo , Ciclo Celular , Movimento Celular , Proliferação de Células , Inibidor de Quinase Dependente de Ciclina p21/genética , DNA (Citosina-5-)-Metiltransferase 1/genética , Epigênese Genética , Feminino , Regulação da Expressão Gênica , Inativação Gênica , Células da Granulosa/metabolismo , Humanos , MAP Quinase Quinase Quinases/genética , Ovário/metabolismo , Síndrome do Ovário Policístico/genética , Síndrome do Ovário Policístico/metabolismo , Prognóstico , Regiões Promotoras Genéticas , Células Tumorais Cultivadas , Ubiquitinação
10.
FASEB J ; 34(7): 9563-9574, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32469087

RESUMO

Insulin resistance (IR) disrupts ovarian functions in polycystic ovary syndrome (PCOS). The contributing factors remains elusive. High mobility group box 1 (HMGB1), a damage-associated molecular pattern molecule, has been shown to be related to IR and autophagy, respectively, in peripheral tissues. Here, we investigated whether increased HMGB1 contributes to IR in granulosa cells of PCOS patients via induction of aberrant autophagy. Results showed that HMGB1 abundance in the follicular fluid was significantly increased with enhanced autophagy in granulosa cells in PCOS patients with IR. HMGB1 exacerbated autophagy in granulosa cells as evinced by increased LC3B II/I ratio and ATG7 as well as decreased p62, the markers for autophagy. Concurrently, HMGB1 impaired insulin sensitivities by attenuating the abundance of insulin receptor substrate-1, Akt phosphorylation, GLUT4 translocation, and glucose uptake in granulosa cells, which were reversed by blocking autophagy pathways with siRNA-mediated knockdown of ATG7 or with chloroquine and bafilomycin A1, the lysosome inhibitors. In conclusion, our results indicate that increased HMGB1 contributes to IR development in granulosa cells of PCOS patients, which is associated with exacerbation of autophagy by HMGB1. Control of HMGB1 production may be benefical for the improvement of insulin sensitivity in granulosa cells in PCOS.


Assuntos
Autofagia , Líquido Folicular/metabolismo , Células da Granulosa/patologia , Proteína HMGB1/metabolismo , Resistência à Insulina , Síndrome do Ovário Policístico/patologia , Adulto , Estudos de Casos e Controles , Feminino , Células da Granulosa/metabolismo , Proteína HMGB1/genética , Humanos , Fosforilação , Síndrome do Ovário Policístico/genética , Síndrome do Ovário Policístico/metabolismo , Transdução de Sinais
11.
FASEB J ; 33(7): 8148-8160, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30917001

RESUMO

Human amnion fibroblasts produce abundant prostaglandin E2 (PGE2), which plays a crucial role in parturition by stimulating not only myometrial contraction and cervical ripening but also the expression of the rate-limiting enzyme in PGE2 synthesis-namely, cyclooxygenase-2 (COX-2). This feed-forward induction of COX-2 expression by PGE2 is mediated via its receptors coupled with the cAMP and PKA pathway and subsequent phosphorylation of the transcription factors cAMP-response element binding protein (CREB) and signal transducer and activator of transcription 3 (STAT3). Although prostaglandin E receptor (EP)-2 and EP4 for PGE2 are coupled with activation of the cAMP and PKA pathway, the exact roles of these 2 receptors in the regulation of COX-2 expression in amnion fibroblasts remain to be determined. Here, we clarify this issue by employing human amnion tissue and fibroblasts with the long-term objective of specific targeting of prostaglandin synthesis in prevention of preterm birth. We find that an EP2 agonist caused long-lasting increases in CREB phosphorylation and COX-2 expression, whereas an EP4 agonist induced only transient increases in CREB phosphorylation and COX-2 expression in amnion fibroblasts. Moreover, only EP2 stimulation increased STAT3 phosphorylation, whereas only EP4 stimulation increased PI3K activity. EP4 antagonist or inhibition of PI3K enhanced the induction of CREB and STAT3 phosphorylation and COX-2 expression by PGE2 or EP2 stimulation, which was attenuated by EP4 overexpression. Of interest, PGE2 and cortisol, both well-demonstrated stimulants of COX-2 expression in amnion fibroblasts, increased EP2 but decreased EP4 receptor expression. Furthermore, increased EP2 but decreased EP4 abundance were observed in amnion tissue at parturition. We conclude that EP2 and EP4 receptors play different roles in the regulation of COX-2 expression in human amnion fibroblasts. EP2 is the dominant PGE2 receptor mediating the induction of COX-2 at parturition, which can be attenuated by simultaneous activation of PI3K coupled to the EP4 receptor.-Lu, J.-W., Wang, W.-S., Zhou, Q., Gan, X.-W., Myatt, L., Sun, K. Activation of prostaglandin EP4 receptor attenuates the induction of cyclooxygenase-2 expression by EP2 receptor activation in human amnion fibroblasts: implications for parturition.


Assuntos
Âmnio/metabolismo , Ciclo-Oxigenase 2/biossíntese , Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Parto , Receptores de Prostaglandina E Subtipo EP2/metabolismo , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Feminino , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Gravidez , Nascimento Prematuro/metabolismo , Fator de Transcrição STAT3/metabolismo
12.
FASEB J ; 33(10): 11303-11313, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31311314

RESUMO

The insulin resistance (IR) of ovarian granulosa cells from polycystic ovary syndrome (PCOS) aggravates the abnormalities in steroidogenesis and anovulation, and chemerin is an adipokine involved in regulating adipogenesis and glucose homeostasis. The role and underlying mechanism of chemerin in developing IR of the granulosa cells from PCOS remain unclear. Plasma, follicular fluid, and human granulosa-lutein cells (hGLs) were collected from non-PCOS and patients with PCOS with or without IR. The chemerin levels were elevated in both follicular fluid and hGL samples from patients with PCOS with IR, and the hGLs from patients with PCOS with IR showed decreased insulin sensitivity and impaired glucose uptake capacity. Moreover, treatment of chemerin attenuated insulin-stimulated glucose uptake by decreasing phosphorylation of insulin receptor substrate (IRS)1/2 Tyr612, phosphorylation of protein kinase B Ser473, and membrane translocation of glucose transporter type 4 through increasing Ser307 phosphorylation of IRS1 in cultured hGLs. These effects could be abolished by small interfering RNA-mediated knockdown of chemokine-like receptor 1. Furthermore, insulin induced the expression of chemerin in hGLs. Our findings demonstrate a novel role of chemerin in the metabolic dysfunction of PCOS, which suggested that chemerin and its receptor can be further implicated as potential therapeutic targets in the future treatment of PCOS.-Li, X., Zhu, Q., Wang, W., Qi, J., He, Y., Wang, Y., Lu, Y., Wu, H., Ding, Y., Sun, Y. Elevated chemerin induces insulin resistance in human granulosa-lutein cells from polycystic ovary syndrome patients.


Assuntos
Quimiocinas/metabolismo , Células da Granulosa/metabolismo , Resistência à Insulina/fisiologia , Insulina/metabolismo , Células Lúteas/metabolismo , Síndrome do Ovário Policístico/metabolismo , Adipocinas/metabolismo , Adulto , Feminino , Humanos , Proteínas Substratos do Receptor de Insulina/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Ovário/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Quimiocinas/metabolismo
13.
FASEB J ; 33(2): 2770-2781, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30303742

RESUMO

Preterm premature rupture of fetal membranes precedes 30-40% of preterm births. Activation of matrix metalloproteases (MMPs) is the one of the major causes of extracellular matrix (ECM) degradation in membrane rupture. Increased cortisol, regenerated by 11ß-hydroxysteroid dehydrogenase 1 in the amnion at parturition, is known to participate in a number of parturition-pertinent events. However, whether cortisol has a role in the regulation of MMPs in the membranes is not known. Here, we addressed this issue using human amnion tissue, the most tensile layer of the membranes. RNA-sequencing revealed that cortisol induced MMP7 expression dramatically in amnion fibroblasts, which was confirmed by real-time quantitative RT-PCR and Western blotting analysis in cortisol-treated amnion explants and fibroblasts. Measurement of collagen IV α5 chain (COL4A5), a substrate for MMP-7, showed that cortisol reduced its extracellular abundance, which was blocked by an antibody against MMP-7. Moreover, increased MMP-7 but decreased COL4A5 abundance was observed in the amnion tissue following labor-initiated spontaneous rupture of membranes. Mechanistic studies showed that cortisol increased the phosphorylation of c-Jun and the expression of c-Fos, the 2 major components of activated protein 1 (AP-1), respectively. The knocking down of c-Fos or c-Jun significantly attenuated the induction of MMP7 expression by cortisol. Chromatin immunoprecipitation assays showed that cortisol stimulated the enrichment of c-Fos and c-Jun at the AP-1 binding site in the MMP7 promoter. The data suggest that induction of MMP7 by cortisol via AP-1 may be a contributing factor to ECM degradation in membrane rupture at parturition.-Wang, L.-Y., Wang, W.-S., Wang, Y.-W., Lu, J.-W., Lu, Y., Zhang, C.-Y., Li, W.-J., Sun, K., Ying, H. Drastic induction of MMP-7 by cortisol in the human amnion: implications for membrane rupture at parturition.


Assuntos
Âmnio/enzimologia , Ruptura Prematura de Membranas Fetais/patologia , Fibroblastos/enzimologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Hidrocortisona/efeitos adversos , Metaloproteinase 7 da Matriz/metabolismo , Parto , Âmnio/efeitos dos fármacos , Anti-Inflamatórios/efeitos adversos , Células Cultivadas , Ativação Enzimática , Feminino , Ruptura Prematura de Membranas Fetais/induzido quimicamente , Ruptura Prematura de Membranas Fetais/enzimologia , Fibroblastos/efeitos dos fármacos , Humanos , Gravidez
14.
Cell Mol Life Sci ; 76(1): 13-26, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30225585

RESUMO

The fetus is shielded from the adverse effects of excessive maternal glucocorticoids by 11ß-HSD2, an enzyme which is expressed in the syncytial layer of the placental villi and is capable of converting biologically active cortisol into inactive cortisone. Impairment of this placental glucocorticoid barrier is associated with fetal intrauterine growth restriction (IUGR) and development of chronic diseases in later life. Ontogeny studies show that the expression of 11ß-HSD2 is initiated at a very early stage after conception and increases with gestational age but declines around term. The promoter for HSD11B2, the gene encoding 11ß-HSD2, has a highly GC-rich core. However, the pattern of methylation on HSD11B2 may have already been set up in the blastocyst when the trophoblast identity is committed. Instead, hCG-initiated signals appear to be responsible for the upsurge of 11ß-HSD2 expression during trophoblast syncytialization. By activating the cAMP/PKA pathway, hCG not only alters the modification of histones but also increases the expression of Sp1 which activates the transcription of HSD11B2. Adverse conditions such as stress, hypoxia and nutritional restriction can cause IUGR of the fetus. It appears that different causes of IUGR may attenuate HSD11B2 expression differentially in the placenta. While stress and nutritional restriction may reduce HSD11B2 expression by increasing its methylation, hypoxia may decrease HSD11B2 expression via alternative mechanisms rather than by methylation. Herein, we summarize the advances in the study of mechanisms underlying the establishment of the placental glucocorticoid barrier and the attenuation of this barrier by adverse conditions during pregnancy.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 2/metabolismo , Feto/fisiologia , Glucocorticoides/metabolismo , Placenta/fisiologia , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/genética , Animais , Metilação de DNA , Epigênese Genética , Feminino , Retardo do Crescimento Fetal/etiologia , Retardo do Crescimento Fetal/genética , Retardo do Crescimento Fetal/metabolismo , Regulação da Expressão Gênica , Glucocorticoides/genética , Histonas/genética , Histonas/metabolismo , Humanos , Hidrocortisona/genética , Hidrocortisona/metabolismo , Gravidez , Regiões Promotoras Genéticas , Transdução de Sinais
15.
Biochem Biophys Res Commun ; 516(2): 402-407, 2019 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-31217075

RESUMO

Gestational diabetes mellitus (GDM) is often accompanied by the development of hyperinsulinemia as an adaptation to increased insulin demand, but this subsequently causes insulin resistance. Loss of function in pancreatic ß-cells further aggravates the development of GDM. The level of serum platelet-derived growth factor (PDGF) reportedly increases in GDM patients. The present study investigated whether enhanced PDGF signaling directly causes ß-cell dysfunction during gestation. Serum PDGF levels were negatively correlated with ß-cell function in GDM patients. Administration of PDGF-BB disrupted glucose tolerance and ß-cell function without inducing apoptosis in gestational mice but had no similar effect in non-gestational mice. The ß-cell-specific genes encoding insulin synthesis proteins were decreased in the islets of PDGF-BB-treated gestational mice. In vitro experiments using INS1 insulinoma cells showed that PDGF-BB promoted cell proliferation, whereas it downregulated ß-cell-specific genes. Taken together, these findings suggested that PDGF reduces ß-cell function during gestation possibly through ß-cell dedifferentiation.


Assuntos
Diabetes Gestacional/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais , Adulto , Animais , Becaplermina/administração & dosagem , Becaplermina/farmacologia , Desdiferenciação Celular/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Diabetes Gestacional/sangue , Feminino , Teste de Tolerância a Glucose , Proteínas de Homeodomínio/metabolismo , Humanos , Insulina/metabolismo , Secreção de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/efeitos dos fármacos , Insulinoma/patologia , Masculino , Camundongos Endogâmicos C57BL , Gravidez , Transporte Proteico/efeitos dos fármacos , Ratos , Transativadores/metabolismo
16.
Clin Sci (Lond) ; 133(3): 515-530, 2019 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-30683734

RESUMO

The de novo synthesis of serum amyloid A1 (SAA1) is augmented in human fetal membranes at parturition. However, its role in parturition remains largely unknown. Here, we investigated whether SAA1 was involved in the rupture of fetal membranes, a crucial event in parturition accompanied with extensive degradation of collagens. Results showed that SAA1 decreased both intracellular and extracellular COL1A1 and COL1A2 abundance, the two subunits of collagen I, without affecting their mRNA levels in human amnion fibroblasts. These reductions were completely blocked only with inhibition of both matrix metalloproteases (MMPs) and autophagy. Consistently, SAA1 increased MMP-2/9 abundance and the markers for autophagic activation including autophagy related (ATG) 7 (ATG7) and the microtubule-associated protein light chain 3 ß (LC3B) II/I ratio with the formation of LC3 punctas and autophagic vacuoles in the fibroblasts. Moreover, the autophagic degradation of COL1A1/COL1A2 and activation of MMP-2/9 by SAA1 were blocked by inhibitors for the toll-like receptors 2/4 (TLR2/4) or NF-κB. Finally, reciprocal corresponding changes of SAA1 and collagen I were observed in the amnion following spontaneous rupture of membranes (ROM) at parturition. Conclusively, SAA1 may participate in membrane rupture at parturition by degradating collagen I via both autophagic and MMP pathways. These effects of SAA1 appear to be mediated by the TLR2/4 receptors and the NF-κB pathway.


Assuntos
Âmnio/metabolismo , Colágeno Tipo I/metabolismo , Parto/metabolismo , Proteína Amiloide A Sérica/metabolismo , Autofagia , Colágeno Tipo I/genética , Cadeia alfa 1 do Colágeno Tipo I , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Parto/genética , Proteólise , Proteína Amiloide A Sérica/genética , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
17.
J Biol Chem ; 292(18): 7578-7587, 2017 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-28302719

RESUMO

The expression of 11ß-hydroxysteroid dehydrogenase type 2 (11ß-HSD2), which acts as a placental glucocorticoid barrier, is silenced in cytotrophoblasts but substantially up-regulated during syncytialization. However, the repressive mechanism of 11ß-HSD2 expression before syncytialization and how this repression is lifted during syncytialization remain mostly unresolved. Here we found that enhancer of zeste homolog 2 (EZH2) accounts for the silence of 11ß-HSD2 expression via trimethylation of histone H3 lysine 27 at the promoter of the 11ß-HSD2 gene. Further studies revealed that, upon syncytialization, human chorionic gonadotropin reduced the phosphorylation of retinoblastoma protein (pRB) via activation of the cAMP/PKA pathway, which sequesters E2F transcription factor 1 (E2F1), the transcription factor for EZH2 expression. As a result of inactivation of the pRB-E2F1-EZH2 pathway, the repressive marker trimethylation of histone H3 lysine 27 at the 11ß-HSD2 promoter is removed, which leads to the robust expression of 11ß-HSD2 during syncytialization.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 2/biossíntese , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Regulação Enzimológica da Expressão Gênica/fisiologia , Placenta/enzimologia , Proteínas da Gravidez/metabolismo , Proteínas Repressoras/metabolismo , Sistemas do Segundo Mensageiro/fisiologia , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/genética , Adulto , AMP Cíclico/genética , AMP Cíclico/metabolismo , Fator de Transcrição E2F1/genética , Fator de Transcrição E2F1/metabolismo , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Feminino , Histonas/genética , Histonas/metabolismo , Humanos , Metilação , Gravidez , Proteínas da Gravidez/genética , Proteínas Repressoras/genética , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/metabolismo
18.
Hum Reprod ; 33(11): 2096-2106, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30272163

RESUMO

STUDY QUESTION: Is overexpression of lysyl oxidase (LOX), an enzyme responsible for the cross-linking of collagens, a cause of anovulation in polycystic ovary syndrome (PCOS)? SUMMARY ANSWER: LOX overexpression was present in PCOS ovaries, due at least in part to interleukin-1ß (IL-1ß), and inhibition of LOX activity with ß-aminopropionitrile (BAPN) ameliorated polycystic ovary morphology and anovulation. WHAT IS KNOWN ALREADY: Aberrant ovarian extracellular matrix (ECM) remodeling and inflammation may contribute to the development of PCOS. It remains unknown whether proinflammatory IL-1ß is a contributing factor for LOX overexpression in PCOS ovaries and whether inhibition of LOX can improve PCOS conditions. STUDY DESIGN, SIZE, DURATION: LOX and IL-1ß abundance in the granulosa cells and follicular fluid was compared between non-PCOS (n = 30) and PCOS (n = 39) patients. The effect and mechanism of IL-1ß on LOX expression was examined in cultured primary human granulosa cells. The improvements in PCOS conditions by LOX inhibition with BAPN was investigated in a dehydroepiandrosterone (DHEA)-induced PCOS rat model. PARTICIPANTS/MATERIALS, SETTING, METHODS: The abundance of LOX and IL-1ß was measured with quantitative real-time polymerase chain reaction (qRT-PCR), LOX activity assays and enzyme-linked immunosorbent assays (ELISA), respectively. The effect of IL-1ß on LOX expression was examined in the presence or absence of inhibitors for signaling molecules and small interfering RNA-mediated knockdown of the putative transcription factor. Chromatin immunoprecipitation assays were conducted to further identify the responsible transcription factor. The role of LOX in ovulation was investigated in a DHEA-induced PCOS rat model with administration of the LOX inhibitor BAPN. The numbers of retrieved total oocytes and MII oocytes were recorded upon ovarian stimulation. MAIN RESULTS AND THE ROLE OF CHANCE: Increased abundance of LOX (P < 0.05) and IL-1ß (P < 0.05) was observed in the granulosa cells and follicular fluid in PCOS patients. IL-1ß increased LOX expression via activation of ERK1/2 and JNK and subsequent activation of the transcription factor c-Jun. Inhibition of LOX with BAPN ameliorated irregular estrous cyclicity (P < 0.05), polycystic ovary morphology and anovulation (P < 0.05) in PCOS rats, but appeared to be ineffective in the improvement of oocyte quality. LARGE SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: Ovarian tissue-directed specific inhibition of LOX in combination with oocyte quality-improving drugs may be more effective in the treatment of PCOS. WIDER IMPLICATIONS OF THE FINDINGS: Inflammation of the ovary is a contributing factor for the aberrant expression of LOX in the PCOS ovary, and inhibition of LOX together with anti-inflammatory therapy may improve the core features of PCOS. STUDY FUNDING/COMPETING INTEREST(S): This work was supported by National Key R & D Program of China (2017YFC1001403) and Doctorial Innovation Fund of Shanghai Jiao Tong University School of Medicine (BXJ201718). The authors declare no competing financial interests.


Assuntos
Líquido Folicular/metabolismo , Síndrome do Ovário Policístico/metabolismo , Proteína-Lisina 6-Oxidase/metabolismo , Adulto , Animais , Anovulação/etiologia , Anovulação/genética , Anovulação/metabolismo , Western Blotting , Estudos de Casos e Controles , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Feminino , Regulação da Expressão Gênica , Células da Granulosa/metabolismo , Humanos , Interleucina-1beta , Ratos , Reação em Cadeia da Polimerase em Tempo Real
19.
Reproduction ; 156(5): 439-449, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30328340

RESUMO

Polycystic ovary syndrome (PCOS) is a major cause of infertility in women of reproductive age. However, its exact etiology remains unknown. In this study, we sequenced miRNAs in human follicular fluid and identified 16 downregulated and 3 upregulated miRNAs in PCOS group compared with non-PCOS group. Among the differential expressed miRNAs, miR-335-5p was verified lower abundance in PCOS than non-PCOS group using quantitative real-time PCR. Besides, miR-335-5p negatively correlated with antral follicle count, anti-Müllerian hormone and total testosterone. Bioinformatics analysis identified serum/glucocorticoid-regulated kinase family member 3 (SGK3) as a potential target gene of miR-335-5p. SGK3 is involved in protein kinase B-mammalian target of rapamycin kinase (AKT-mTOR) signaling pathway and cell proliferation. Western blotting and cell counting kit-8 assays demonstrated that miR-335-5p mimic reduced, while miR-335-5p inhibitor increased, SGK3 abundance, AKT-mTOR pathway and cell proliferation in human granulosa-like tumor KGN cells. Dual-luciferase reporter assays showed that miR-335-5p binds to the 3' untranslated region of SGK3 mRNA. Furthermore, miR-335-5p was decreased and SGK3 was elevated in human granulosa cells obtained from PCOS patients as compared with non-PCOS controls. These findings suggested that miR-335-5p is involved in granulosa cells proliferation by reducing SGK3 expression, which might provide a molecular target to improve dysfunctional granulosa cells in patients with PCOS.


Assuntos
Proliferação de Células , Células da Granulosa/enzimologia , MicroRNAs/metabolismo , Síndrome do Ovário Policístico/enzimologia , Proteínas Serina-Treonina Quinases/metabolismo , Adulto , Estudos de Casos e Controles , Linhagem Celular , Feminino , Regulação Enzimológica da Expressão Gênica , Células da Granulosa/patologia , Humanos , MicroRNAs/genética , Síndrome do Ovário Policístico/genética , Síndrome do Ovário Policístico/patologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo
20.
Immunol Cell Biol ; 95(10): 943-952, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28878297

RESUMO

Preterm premature rupture of membranes (pPROMs) account for one-third of preterm births, a leading cause of neonatal death. Understanding the mechanism of membrane rupture is thus of clinical significance in the prevention of preterm birth. Parturition at both term and preterm is associated with increased abundance of proinflammatory cytokines in the fetal membranes regardless of the presence of infection, which is believed to induce rupture of membranes through activation of the matrix metalloproteinases. It remains unknown whether there are any alternative mechanisms underpinning proinflammatory cytokine-induced rupture of membranes. Here we showed that there were reciprocal increases in interleukin-1ß (IL-1ß) and decreases in lysyl oxidase (LOX), a collagen crosslinking enzyme, in the human amnion tissue following spontaneous rupture of membrane at term and pPROM. Studies using human amnion tissue explants revealed that IL-1ß inhibited the expression of LOX, which can be reproduced in cultured human amnion fibroblasts. Mechanistic study revealed that IL-1ß inhibited LOX expression through activation of p38 and Erk1/2 mitogen-activated protein kinase pathways, which resulted in the phosphorylation of the nuclear factor kappa light-chain enhancer of activated B (NF-κB) cell subunit p65 as well as GATA binding protein 3 (GATA3). Subsequently, activated NF-κB interacted with GATA3 at the NF-κB binding site of LOX promoter to inhibit its expression. Conclusively, this study has revealed an alternative mechanism that IL-1ß may contribute to the rupture of membranes by attenuating collagen crosslinking through downregulation of LOX expression in amnion fibroblasts.


Assuntos
Âmnio/patologia , Ruptura Prematura de Membranas Fetais/imunologia , Fibroblastos/fisiologia , Fator de Transcrição GATA3/metabolismo , Interleucina-1beta/metabolismo , NF-kappa B/metabolismo , Nascimento Prematuro/imunologia , Proteína-Lisina 6-Oxidase/metabolismo , Células Cultivadas , Colágeno/metabolismo , Feminino , Regulação da Expressão Gênica , Humanos , Gravidez , Regiões Promotoras Genéticas/genética , Proteína-Lisina 6-Oxidase/genética , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA