Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Endocrinology ; 165(2)2023 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-38146648

RESUMO

Progesterone synthesized in the placenta is essential for pregnancy maintenance. CYP11A1 is a key enzyme in progesterone synthesis, and its expression increases greatly during trophoblast syncytialization. However, the underlying mechanism remains elusive. Here, we demonstrated that passive demethylation of CYP11A1 promoter accounted for the upregulation of CYP11A1 expression during syncytialization with the participation of the transcription factor C/EBPα. We found that the methylation rate of a CpG locus in the CYP11A1 promoter was significantly reduced along with decreased DNA methyltransferase 1 (DNMT1) expression and its enrichment at the CYP11A1 promoter during syncytialization. DNMT1 overexpression not only increased the methylation of this CpG locus in the CYP11A1 promoter, but also decreased CYP11A1 expression and progesterone production. In silico analysis disclosed multiple C/EBPα binding sites in both CYP11A1 and DNMT1 promoters. C/EBPα expression and its enrichments at both the DNMT1 and CYP11A1 promoters were significantly increased during syncytialization. Knocking-down C/EBPα expression increased DNMT1 while it decreased CYP11A1 expression during syncytialization. Conclusively, C/EBPα plays a dual role in the regulation of CYP11A1 during syncytialization. C/EBPα not only drives CYP11A1 expression directly, but also indirectly through downregulation of DNMT1, which leads to decreased methylation in the CpG locus of the CYP11A1 promoter, resulting in increased progesterone production during syncytialization.


Assuntos
Proteína alfa Estimuladora de Ligação a CCAAT , Enzima de Clivagem da Cadeia Lateral do Colesterol , DNA (Citosina-5-)-Metiltransferase 1 , Placenta , Feminino , Humanos , Gravidez , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Enzima de Clivagem da Cadeia Lateral do Colesterol/genética , Enzima de Clivagem da Cadeia Lateral do Colesterol/metabolismo , Metilação de DNA , Placenta/metabolismo , Progesterona/metabolismo , Trofoblastos/metabolismo , DNA (Citosina-5-)-Metiltransferase 1/genética , DNA (Citosina-5-)-Metiltransferase 1/metabolismo
2.
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
3.
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
4.
Endocrinology ; 163(11)2022 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-36048433

RESUMO

Fetal membrane activation is seen as being one of the crucial triggering components of human parturition. Increased prostaglandin E2 (PGE2) production, a common mediator of labor onset in virtually all species, is recognized as one of the landmark events of membrane activation. Fetal membranes are also equipped with a high capacity of cortisol regeneration by 11ß-hydroxysteroid dehydrogenase 1 (11ß-HSD1), and the cortisol regenerated potently induces PGE2 synthesis, an effect normally suppressed by progesterone during gestation. There is no precipitous decline of progesterone synthesis in human parturition. It is intriguing how this suppression is lifted in parturition. Here, we investigated this issue by using human amnion tissue and primary amnion fibroblasts which synthesize the most PGE2 in the fetal membranes. Results showed that the expression of 11ß-HSD1 and aldo-keto reductase family 1 member C1 (AKR1C1), a progesterone-inactivating enzyme, increased in parallel in human amnion tissue with gestational age toward the end of gestation and at parturition. Cortisol induced AKR1C1 expression via the transcription factor CCAAT enhancer binding protein δ (C/EBPδ) in amnion fibroblasts. Inhibition of AKR1C1 not only blocked progesterone catabolism induced by cortisol, but also enhanced the suppression of cortisol-induced cyclooxygenase-2 (COX-2) expression by progesterone in amnion fibroblasts. In conclusion, our results indicate that cortisol regenerated in the fetal membranes triggers local progesterone withdrawal through enhancement of AKR1C1-mediated progesterone catabolism in amnion fibroblasts, so that the suppression of progesterone on the induction of COX-2 expression and PGE2 synthesis by cortisol can be lifted for parturition.


Assuntos
Âmnio , Hidrocortisona , Feminino , Humanos , Gravidez , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/metabolismo , Aldo-Ceto Redutases/metabolismo , Âmnio/metabolismo , Proteína delta de Ligação ao Facilitador CCAAT/metabolismo , Proteína delta de Ligação ao Facilitador CCAAT/farmacologia , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Fibroblastos/metabolismo , Hidrocortisona/metabolismo , Parto/metabolismo , Progesterona/metabolismo
5.
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
6.
Transl Res ; 230: 55-67, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33129993

RESUMO

Polycystic ovary syndrome (PCOS) is one of the most common endocrine disorders in reproductive-age women. Reduced progesterone levels are associated with luteal phase deficiency in women with PCOS. The levels of C-X-C motif chemokine ligand-14 (CXCL14) were previously reported to be decreased in human-luteinized granulosa (hGL) cells derived from PCOS patients. However, the function of CXCL14 in hGL cells and whether CXCL14 affects the synthesis of progesterone in hGL cells remain unclear. In the present study, the levels of CXCL14 were reduced in follicular fluid and hGL cells in PCOS patients, accompanied by decreased progesterone levels in follicular fluid and decreased steroidogenic acute regulatory (STAR) expression in hGL cells. CXCL14 administration partially reversed the low progesterone production and STAR expression in hGL cells obtained from PCOS patients. In primary hGL cells, CXCL14 upregulated STAR expression and progesterone production. CXCL14 activated the phosphorylation of cyclic adenosine monophosphate response element-binding protein (CREB) and CREB inhibitor attenuated the modulation of StAR expression by CXCL14. P38 and Jun N-terminal kinase (JNK) pathways were also activated by CXCL14 and inhibition of p38 and JNK attenuated the increase of phosphorylation of CREB, STAR expression and progesterone production caused by CXCL14. Our findings revealed the novel role of CXCL14 in upregulation of STAR expression and progesterone synthesis through CREB phosphorylation via activation of p38 and JNK pathways in hGL cells. This is likely contributing to the dysfunction in steroidogenesis in granulosa cells from PCOS patients.


Assuntos
Quimiocinas CXC/farmacologia , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/metabolismo , Fosfoproteínas/metabolismo , Progesterona/biossíntese , Adulto , Antracenos/farmacologia , Células Cultivadas , Enzima de Clivagem da Cadeia Lateral do Colesterol/genética , Enzima de Clivagem da Cadeia Lateral do Colesterol/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Inibidores Enzimáticos/farmacologia , Feminino , Flavonoides/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Humanos , Imidazóis/farmacologia , Fosfoproteínas/genética , Síndrome do Ovário Policístico , Progesterona Redutase/genética , Progesterona Redutase/metabolismo , Piridinas/farmacologia
7.
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
8.
Front Immunol ; 11: 1038, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32582166

RESUMO

Serum amyloid A1 (SAA1) is an acute phase protein produced mainly by the liver to participate in immunomodulation in both sterile and non-sterile inflammation. However, non-hepatic tissues can also synthesize SAA1. It remains to be determined whether SAA1 synthesized locally in the placenta participates in parturition via eliciting inflammatory reactions. In this study, we investigated this issue by using human placenta and a mouse model. We found that SAA1 mRNA and protein were present in human placental villous trophoblasts, which was increased upon syncytialization as well as treatments with lipopolysaccharides (LPS), tumor necrosis factor-α (TNF-α), and cortisol. Moreover, significant increases in SAA1 abundance were observed in the placental tissue or in the maternal blood in spontaneous deliveries without infection at term and in preterm birth with histological chorioamnionitis. Serum amyloid A1 treatment significantly increased parturition-pertinent inflammatory gene expression including interleukin-1ß (IL-1ß), IL-8, TNF-α, and cyclooxygenase-2 (COX-2), along with increased PGF2α production in syncytiotrophoblasts. Mouse study showed that SAA1 was present in the placental junctional zone and yolk sac membrane, which was increased following intraperitoneal administration of LPS. Intraperitoneal injection of SAA1 not only induced preterm birth but also increased the abundance of IL-1ß, TNF-α, and COX-2 in the mouse placenta. Conclusively, SAA1 can be synthesized in the human placenta, which is increased upon trophoblast syncytialization. Parturition is accompanied with increased SAA1 abundance in the placenta. Serum amyloid A1 may participate in parturition in the presence and absence of infection by inducing the expression of inflammatory genes in the placenta.


Assuntos
Parto/metabolismo , Placenta/metabolismo , Proteína Amiloide A Sérica/biossíntese , Adulto , Animais , Corioamnionite/genética , Corioamnionite/imunologia , Corioamnionite/metabolismo , Membranas Extraembrionárias/imunologia , Membranas Extraembrionárias/metabolismo , Feminino , Expressão Gênica , Humanos , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Parto/genética , Parto/imunologia , Placenta/imunologia , Gravidez , Nascimento Prematuro/genética , Nascimento Prematuro/imunologia , Nascimento Prematuro/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteína Amiloide A Sérica/genética , Proteína Amiloide A Sérica/imunologia , Trofoblastos/imunologia , Trofoblastos/metabolismo
9.
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
10.
J Cancer ; 10(20): 4939-4946, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31598166

RESUMO

Objective: The lack of effective therapeutic targets poses a leading challenge to prolong survival and improve the quality of life for pancreatic cancer patients. Proteasome 26S subunit ATPase 2 (PSMC2), a recently discovered gene, has been implicated in certain human carcinogenesis. However, limited data is available concerning the functional significance of PSMC2 in cancer cell growth, and whether it plays a role in pancreatic carcinogenesis remains unknown. Materials and Methods: Quantitative RT-PCR (qRT-PCR) was performed to assess mRNA expression levels of PSMC2 in different pancreatic cancer cell lines. Knockdown of PSMC2 was achieved by using short hairpin RNA (shRNA). The effects of PSMC2 silencing on pancreatic cancer cell proliferation and apoptosis were evaluated by the MTT cell proliferation assay, Celigoassays, Annexin V fluorescence-activated cell sorting (FACS) assay and Caspase-3/7 array. Results: High expression of PSMC2 was detected in three pancreatic cancer cell lines (SW1990, PANC-1, and AsPC-1). Knockdown of PSMC2 in SW1990 cells inhibited proliferation and enhanced apoptosis. Conclusions: Our primary study suggests that PSMC2 might be involved in the progression of pancreatic cancer and may serve as a potential therapeutic target.

11.
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
12.
Pancreas ; 48(7): 904-912, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31268976

RESUMO

OBJECTIVES: Tumor-associated macrophages are dominant players in establishing the inmmunosuppressive microenvironment in pancreatic ductal adenocarcinoma (PDAC). Immune checkpoint inhibitor monotherapy has achieved limited clinical effectiveness. To date, the interaction of macrophages and checkpoint regulators and their correlation with clinicopathologic characteristics in PDAC have been largely unavailable. METHODS: Macrophages and immune checkpoint expression were assessed by immunohistochemistry from 80 PDAC samples. Clinicopathologic features and the prognostic value of each marker were evaluated. In vitro changes in the expression of immune markers in cocultured macrophages and PDAC cells were detected by Western blot and immunosorbance assays. RESULTS: The macrophages marker CD163 and the checkpoint marker programmed death-ligand 1 (PD-L1) remained as the independent prognostic factors for overall survival (hazard ratio, 2.543; P = 0.017 and hazard ratio, 2.389; P = 0.021). Furthermore, integrated analysis of CD163 and PD-L1 served as more optimal indicators of survival (P = 0.000). In vitro coculture of macrophages and PDAC cells significantly increased the expression of CD163 and PD-L1, compared with monocultured counterpart (P < 0.05). CONCLUSIONS: Combined analysis of CD163 and PD-L1 was enhanced indicators of survival in PDAC patients. The interaction of macrophages and immune checkpoints implied the value of the combination therapy.


Assuntos
Antígenos CD/imunologia , Antígenos de Diferenciação Mielomonocítica/imunologia , Antígeno B7-H1/imunologia , Biomarcadores Tumorais/imunologia , Carcinoma Ductal Pancreático/imunologia , Macrófagos/imunologia , Neoplasias Pancreáticas/imunologia , Receptores de Superfície Celular/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Antígeno B7-H1/metabolismo , Biomarcadores Tumorais/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Feminino , Humanos , Estimativa de Kaplan-Meier , Macrófagos/metabolismo , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Prognóstico , Receptores de Superfície Celular/metabolismo , Microambiente Tumoral/imunologia
13.
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
14.
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
15.
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
16.
J Clin Endocrinol Metab ; 103(7): 2457-2467, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29618067

RESUMO

Context: Endometrial insulin resistance (IR) may account for the endometrial dysfunction in polycystic ovary syndrome (PCOS). The underlying mechanism remains to be elucidated. Objective: To investigate whether the abundance of 11ß-hydroxysteroid dehydrogenases (11ß-HSDs) 1 and 2 and cortisol as well as the insulin signaling pathway are altered in PCOS endometrium and to clarify the relationship between endometrial IR and local cortisol. Design: We measured cortisol and cortisone concentrations, 11ß-HSD1 and 11ß-HSD2, and core insulin signaling molecules in endometrial biopsies collected from non-PCOS and PCOS with or without IR patients on the seventh day after human chorionic gonadotropin injection. We also studied the effects of cortisol on glucose uptake and the insulin signaling pathway in primary cultured endometrial epithelial cells (EECs). Results: The cortisol concentration was elevated, whereas 11ß-HSD2 expression was diminished in endometrial biopsies obtained from PCOS with IR patients compared with those from non-PCOS and PCOS without IR patients. The implantation rate was relatively impaired and the endometrial insulin signaling pathway was defective in PCOS with IR patients. In addition, cortisol attenuated insulin-stimulated glucose uptake in EECs, which was mediated by inhibition of Akt phosphorylation and glucose transporter type 4 translocation via induction of phosphatase and tensin homolog deleted on chromosome ten (PTEN). Conclusions: Decreased oxidation of cortisol and defects of insulin signaling in endometrium were observed in PCOS with IR patients. The excessive cortisol level, derived from the reduction of 11ß-HSD2, might contribute to the development of endometrial IR by inhibiting the insulin signaling pathway via induction of PTEN expression in EECs.


Assuntos
Endométrio/metabolismo , Hidrocortisona/metabolismo , Resistência à Insulina/fisiologia , Síndrome do Ovário Policístico/metabolismo , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/metabolismo , Adulto , Gonadotropina Coriônica/administração & dosagem , Cortisona/metabolismo , Implantação do Embrião , Endométrio/citologia , Células Epiteliais , Feminino , Humanos , Insulina/metabolismo , Transdução de Sinais
17.
Sci Signal ; 10(506)2017 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-29162743

RESUMO

Phosphorylation of the transcription factors cyclic adenosine monophosphate response element-binding protein (CREB) and signal transducer and activator of transcription 3 (STAT3) by protein kinase A (PKA) is required for the cortisol-induced production of cyclooxygenase-2 (COX-2) and prostaglandin E2 (PGE2) in human amnion fibroblasts, which critically mediates human parturition (labor). We found that PKA was confined in the nucleus by A-kinase-anchoring protein 95 (AKAP95) in amnion fibroblasts and that this localization was key to the cortisol-induced expression of PTGS2, the gene encoding COX-2. Cortisol increased the abundance of nuclear PKA by stimulating the expression of the gene encoding AKAP95. Knockdown of AKAP95 not only reduced the amounts of nuclear PKA and phosphorylated CREB but also attenuated the induction of PTGS2 expression in primary human amnion fibroblasts treated with cortisol, whereas the phosphorylation of STAT3 in response to cortisol was not affected. The abundances of AKAP95, phosphorylated CREB, and COX-2 were markedly increased in human amnion tissue after labor compared to those in amnion tissues from cesarean sections without labor. These results highlight an essential role for PKA that is anchored in the nucleus by AKAP95 in the phosphorylation of CREB and the consequent induction of COX-2 expression by cortisol in amnion fibroblasts, which may be important in human parturition.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Âmnio/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Ciclo-Oxigenase 2/biossíntese , Hidrocortisona/farmacologia , Proteínas de Ancoragem à Quinase A/fisiologia , Âmnio/citologia , Âmnio/efeitos dos fármacos , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Ciclo-Oxigenase 2/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Parto , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais
18.
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
19.
Endocrinology ; 158(4): 1005-1014, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28323983

RESUMO

Rupture of fetal membranes can initiate parturition at both term and preterm. Collagen is the crucial factor determining the tensile strength of the membranes. Toward the end of gestation, a feed-forward regeneration of cortisol via 11ß-hydroxysteroid dehydrogenase 1 exists in fetal membranes. It remains undetermined whether cortisol contributes to collagen reduction in fetal membranes. In this study, we have examined whether cortisol accumulation is a causative factor for collagen reduction in human amnion fibroblasts, the major source of collagens in the membranes. Cortisol had no effect on collagen 1A1 (COL1A1) and 1A2 (COL1A2) messenger RNA (mRNA) abundance but decreased their protein abundance. The latter effect was affected by neither mRNA transcription inhibitor nor protein translation inhibitor. Mechanistic studies revealed that the reduction in COL1A1 but not COL1A2 protein by cortisol was blocked by lysosome inhibitor chloroquine or small interfering RNA (siRNA)-mediated knockdown of autophagy-related protein 7, an essential protein for autophagy, whereas the proteasome inhibitors MG132 and bortezomib were ineffective. Further analysis showed that cortisol dose dependently increased the ratio of LC3II/LC3I, a marker of lysosome activation, an effect blocked by the glucocorticoid receptor (GR) antagonist RU486 and siRNA-mediated knockdown of GR. Consistently, cortisol decreased COL1A1 and COL1A2 protein abundance in amnion tissue explants, and decreased COL1A1 and COL1A2 protein abundance was observed at parturition in the amnion tissue. Conclusively, cortisol regeneration in fetal membranes may contribute to rupture of fetal membranes at parturition by reducing collagen protein abundance. Lysosome-mediated autophagy accounts for the reduction in COL1A1 by cortisol, but the mechanism underlying the reduction in COL1A2 awaits further investigation.


Assuntos
Âmnio/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Colágeno Tipo I/metabolismo , Fibroblastos/efeitos dos fármacos , Hidrocortisona/farmacologia , Lisossomos/metabolismo , Âmnio/citologia , Âmnio/metabolismo , Autofagia/fisiologia , Bortezomib/farmacologia , Células Cultivadas , Colágeno Tipo I/genética , Cadeia alfa 1 do Colágeno Tipo I , Inibidores de Cisteína Proteinase/farmacologia , Relação Dose-Resposta a Droga , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Leupeptinas/farmacologia , RNA Interferente Pequeno , Receptores de Glucocorticoides/metabolismo
20.
Endocrinology ; 157(11): 4487-4498, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27690691

RESUMO

Rupture of fetal membranes (ROM) can initiate parturition at both term and preterm birth. Apoptosis of the amnion epithelium plays a key role in structural remodeling of the membranes preceding ROM. However, the causative factors for apoptosis remain unidentified. Toward the end of gestation, a feed-forward regeneration of cortisol via 11ß-hydroxysteroid dehydrogenase 1 exists in the fetal membranes. Here, we have examined whether cortisol accumulation is a causative factor for amnion cells apoptosis. By using primary human amnion epithelial and fibroblast cells, we demonstrated cortisol induced apoptosis specifically in epithelial cells but not in fibroblasts via reciprocal regulation of tissue-type plasminogen activator (tPA)/plasmin system. Cortisol increased PLAT expression, the gene encoding tPA, via glucocorticoid receptor binding to a glucocorticoid response element in PLAT promoter, thereby increasing plasmin activity in epithelial cells. Further study revealed that a Fas-mediated extrinsic apoptotic pathway was involved in the induction of epithelial cells apoptosis by cortisol, which was blocked by inhibiting either tPA or plasmin. Consistently, cortisol increased cleaved-caspase-3 and tPA abundance in amnion tissue explants. Moreover, the abundance of cortisol, cleaved-caspase-3, and tPA was significantly increased in amnion tissue after labor-initiated spontaneous rupture of membranes. In conclusion, local accumulation of cortisol is a causative factor for amnion epithelial apoptosis via activation of tPA/plasmin system toward the end of gestation. This may contribute to the ROM at both term and preterm birth.


Assuntos
Âmnio/citologia , Células Epiteliais/citologia , Células Epiteliais/metabolismo , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/metabolismo , Âmnio/metabolismo , Apoptose/genética , Apoptose/fisiologia , Western Blotting , Caspase 3/metabolismo , Células Cultivadas , Imunoprecipitação da Cromatina , Membranas Extraembrionárias/metabolismo , Feminino , Fibrinolisina/genética , Fibrinolisina/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Hidrocortisona/metabolismo , Marcação In Situ das Extremidades Cortadas , Técnicas In Vitro , Gravidez , Regiões Promotoras Genéticas/genética , Ligação Proteica , RNA Interferente Pequeno , Ativador de Plasminogênio Tecidual/genética , Ativador de Plasminogênio Tecidual/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA