Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
FASEB J ; 34(3): 3820-3837, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31944402

RESUMO

Malic enzyme 1 (Me1), a member of the malic enzymes involving in glycolytic pathway and citric acid cycle, is essential for the energy metabolism and maintenance of intracellular redox balance state, but its physiological role and regulatory mechanism in the uterine decidualization are still unknown. Current study showed that Me1 was strongly expressed in decidual cells, and could promote the proliferation and differentiation of stromal cells followed by an accelerated cell cycle transition, indicating an importance of Me1 in the uterine decidualization. Silencing of Me1 attenuated NADPH generation and reduced GR activity, while addition of NADPH improved the defect of GR activity elicited by Me1 depletion. Further analysis found that Me1 modulated intracellular GSH content via GR. Meanwhile, Me1 played a role in maintaining mitochondrial function as indicated by these observations that blockadge of Me1 led to the accumulation of mitochondrial O2- level and decreased ATP production and mtDNA copy numbers accompanied with defective mitochondrial membrane potential. In uterine stromal cells, progesterone induced Me1 expression through PR-cAMP-PKA pathway. Knockdown of HB-EGF might impede the regulation of progesterone and cAMP on Me1. Collectively, Me1 is essential for uterine decidualization in response to progesterone/cAMP/PKA/HB-EGF pathway and plays an important role in preventing mitochondrial dysfunction.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/metabolismo , Malato Desidrogenase/metabolismo , Progesterona/metabolismo , Útero/metabolismo , Trifosfato de Adenosina , Fosfatase Alcalina/metabolismo , Animais , Western Blotting , Ciclo Celular/genética , Ciclo Celular/fisiologia , Proliferação de Células/genética , Proliferação de Células/fisiologia , Feminino , Imunofluorescência , Glutationa/metabolismo , Glutationa Redutase/metabolismo , Hibridização In Situ , Malato Desidrogenase/genética , Potencial da Membrana Mitocondrial , Camundongos , Gravidez , Interferência de RNA , Reação em Cadeia da Polimerase em Tempo Real , Células Estromais/metabolismo
2.
J Cell Mol Med ; 24(12): 7023-7033, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32391984

RESUMO

Serpinb6b is a novel member of Serpinb family and found in germ and somatic cells of mouse gonads, but its physiological function in uterine decidualization remains unclear. The present study revealed that abundant Serpinb6b was noted in decidual cells, and advanced the proliferation and differentiation of stromal cells, indicating a creative role of Serpinb6b in uterine decidualization. Further analysis found that Serpinb6b modulated the expression of Mmp2 and Mmp9. Meanwhile, Serpinb6b was identified as a target of Bmp2 regulation in stromal differentiation. Treatment with rBmp2 resulted in an accumulation of intracellular cAMP level whose function in this differentiation program was mediated by Serpinb6b. Addition of PKA inhibitor H89 impeded the Bmp2 induction of Serpinb6b, whereas 8-Br-cAMP rescued the defect of Serpinb6b expression elicited by Bmp2 knock-down. Attenuation of Serpinb6b greatly reduced the induction of constitutive Wnt4 activation on stromal cell differentiation. By contrast, overexpression of Serpinb6b prevented this inhibition of differentiation process by Wnt4 siRNA. Moreover, blockage of Wnt4 abrogated the up-regulation of cAMP on Serpinb6b. Collectively, Serpinb6b mediates uterine decidualization via Mmp2/9 in response to Bmp2/cAMP/PKA/Wnt4 pathway.


Assuntos
Proteína Morfogenética Óssea 2/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Decídua/metabolismo , Serpinas/metabolismo , Transdução de Sinais , Proteína Wnt4/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Feminino , Metaloproteinases da Matriz/metabolismo , Camundongos , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Serpinas/genética , Células Estromais/citologia , Células Estromais/metabolismo
3.
J Cell Physiol ; 235(9): 6023-6031, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-31960430

RESUMO

The desert hedgehog (Dhh) is crucial for spermatogenesis and Leydig cell differentiation, but little is known regarding its physiological function in cartilage. In this study, Dhh mRNA was abundant in antler chondrocytes, where it advanced cell proliferation concomitant with accelerated transition from the G1 to the S phase and induced elevation of the hypertrophic chondrocyte markers, Col X and Runx2. Silencing of Ptch1 resulted in appreciable Smo accumulation and enhanced rDhh stimulation of Smo, whose impediment by cyclopamine obscured the proliferative function of Dhh and alleviated its guidance of chondrocyte differentiation. Further analysis evidenced the noteworthy positive action of Smo in the bridging between Dhh and Gli transcription factors. Obstruction of Gli1 by GANT58 caused the failed stimulation of Col X and Runx2 by rDhh. Analogously, siRNA against Gli1-3 hindered chondrocyte differentiation in the context of rDhh. Simultaneously, Gli transcription factors mediated the regulation of Dhh on Foxa1, Foxa2, and Foxa3, whose knockdown impaired chondrocyte differentiation. Attenuation of Foxa antagonized the augmentation of Col X and Runx2 generated by rDhh. Collectively, Dhh signaling through its target Foxa appears to induce antler chondrocyte proliferation and differentiation.


Assuntos
Chifres de Veado/crescimento & desenvolvimento , Condrogênese/genética , Fatores de Transcrição Forkhead/genética , Espermatogênese/genética , Animais , Chifres de Veado/metabolismo , Cartilagem/crescimento & desenvolvimento , Cartilagem/metabolismo , Ciclo Celular/genética , Diferenciação Celular/genética , Condrócitos/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Cervos/genética , Cervos/crescimento & desenvolvimento , Proteínas Hedgehog/genética , Células Intersticiais do Testículo/citologia , Células Intersticiais do Testículo/patologia , Masculino , Transdução de Sinais
4.
Exp Physiol ; 105(2): 282-292, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31803965

RESUMO

NEW FINDINGS: What is the central question of this study? What are the potential therapeutic roles of ginsenoside Rb1 and hydroxysafflor yellow A (HSYA) in polycystic ovary syndrome (PCOS). What is the main finding and its importance? HSYA restored the oestrous cycles of PCOS mice, reduced follicular cysts in ovaries and rescued abnormal hormone secretion; ginsenoside Rb1 did not ameliorate the main symptoms of PCOS mice. HSYA alleviated oxidative stress along with an enhancement of antioxidant enzyme activity. This highlights a potential role of HSYA in PCOS therapy. ABSTRACT: Polycystic ovary syndrome (PCOS) is the most common endocrine disease resulting in female infertility. Hydroxysafflor yellow A (HSYA) and ginsenoside Rb1 have been shown to have antioxidant properties, but little is known about their impact in PCOS. Here dehydroepiandrosterone was used to induce PCOS in a mouse model that was characterized by an irregular oestrous cycle, cystic follicles and an elevated serum testosterone level. Supplementation of HSYA restored the oestrous cycle of PCOS mice, reduced follicular cysts in PCOS mouse ovaries and brought about a decline in serum testosterone level, while ginsenoside Rb1 did not ameliorate the above symptoms of PCOS mice. After HSYA treatment, there was elevation of serum oestradiol, progesterone, luteinizing hormone and anti-Müllerian hormone levels and a reduction of follicle-stimulating hormone level, but ginsenoside Rb1 only rescued the levels of follicle-stimulating hormone and anti-Müllerian hormone. Further analysis evidenced that HSYA reversed the expression of steroid hormone secretion-related genes Star, Hsd3b1, Cyp11a1 and Cyp19a1. In PCOS mice HSYA weakened the elevation of ovarian malondialdehyde, which is regarded as a biomarker for oxidative stress. Moreover, HSYA improved reduced glutathione content accompanied by a simultaneous increase in reduced to oxidized glutathione ratio, and enhanced the activities of the antioxidant enzymes superoxide dismutase, glutathione peroxidase and catalase. Collectively, HSYA exerted beneficial effects on PCOS mice by restoring hormone secretion and alleviating oxidative stress.


Assuntos
Chalcona/análogos & derivados , Estresse Oxidativo/efeitos dos fármacos , Hormônios Peptídicos/sangue , Pigmentos Biológicos/uso terapêutico , Síndrome do Ovário Policístico/sangue , Síndrome do Ovário Policístico/tratamento farmacológico , Quinonas/uso terapêutico , Animais , Chalcona/farmacologia , Chalcona/uso terapêutico , Feminino , Hormônio Foliculoestimulante/sangue , Hormônio Luteinizante/sangue , Camundongos , Camundongos Endogâmicos ICR , Estresse Oxidativo/fisiologia , Pigmentos Biológicos/farmacologia , Progesterona/sangue , Quinonas/farmacologia , Resultado do Tratamento
5.
Cell Biol Int ; 44(2): 433-445, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31579960

RESUMO

Genistein is an isoflavone that has estrogen (E2 )-like activity and is beneficial for follicular development, but little is known regarding its function in oxidative stress (OS)-mediated granulosa cell (GC) injury. Here, we found that after exposure to H2 O2 , Genistein weakened the elevated levels of intracellular reactive oxygen species (ROS) and malondialdehyde (MDA), which were regarded as the biomarkers for OS, and rescued glutathione (GSH) content and GSH/GSSG ratio accompanying with a simultaneous increase in cyclic adenosine monophosphate (cAMP) level, whereas addition of protein kinase A (PKA) inhibitor H89 impeded the effects of Genistein on the levels of ROS and MDA. Further analysis evidenced that Genistein enhanced the activities of antioxidant enzymes superoxide dismutase (SOD), GSH-peroxidase (GSH-Px), and catalase (CAT) in H2 O2 -treated GCs, but this enhancement was attenuated by H89. Under OS, Genistein improved cell viability and lessened the apoptotic rate of GCs along with a reduction in the activity of Casp3 and levels of Bax and Bad messenger RNA (mRNA), while H89 reversed the above effects. Moreover, Genistein treatment caused an obvious elevation in mitochondrial membrane potential (MMP) followed by a decline in the levels of intracellular mitochondrial superoxide, but H89 inhibited the regulation of Genistein on MMP and mitochondrial superoxide. Supplementation of Genistein promoted the secretion of E2 and increased the expression of Star and Cyp19a1 mRNA, whereas suppressed the level of progesterone (P4 ) accompanied with a decline in the level of Hsd3b1 mRNA expression. H89 blocked the regulation of Genistein on the secretion of E2 and P4 , and alleviated the ascending of Star and Cyp19a1 elicited by Genistein. Collectively, Genistein protects GCs from OS via cAMP-PKA signaling.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Genisteína/farmacologia , Células da Granulosa/efeitos dos fármacos , Ovário/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Animais , Sobrevivência Celular , Feminino , Glutationa/metabolismo , Células da Granulosa/metabolismo , Células da Granulosa/patologia , Potencial da Membrana Mitocondrial , Camundongos , Camundongos Endogâmicos ICR , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Ovário/metabolismo , Ovário/patologia , Fitoestrógenos/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Superóxidos/metabolismo
6.
J Cell Physiol ; 233(2): 1612-1626, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28657144

RESUMO

Ptn is a pleiotropic growth factor involving in the regulation of cellular proliferation and differentiation, but its biological function in uterine decidualization remains unknown. Here, we showed that Ptn was highly expressed in the decidual cells, and could induce the proliferation of uterine stromal cells and expression of Prl8a2 and Prl3c1 which were two well-established differentiation markers for decidualization, suggesting an important role of Ptn in decidualization. In the uterine stromal cells, progesterone stimulated the expression of Ptn accompanied with an accumulation of intracellular cAMP level. Silencing of Ptn impeded the induction of progesterone and cAMP on the differentiation of uterine stromal cells. Administration of PKA inhibitor H89 resulted in a blockage of progesterone on Ptn expression. Further analysis evidenced that regulation of progesterone and cAMP on Ptn was mediated by C/EBPß. During in vitro decidualization, knockdown of Ptn could weaken the up-regulation of Prl8a2 and Prl3c1 elicited by C/EBPß overexpression, while constitutive activation of Ptn reversed the repressive effects of C/EBPß siRNA on the expression of Prl8a2 and Prl3c1. Meanwhile, Ptn might mediate the regulation of C/EBPß on Hand2 which was a downstream target of Ptn in the differentiation of uterine stromal cells. Attenuation of Ptn or C/EBPß by specific siRNA blocked the stimulation of Hand2 by progesterone and cAMP. Collectively, Ptn may play a vital role in the progesterone-induced decidualization pathway.


Assuntos
8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Proteínas de Transporte/metabolismo , Diferenciação Celular/efeitos dos fármacos , Citocinas/metabolismo , Decídua/efeitos dos fármacos , Progesterona/farmacologia , Células Estromais/efeitos dos fármacos , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Proteína beta Intensificadora de Ligação a CCAAT/genética , Proteínas de Transporte/genética , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Citocinas/genética , Decídua/citologia , Decídua/metabolismo , Feminino , Regulação da Expressão Gênica , Camundongos , Gravidez , Prolactina/análogos & derivados , Prolactina/genética , Prolactina/metabolismo , Interferência de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células Estromais/metabolismo , Fatores de Tempo , Transfecção
7.
J Cell Physiol ; 233(2): 1129-1145, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28436029

RESUMO

Although ATRA is involved in regulating the proliferation and differentiation of chondrocytes, its underlying mechanism remains unknown. Here we showed that ATRA could stimulate the proliferation of antler chondrocytes and expression of COL X and MMP13 which were two well-known markers for hypertrophic chondrocytes. Silencing of CRABP2 prevented the induction of ATRA on chondrocyte terminal differentiation, while overexpression of CRABP2 exhibited the opposite effects. CYP26A1 and CYP26B1 weakened the sensitivity of antler chondrocytes to ATRA. Further analysis evidenced that ATRA might induce chondrocyte terminal differentiation and modulate the expression of BMP2, WNT4, and RUNX1 through RARα/RXRα. Knockdown of BMP2 enhanced the induction of ATRA on the expression of COL X and MMP13, whereas overexpression of BMP2 abrogated this effectiveness. WNT4 might mediate the effects of ATRA and BMP2 on chondrocyte terminal differentiation. Dysregulation of BMP2 impaired the regulation of ATRA on WNT4 expression. Administration of ATRA to antler chondrocytes transfected with RUNX1 siRNA failed to induce the differentiation. Conversely, rRUNX1 strengthened the stimulation of ATRA on the expression of COL X and MMP13. Simultaneously, RUNX1 was a downstream effector of BMP2 and WNT4 in chondrocyte terminal differentiation. Moreover, WNT4 might play an important role in the crosstalk between BMP2 and RUNX1. Attenuation of BMP2 or WNT4 enhanced the interaction between ATRA and RUNX1, while constitutive expression of BMP2 or WNT4 reversed the regulation of ATRA on RUNX1. Collectively, WNT4 may act downstream of BMP2 to mediate the effects of ATRA on the terminal differentiation of antler chondrocytes through targeting RUNX1.


Assuntos
Chifres de Veado/efeitos dos fármacos , Proteína Morfogenética Óssea 2/metabolismo , Diferenciação Celular/efeitos dos fármacos , Condrócitos/efeitos dos fármacos , Condrogênese/efeitos dos fármacos , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Tretinoína/farmacologia , Via de Sinalização Wnt/efeitos dos fármacos , Proteína Wnt4/metabolismo , Animais , Chifres de Veado/citologia , Chifres de Veado/metabolismo , Proteína Morfogenética Óssea 2/genética , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Condrócitos/metabolismo , Colágeno Tipo X/genética , Colágeno Tipo X/metabolismo , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Cervos , Regulação da Expressão Gênica , Metaloproteinase 13 da Matriz/genética , Metaloproteinase 13 da Matriz/metabolismo , Interferência de RNA , Receptores do Ácido Retinoico/agonistas , Receptores do Ácido Retinoico/genética , Receptores do Ácido Retinoico/metabolismo , Ácido Retinoico 4 Hidroxilase/genética , Ácido Retinoico 4 Hidroxilase/metabolismo , Fatores de Tempo , Transfecção , Proteína Wnt4/genética
8.
Cell Physiol Biochem ; 48(6): 2399-2408, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30121651

RESUMO

BACKGROUND/AIMS: High mobility group box 1 (Hmgb1) is associated with a variety of physiological processes including embryonic development, cell proliferation and differentiation, but little information is available regarding its biological role in decidualization. METHODS: In situ hybridization, real-time PCR, RNA interference, gene overexpression and MTS assay were used to analyze the spatiotemporal expression of Hmgb1 in mouse uterus during the pre-implantation period, and explore its function and regulatory mechanisms during uterine decidualization. RESULTS: Hmgb1 mRNA was obviously observed in uterine epithelium on day 2 and 3 of pregnancy, but its expression was scarcely detected on day 4 of pregnancy. With the onset of embryo implantation, abundant Hmgb1 expression was noted in the subluminal stromal cells around the implanting blastocyst at implantation sites. Meanwhile, the accumulation of Hmgb1 mRNA was visualized in the decidual cells. Hmgb1 advanced the proliferation of uterine stromal cells and induced the expression of prolactin family 8, subfamily a, member 2 (Prl8a2), a reliable differentiation marker for decidualization. In uterine stromal cells, cAMP analogue 8-Br-cAMP up-regulated the expression of Hmgb1, but the up-regulation was abrogated by protein kinase A (PKA) inhibitor H89. Silencing of Hmgb1 by specific siRNA impeded the induction of 8-Br-cAMP on Prl8a2. Further analysis evidenced that Hmgb1 was a critical mediator of Kruppel-like factor 5 (Klf5) function in stromal differentiation. Knockdown of bone morphogenetic protein 2 (Bmp2) prevented the up-regulation of Prl8a2 elicited by Hmgb1 overexpression, whereas addition of exogenous recombinant Bmp2 protein (rBmp2) reversed the repression of Hmgb1 siRNA on Prl8a2 expression. CONCLUSION: Hmgb1 may play an important role during mouse uterine decidualization.


Assuntos
Proteína Morfogenética Óssea 2/metabolismo , Diferenciação Celular/efeitos dos fármacos , Proteína HMGB1/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Prolactina/metabolismo , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Animais , Proteína Morfogenética Óssea 2/antagonistas & inibidores , Proteína Morfogenética Óssea 2/genética , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Implantação do Embrião , Feminino , Proteína HMGB1/antagonistas & inibidores , Proteína HMGB1/genética , Isoquinolinas/farmacologia , Fatores de Transcrição Kruppel-Like/genética , Camundongos , Gravidez , Prolactina/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/farmacologia , Células Estromais/citologia , Células Estromais/metabolismo , Sulfonamidas/farmacologia , Regulação para Cima/efeitos dos fármacos , Útero/citologia
9.
J Exp Zool B Mol Dev Evol ; 330(4): 215-224, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29781132

RESUMO

Although Egr2 is involved in regulating the folliculogenesis and ovulation, there is almost no data describing its physiological function in embryo implantation and decidualization. Here, we showed that Egr2 mRNA was distinctly accumulated in subluminal stromal cells around implanting blastocyst on day 5 of pregnancy as well as in estrogen-activated implantation uterus. Estrogen induced the expression of Egr2 in uterine epithelia. Elevated expression of Egr2 mRNA was also observed in the decidual cells. Silencing of Egr2 by specific siRNA weakened the proliferation of uterine stromal cells and reduced the expression of Ccnd1, Ccnd3, Cdk4, and Cdk6. Furthermore, Egr2 advanced the expression of Prl8a2, Prl3c1, and Pgr, the well-established differentiation markers for decidualization. Administration of exogenous recombinant heparin-binding EGF-like growth factor (rHB-EGF) to uterine stromal cells resulted in an increase in the level of Egr2 mRNA. Moreover, siRNA-mediated attenuation of Egr2 impeded the stimulation of HB-EGF on stromal cell differentiation. Knockdown of Egr2 led to a reduction in the expression of Cox-2, mPGES-1, Vegf, Trp53, and Mmp2. Further analysis found that Egr2 may serve as an intermediate to mediate the regulation of HB-EGF on Cox-2, mPGES-1, Vegf, Trp53, Mmp2, and Ccnd3. Collectively, Egr2 may play an important role during embryo implantation and decidualization.


Assuntos
Proteína 2 de Resposta de Crescimento Precoce/metabolismo , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/farmacologia , Células Estromais/efeitos dos fármacos , Animais , Diferenciação Celular , Proliferação de Células , Proteína 2 de Resposta de Crescimento Precoce/genética , Implantação do Embrião/genética , Feminino , Perfilação da Expressão Gênica , Masculino , Camundongos , Gravidez , RNA Mensageiro , RNA Interferente Pequeno , Útero/metabolismo
10.
Cell Physiol Biochem ; 44(5): 1681-1695, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29216626

RESUMO

BACKGROUND/AIMS: Hmgn2 is involved in regulating embryonic development, but its physiological function during embryo implantation and decidualization remains unknown. METHODS: In situ hybridization, real-time PCR, RNA interference, gene overexpression and MTS assay were used to examine the expression of Hmgn2 in mouse uterus during the pre-implantation period and explore its function and regulatory mechanisms in epithelial adhesion junction and stromal cell proliferation and differentiation. RESULTS: Hmgn2 was primarily accumulated in uterine luminal epithelia on day 4 of pregnancy and subluminal stromal cells around the implanting blastocyst at implantation sites on day 5. Similar results were observed during delayed implantation and activation. Meanwhile, Hmgn2 expression was visualized in the decidua. In uterine epithelial cells, silencing of Hmgn2 by specific siRNA reduced the expression of adhesion molecules Cdh1, Cdh2 and Ctnnb1 and enhanced the expression of Muc1, whereas constitutive activation of Hmgn2 exhibited the opposite effects, suggesting a role for Hmgn2 in attachment reaction during embryo implantation. Estrogen stimulated the expression of Hmgn2 in uterine epithelia, but the stimulation was abrogated by ER antagonist ICI 182,780. Further analysis evidenced that attenuation of Hmgn2 might eliminate the regulation of estrogen on the expression of Cdh1, Cdh2 and Ctnnb1. In uterine stromal cells, progesterone induced the accumulation of Hmgn2 which advanced the expression of Prl8a2 and Prl3c1, two well-known differentiation markers for decidualization, but did not affect the proliferation of stromal cells. Knockdown of Hmgn2 blocked the progesterone-induced differentiation of uterine stromal cells. Moreover, Hmgn2 might serve as an intermediate to mediate the regulation of progesterone on Hand2. CONCLUSION: Hmgn2 may play an important role during embryo implantation and decidualization.


Assuntos
Decídua/metabolismo , Implantação do Embrião , Proteína HMGN2/metabolismo , Animais , Caderinas/metabolismo , Proteínas Cdh1/metabolismo , Diferenciação Celular/efeitos dos fármacos , Estradiol/análogos & derivados , Estradiol/farmacologia , Estrogênios/farmacologia , Feminino , Fulvestranto , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Proteína HMGN2/antagonistas & inibidores , Proteína HMGN2/genética , Camundongos , Mucina-1/metabolismo , Gravidez , Progesterona/farmacologia , Prolactina/metabolismo , Interferência de RNA , Células Estromais/citologia , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo , Útero/metabolismo , beta Catenina/metabolismo
11.
J Exp Zool B Mol Dev Evol ; 328(6): 575-586, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28643469

RESUMO

Although all-trans retinoic acid (ATRA) is involved in the regulation of cartilage growth and development, its regulatory mechanisms remain unknown. Here, we showed that ATRA could induce the expression of COL9A1 in antler chondrocytes. Silencing of cellular retinoic acid binding protein 2 (CRABP2) could impede the ATRA-induced upregulation of COL9A1, whereas overexpression of CRABP2 presented the opposite effect. RARα agonist Am80 induced the expression of COL9A1, whereas treatment with RARα antagonist Ro 41-5253 or RXRα small-interfering RNA (siRNA) caused an obvious blockage of ATRA on COL9A1. In antler chondrocytes, CYP26A1 and CYP26B1 weakened the sensitivity of ATRA to COL9A1. Simultaneously, Bone morphogenetic protein 2 (BMP2) and WNT4 mediated the regulation of ATRA on COL9A1 expression. Knockdown of WNT4 could abrogate the inhibitory effect of BMP2 overexpression on COL9A1. Conversely, constitutive expression of WNT4 reversed the upregulation of COL9A1 elicited by BMP2 siRNA. Together these data indicated that WNT4 might act downstream of BMP2 to mediate the effect of ATRA on COL9A1 expression. Further analysis evidenced that attenuation of runt-related transcription factor 1 (RUNX1) could prevent the stimulation of ATRA on COL9A1 expression, while exogenous rRUNX1 further enhanced this effectiveness. Moreover, RUNX1 might serve as an intermediate to mediate the regulation of BMP2 and WNT4 on COL9A1 expression. Collectively, ATRA signaling might regulate the expression of COL9A1 through BMP2-WNT4-RUNX1 pathway.


Assuntos
Chifres de Veado/citologia , Proteína Morfogenética Óssea 2/metabolismo , Colágeno Tipo IX/metabolismo , Regulação da Expressão Gênica/fisiologia , Transdução de Sinais/fisiologia , Tretinoína/metabolismo , Animais , Proteína Morfogenética Óssea 2/genética , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Colágeno Tipo IX/genética , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Proteína Wnt4/genética , Proteína Wnt4/metabolismo
12.
Cell Biol Int ; 41(3): 296-308, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28067449

RESUMO

Although 13cRA is involved in the regulation of cellular proliferation and differentiation, its physiological roles in chondrocyte proliferation and differentiation still remain unknown. Here, we showed that 13cRA could induce the proliferation of sika deer antler chondrocytes and expression of Ccnd3 and Cdk6. Administration of 13cRA to antler chondrocytes resulted in an obvious increase in the expression of chondrocyte marker Col II and hypertrophic chondrocyte marker Col X. Silencing of Crabp2 expression by specific siRNA could prevent the 13cRA-induced up-regulation of Col X, whereas overexpression of Crabp2 showed the opposite effects. Further study found that Crabp2 mediated the regulation of 13cRA on the expression of Runx3 which was highly expressed in the antler cartilage and inhibited the differentiation of antler chondrocytes. Moreover, attenuation of Runx3 expression greatly raised 13cRA-induced chondrocyte differentiation. Simultaneously, 13cRA could stimulate the expression of Cyp26a1 and Cyp26b1 in the antler chondrocytes. Inhibition of Cyp26a1 and/or Cyp26b1 reinforced the effects of 13cRA on the expression of Col X and Runx3, while overexpression of Cyp26b1 rendered the antler chondrocytes hyposensitive to 13cRA. Collectively, 13cRA may play an important role in the differentiation of antler chondrocytes through targeting Runx3. Crabp2 enhances the effects of 13cRA on chondrocyte differentiation, while Cyp26a1 and Cyp26b1 weaken the sensitivity of antler chondrocytes to 13cRA.


Assuntos
Diferenciação Celular/fisiologia , Condrócitos/fisiologia , Condrogênese/fisiologia , Subunidade alfa 3 de Fator de Ligação ao Core/biossíntese , Isotretinoína/farmacologia , Animais , Chifres de Veado , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Células Cultivadas , Condrócitos/efeitos dos fármacos , Condrogênese/efeitos dos fármacos , Subunidade alfa 3 de Fator de Ligação ao Core/antagonistas & inibidores , Cervos , Sistemas de Liberação de Medicamentos/métodos , Isotretinoína/metabolismo
13.
Cell Tissue Res ; 366(2): 393-401, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27147263

RESUMO

Although Runx2 is involved in the regulation of cellular differentiation, its physiological roles in the differentiation of uterine stromal cells during decidualization still remain unknown. The aim of this study was to examine the expression, regulation and function of Runx2 in mouse uterus during decidualization. The results showed that Runx2 was highly expressed in the decidua and oil-induced decidualized cells. In the uterine stromal cells, recombinant human Runx2 (rRunx2) could induce the expression of Prl8a2 and Prl3c1 which are two well-known differentiation markers for decidualization, while inhibition of Runx2 with specific siRNA reduced their expression. Further study found that rRunx2 could improve the expression of Prl8a2 and Prl3c1 in the C/EBPß siRNA-transfected stromal cells. In the stromal cells, cAMP analogue 8-Br-cAMP could induce the expression of Runx2. Moreover, the induction was blocked by PKA inhibitor H89. Simultaneously, attenuation of C/EBPß with siRNA could also reduce the cAMP-induced Runx2 expression. Furthermore, siRNA-mediated silencing of Runx2 expression alleviated the effects of cAMP on the differentiation of stromal cells. Runx2 might act downstream of C/EBPß to regulate the expression of Cox-2, Vegf and Mmp9 in the uterine stromal cells. Collectively, Runx2 may play an important role during mouse decidualization.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Diferenciação Celular , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Útero/citologia , Animais , Subunidade alfa 1 de Fator de Ligação ao Core/genética , AMP Cíclico/metabolismo , Ciclo-Oxigenase 2/metabolismo , Decídua/citologia , Feminino , Hibridização In Situ , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Gravidez , Células Estromais/citologia , Células Estromais/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
14.
Cell Tissue Res ; 362(1): 231-40, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25896884

RESUMO

Runx1 transcription factor is a key developmental regulator. However, little is known about the effects of Runx1 on embryo implantation and decidualization. The aim of this study is to examine the expression and regulation of Runx1 in mouse uterus during the peri-implantation period. There was no evident Runx1 mRNA signal on days 1-4 of pregnancy. On day 5 of pregnancy, Runx1 mRNA was mainly localized in the subluminal stroma surrounding the implanting blastocyst. A similar result was observed in the estrogen-activated implantation uterus. Simultaneously, a high level of Runx1 mRNA expression was detected on days 6-8 of pregnancy and under artificial decidualization. 8-Br-cAMP could induce the expression of Runx1 mRNA in the uterine stromal cells. Moreover, the induction was obviously blocked by PKA inhibitor H89. Inhibition of Runx1 with specific siRNA could decrease the proliferation of stromal cells and expression of decidual markers Prl8a2 and Prl3c1 in the uterine stromal cells. Further study found that inhibition of Runx1 could also suppress the expression of Cox-2, mPGES-1 and Mmp2 genes in uterine stromal cells. Estrogen and progesterone could induce the expression of Runx1 mRNA in ovariectomized mouse uterus and uterine stromal cells. Taken together, these data suggest that Runx1 may play an important role during mouse decidualization.


Assuntos
Blastocisto/fisiologia , Subunidade alfa 2 de Fator de Ligação ao Core/biossíntese , Implantação do Embrião/fisiologia , Células Estromais/metabolismo , Útero/metabolismo , Animais , Proliferação de Células , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Feminino , Camundongos , Transfecção
15.
Int J Mol Sci ; 16(2): 4265-80, 2015 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-25690038

RESUMO

ß-hydroxybutyric acid (BHBA) regulates the synthesis and secretion of growth hormone (GH) and prolactin (PRL), but its mechanism is unknown. In this study, we detected the effects of BHBA on the activities of G protein signaling pathways, AMPK-α activity, GH, and PRL gene transcription, and GH and PRL secretion in dairy cow anterior pituitary cells (DCAPCs). The results showed that BHBA decreased intracellular cAMP levels and a subsequent reduction in protein kinase A (PKA) activity. Inhibition of PKA activity reduced cAMP response element-binding protein (CREB) phosphorylation, thereby inhibiting GH and PRL transcription and secretion. The effects of BHBA were attenuated by a specific Gαi inhibitor, pertussis toxin (PTX). In addition, intracellular BHBA uptake mediated by monocarboxylate transporter 1 (MCT1) could trigger AMPK signaling and result in the decrease in GH and PRL mRNA translation in DCAPCs cultured under low-glucose and non-glucose condition when compared with the high-glucose group. This study identifies a biochemical mechanism for the regulatory action of BHBA on GH and PRL gene transcription, translation, and secretion in DCAPCs, which may be one of the factors that regulate pituitary function during the transition period in dairy cows.


Assuntos
Hormônio do Crescimento/metabolismo , Hidroxibutiratos/toxicidade , Adeno-Hipófise/efeitos dos fármacos , Prolactina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Bovinos , Células Cultivadas , AMP Cíclico/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Feminino , Glucose/farmacologia , Hormônio do Crescimento/antagonistas & inibidores , Hormônio do Crescimento/genética , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Toxina Pertussis/farmacologia , Fosforilação/efeitos dos fármacos , Adeno-Hipófise/citologia , Adeno-Hipófise/metabolismo , Prolactina/antagonistas & inibidores , RNA Mensageiro/metabolismo , Simportadores/genética , Simportadores/metabolismo , Fator de Transcrição Pit-1/genética , Fator de Transcrição Pit-1/metabolismo , Transcrição Gênica/efeitos dos fármacos
16.
Microbiol Immunol ; 58(7): 398-408, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24845706

RESUMO

Post-weaning multisystemic wasting syndrome (PMWS) associated with porcine circovirus type 2 (PCV2) has caused the swine industry significant health challenges and economic damage. Although inactivated and subunit vaccines against PMWS have been used widely, so far no DNA vaccine is available. In this study, with the aim of exploring a new route for developing a vaccine against PCV2, the immunogenicity of a DNA vaccine was evaluated in mice. The pEGFP-N1 vector was used to construct a PCV2 Cap gene recombinant vaccine. To assess the immunogenicity of pEGFP-Cap, 80 BALB/c mice were immunized three times at 2 weekly intervals with pEGFP-Cap, LG-strain vaccine, pEGFP-N1 vector or PBS and then challenged with PCV2. IgG and cytokines were assessed by indirect ELISA and ELISA, respectively. Specimens stained with hematoxylin and eosin (HE) and immunohistochemistry (IHC) techniques were examined histopathologically. It was found that vaccination of the mice with the pEGFP-Cap induced solid protection against PCV2 infection through induction of highly specific serum IgG antibodies and cytokines (IFN-γ and IL-10), and a small PCV2 viral load. The mice treated with the pEGFP-Cap and LG-strain developed no histopathologically detectable lesions (HE stain) and IHC techniques revealed only a few positive cells. Thus, this study demonstrated that recombinant pEGFP-Cap substantially alleviates PCV2 infection in mice and provides evidence that a DNA vaccine could be an alternative to PCV2 vaccines against PMWS.


Assuntos
Infecções por Circoviridae/prevenção & controle , Circovirus/genética , Circovirus/imunologia , Vacinas de DNA/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/imunologia , Infecções por Circoviridae/patologia , Infecções por Circoviridae/virologia , Citocinas/sangue , Expressão Gênica , Ordem dos Genes , Imunidade Humoral , Imuno-Histoquímica , Linfonodos/metabolismo , Linfonodos/patologia , Camundongos , Plasmídeos/genética , Proteínas Recombinantes de Fusão/genética , Suínos , Carga Viral
17.
Reproduction ; 145(6): 577-85, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23579188

RESUMO

The aim of this study was to examine the expression and regulation of the crystallin, alpha B (Cryab) gene in mouse uterus during the peri-implantation period by in situ hybridization and real-time PCR. There was no detectable Cryab mRNA signal on days 1-4 of pregnancy. On day 5 of pregnancy when embryo implanted, a high level of Cryab mRNA signal was found in the subluminal stroma surrounding the implanting blastocyst. On days 6-8, Cryab mRNA was strongly expressed in the primary decidua. By real-time PCR, a high level of Cryab expression was detected on days 7 and 8 of pregnancy, although Cryab expression was seen from days 1 to 8. Under in vivo and in vitro artificial decidualization, Cryab expression was significantly elevated. Compared with the progesterone-primed delayed implantation uterus, a high level of Cryab mRNA expression was observed in estrogen-activated implantation uterus. In the uterine stromal cells, cAMP, estrogen, and progesterone could induce the expression of Cryab gene. In the ovariectomized mouse uterus, estrogen could also induce the expression of Cryab while progesterone inhibited its expression. Our data suggest that Cryab may play an important role during mouse embryo implantation and decidualization and that estrogen and progesterone can regulate the expression of Cryab gene.


Assuntos
Decídua/metabolismo , Implantação do Embrião , Endométrio/metabolismo , Período Fértil/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Útero/metabolismo , Cadeia B de alfa-Cristalina/biossíntese , Animais , Decídua/efeitos dos fármacos , Implantação do Embrião/efeitos dos fármacos , Implantação Tardia do Embrião/efeitos dos fármacos , Endométrio/efeitos dos fármacos , Terapia de Reposição de Estrogênios , Estrogênios/farmacologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Hibridização In Situ , Camundongos , Ovariectomia/efeitos adversos , Placentação/efeitos dos fármacos , Gravidez , Progesterona/farmacologia , Progestinas/farmacologia , Pseudogravidez/metabolismo , RNA Mensageiro/metabolismo , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo , Útero/efeitos dos fármacos , Cadeia B de alfa-Cristalina/genética , Cadeia B de alfa-Cristalina/metabolismo
18.
Mol Ther Nucleic Acids ; 28: 435-449, 2022 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-35505966

RESUMO

Transcriptional co-activator with PDZ-binding motif (TAZ), one of core modules of the Hippo pathway, involves inflammatory cell infiltration in the liver, but little information is available regarding its physiological function in the microglia-mediated inflammatory response. Here we revealed that activation of TAZ prevented microglia production of proinflammatory cytokines, indicating TAZ's importance in anti-inflammation. After translocation into the nucleus, TAZ interacted with transcriptional enhanced associate domain (TEAD) and bound to the promoter of nuclear factor erythroid 2-related factor 2 (Nrf2), whose blockage caused inability of TAZ to improve inflammation, implying that Nrf2 is a direct target of TAZ. Further analysis showed that TAZ induced Nrf2 nuclear translocation to enhance antioxidant capacity with attenuation of oxidative stress and the inflammatory response. Under inflammatory conditions, TAZ impeded mitochondrial dysfunction, as indicated by amelioration of ATP levels, mtDNA copy numbers, and mitochondrial membrane potential with an obvious reduction in mitochondrial superoxide, but this impediment was neutralized by blockage of Nrf2. TAZ hindered opening of the mitochondrial permeability transition pore, restrained release of cytochrome c from mitochondria into the cytosol, and was sufficient to rescue microglia from apoptosis dependent on Nrf2. Nrf2 acted as a downstream target of TAZ to repress NF-κB activation by enhancing antioxidant capacity. Collectively, TAZ might ameliorate the microglia-mediated inflammatory response through the Nrf2-reactive oxygen species (ROS)-nuclear factor κB (NF-κB) pathway.

19.
Int J Biol Sci ; 18(6): 2261-2276, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35414789

RESUMO

Yap is required for ovarian follicle and early embryo development, but little information is available regarding its physiological significance in decidualization. Here we determine the effects of YAP on decidualization, mitochondrial function, cell apoptosis and DNA damage, and explore its interplay with Bmp2, Rrm2, GSH and ROS. The results exhibited that Yap was abundant in decidual cells and its inactivation impaired the proliferation and differentiation of stromal cells along with the deferral of G1/S phase transition, indicating Yap importance in decidualization. Bmp2 via Alk2 receptor promoted nuclear translocation of Yap where it might interact with Tead and then bind to the promoter of Rrm2 whose activation rescued the faultiness of differentiation program and attenuated oxidative DNA damage caused by Yap impediment. Meanwhile, Yap had an important part in the crosstalk between Bmp2 and Rrm2. Furthermore, inactivation of Yap resulted in an obvious accumulation of intracellular ROS followed by the abnormal GR activity and GSH content dependent on Rrm2. Replenishment of GSH counteracted the regulation of Yap inactivation on stromal differentiation and DNA damage with distinct reduction for intracellular ROS. Additionally, blockage of Yap caused the enhancement of stromal cell apoptosis and brought about mitochondrial dysfunction as indicated by the aberration for ATP level, mtDNA copy number and mitochondrial membrane potential concomitant with the opening of mitochondrial permeability transition pore, but these abnormalities were neutralized by GSH. Administration of mitochondrial antioxidant Mito-TEMPO rescued the fault of stromal differentiation conferred by Yap inactivation. Collectively, Yap was essential for uterine decidualization through Rrm2/GSH/ROS pathway in response to Bmp2.


Assuntos
Células Estromais , Útero , Diferenciação Celular/fisiologia , Feminino , Humanos , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Células Estromais/metabolismo , Útero/metabolismo
20.
Exp Mol Med ; 53(9): 1307-1318, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34497345

RESUMO

TAZ, as a crucial effector of Hippo pathway, is required for spermatogenesis and fertilization, but little is known regarding its physiological function in uterine decidualization. In this study, we showed that TAZ was localized in the decidua, where it promoted stromal cell proliferation followed by accelerated G1/S phase transition via Ccnd3 and Cdk4 and induced the expression or activity of stromal differentiation markers Prl8a2, Prl3c1 and ALP, indicating the importance of TAZ in decidualization. Knockdown of TAZ impeded HB-EGF induction of stromal cell proliferation and differentiation. Under oxidative stress, TAZ protected stromal differentiation against oxidative damage by reducing intracellular ROS and enhancing cellular antioxidant capacity dependent on the Nrf2/ARE/Foxo1 pathway. TAZ strengthened the transcriptional activity of Nrf2 which directly bound to the antioxidant response element (ARE) of Foxo1 promoter region. Additionally, silencing TAZ caused accumulation of intracellular ROS through heightening NOX activity whose blockade by APO reversed the disruption in stromal differentiation. Further analysis revealed that TAZ might restore mitochondrial function, as indicated by the increase in ATP level, mtDNA copy number and mitochondrial membrane potential with the reduction in mitochondrial superoxide. Additionally, TAZ modulated the activities of mitochondrial respiratory chain complexes I and III whose suppression by ROT and AA resulted in the inability of TAZ to defend against oxidative damage to stromal differentiation. Moreover, TAZ prevented stromal cell apoptosis by upregulating Bcl2 expression and inhibiting Casp3 activity and Bax expression. In summary, TAZ might mediate HB-EGF function in uterine decidualization through Ccnd3 and ameliorate oxidative damage to stromal cell differentiation via Nrf2/ARE/Foxo1 pathway.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Elementos de Resposta Antioxidante , Decídua/fisiologia , Proteína Forkhead Box O1/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo , Transdução de Sinais , Animais , Antioxidantes/metabolismo , Apoptose , Diferenciação Celular , Feminino , Proteína Forkhead Box O1/genética , Regulação da Expressão Gênica , Camundongos , Mitocôndrias/metabolismo , Oxirredução , Estresse Oxidativo/genética , Gravidez , Interferência de RNA , Espécies Reativas de Oxigênio/metabolismo , Células Estromais/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa