Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 249
Filtrar
1.
Immunol Invest ; 51(5): 1313-1329, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34132165

RESUMO

Prolactin (PRL) is a pleiotropic hormone with a key role in pregnancy. In fetal membranes, PRL can regulate the secretion of pro-inflammatory factors, which induces the activation of matrix metalloproteinases (MMPs). The increase and activation of MMPs deregulate the turnover of the extracellular matrix in the fetal membranes, altering its structure and function, causing premature rupture of the membranes and preterm labor. In this work, we evaluate the effect of PRL upon the secretion of MMP-1, MMP-2, MMP-9, MMP-13, and the tissue inhibitors of metalloproteinases (TIMPs) in human fetal membranes after lipopolysaccharide (LPS) challenge. Nine fetal membranes from healthy non-laboring cesarean deliveries at term were cultured in a 2-independent chamber system and pre-treated with 250, 500, 1000 or 4000 ng/ml of PRL for 24 h, then choriodecidual region was stimulated with 500 ng/ml of LPS plus fresh PRL for 24 h. The MMPs and TIMPs secretion were quantified by ELISA, additionally MMP-2 and MMP-9 gelatinolytic activity was measured by zymography. LPS induced the MMP-9 and MMP-1 secretion, but no MMP-2 or MMP-13 in comparison with basal levels. PRL co-treatment decreased the MMP-2, MMP-9 and MMP-1 secretion induced by LPS. The active forms were present in the tissue extract, showing a response consistent with the secretion profile. TIMP-1 and TIMP-2 secretion was decreased after LPS treatment and the PRL co-treatment reverts this effect. The present results support that PRL may favor the balance between these factors involved in the structural maintenance of fetal membranes in an inflammatory event.


Assuntos
Anti-Inflamatórios , Membranas Extraembrionárias , Inflamação , Metaloproteinase 9 da Matriz , Metaloproteinases da Matriz Secretadas , Prolactina , Anti-Inflamatórios/farmacologia , Regulação para Baixo , Membranas Extraembrionárias/efeitos dos fármacos , Membranas Extraembrionárias/metabolismo , Feminino , Humanos , Inflamação/tratamento farmacológico , Inflamação/etiologia , Inflamação/metabolismo , Inflamação/terapia , Lipopolissacarídeos/efeitos adversos , Metaloproteinase 1 da Matriz/metabolismo , Metaloproteinase 13 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Metaloproteinases da Matriz Secretadas/metabolismo , Gravidez , Prolactina/farmacologia , Técnicas de Cultura de Tecidos , Inibidores Teciduais de Metaloproteinases/metabolismo
2.
Placenta ; 97: 79-88, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32792069

RESUMO

INTRODUCTION: We use an in-vitro human fetal membrane (FM) explant-based model to study inflammation-induced FM weakening, a prerequisite for PPROM. In this system, GMCSF is a critical intermediate, both necessary and sufficient for TNFα and thrombin induced FM weakening. α-Lipoic-acid (LA) blocks TNFα and thrombin, as well as GMCSF-induced weakening. Recently, we reported LA concomitantly blocks GMCSF-induction of MMPs 2, 9 and 10 and inhibition of TIMPs 1-3. The aim of this study was to show that LA blocks GMCSF-induced increases in additional proteases and reductions in additional protease inhibitors. METHODS: FM fragments were cultured±LA and then±GMCSF. In other experiments, weak versus strong, fresh FM were cultured without additions. Fragments were strength tested and media analyzed by multiplex protein ELISA for proteases and protease inhibitors. RESULTS: GMCSF induced FM weakening and concomitantly increased several Proteases (Cathepsin-S, Proteinase-3, Elastase-2) and decreased several protease inhibitors (NGAL, Cystatin-C, HE4 and Thrombospondin1). LA inhibited GMCSF-induced FM weakening and all enzymatic changes. Untreated weaker versus stronger regions of fresh FM showed comparable differences in proteases and protease inhibitor patterns to GMCSF-stimulated versus controls. CONCLUSION: LA blocks GMCSF-induced human FM weakening and associated protease increases and inhibitor decreases. The GMCSF-induced spectrum of protease/protease-inhibitor changes is similar to that in the natural weak FM fragments. In concert with previously reported GMCSF-induced changes in MMPs & TIMPs, these other protease and protease-inhibitor changes presumably facilitate FM weakening and rupture. LA blocks these GMCSF effects and therefore may be a useful agent to prevent PPROM.


Assuntos
Membranas Extraembrionárias/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Peptídeo Hidrolases/metabolismo , Ácido Tióctico/farmacologia , Cistatinas/metabolismo , Membranas Extraembrionárias/metabolismo , Feminino , Humanos , Gravidez , Trombina/metabolismo , Trombospondina 1/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
3.
Metallomics ; 12(6): 935-951, 2020 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-32373896

RESUMO

Spontaneous preterm birth, which can affect up to 20% of all pregnancies, is the greatest contributor to perinatal morbidity and mortality. Infection is the leading pathological cause of spontaneous preterm birth. Infection activates the maternal immune system, resulting in the upregulation of pro-inflammatory and pro-labor mediators that activate myometrial contractions and rupture of fetal membranes. Anti-inflammatory agents therefore have the potential for the prevention of spontaneous preterm birth. Selenium, an essential micronutrient, has been shown to be a potent anti-inflammatory regulator. Notably, clinical and epidemiological studies have suggested a link between selenium and preterm birth. Thus, the aim of this study was to assess the effect of selenite (an inorganic form of selenium) on the expression of pro-inflammatory and pro-labor mediators in human gestational tissues. Human fetal membranes and myometrium were pre-incubated with or without selenite before incubation with the bacterial product lipopolysaccharide (LPS) to stimulate inflammation associated with preterm birth. Selenite blocked LPS-induced expression of pro-inflammatory cytokines and chemokines and enzymes involved in remodelling of myometrium and degradation of fetal membranes. Of note, selenite also suppressed myometrial activation induced by inflammation as evidenced by a decrease in LPS-induced prostaglandin signalling and myometrial cell contractility. These effects of selenite were mediated by the MAPK protein ERK as selenite blunted LPS induced activation of ERK. In conclusion, selenite suppresses key mediators involved in inflammation induced activation of mediators involved in active labor in human fetal membranes and myometrium. These findings support recent clinical studies demonstrating selenium supplementation is associated with decreased incidence of spontaneous preterm birth.


Assuntos
Lipopolissacarídeos/farmacologia , Miométrio/efeitos dos fármacos , Miométrio/metabolismo , Selênio/farmacologia , Western Blotting , Quimiocinas/metabolismo , Membranas Extraembrionárias/efeitos dos fármacos , Membranas Extraembrionárias/metabolismo , Feminino , Humanos , Imunoensaio , Interleucina-6/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
4.
J Exp Biol ; 223(Pt 4)2020 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-32001543

RESUMO

Maternal transfer of steroids to eggs can elicit permanent effects on offspring phenotype. Although testosterone was thought to be a key mediator of maternal effects in birds, we now know that vertebrate embryos actively regulate their exposure to maternal testosterone through steroid metabolism, suggesting testosterone metabolites, not testosterone, may elicit the observed phenotypic effects. To address the role steroid metabolism plays in mediating yolk testosterone effects, we used European starling (Sturnus vulgaris) eggs to characterize the timing of testosterone metabolism and determine whether etiocholanolone, a prominent metabolite of testosterone in avian embryos, is capable of affecting early embryonic development. Tritiated testosterone was injected into freshly laid eggs to characterize steroid movement and metabolism during early development. Varying levels of etiocholanolone were also injected into eggs, with incubation for either 3 or 5 days, to test whether etiocholanolone influences the early growth of embryonic tissues. The conversion of testosterone to etiocholanolone was initiated within 12 h of injection, but the increase in etiocholanolone was transient, indicating that etiocholanolone is also subject to metabolism, and that exposure to maternal etiocholanolone is limited to a short period during early development. Exogenous etiocholanolone manipulation had no significant effect on the growth rate of the embryos or extra-embryonic membranes early in development. Thus, the conversion of testosterone to etiocholanolone may be an inactivation pathway that buffers the embryo from maternal steroids, with any effects of yolk testosterone resulting from testosterone that escapes metabolism; alternatively, etiocholanolone may influence processes other than growth or take additional time to manifest.


Assuntos
Desenvolvimento Embrionário/efeitos dos fármacos , Etiocolanolona/farmacologia , Estorninhos/embriologia , Testosterona/metabolismo , Animais , Gema de Ovo/metabolismo , Embrião não Mamífero/metabolismo , Etiocolanolona/metabolismo , Membranas Extraembrionárias/efeitos dos fármacos , Feminino , Estorninhos/metabolismo , Trítio
5.
Reprod Domest Anim ; 54(12): 1651-1659, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31595997

RESUMO

We have shown that dietary supplementation of n-3 polyunsaturated fatty acid (n-3 PUFA)-rich fish oil (FO) around the breeding time improved the utero-ovarian functions in the goat. Here, we investigated the effect of FO supplementation during the periparturient period on serum n-3 PUFA, prostaglandin F2α metabolite (PGFM), placental expulsion, uterine involution, resumption of oestrus and neonatal vigour. Rohilkhandi goat in advanced gestation (n = 16) was divided into two equal groups. One group was supplemented with FO containing 26% n-3 long-chain PUFA at the rate of 156 mg per kg body weight, while the control group was fed isocaloric palm oil (PO) from -3 to +3 week of kidding. Dietary FO increased serum concentration of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) by 7.3- and 6.6-fold, respectively, after 6 weeks of supplementation. Goats in FO group expelled the foetal membranes 99.1 min earlier (p < .01) than those of PO group. Further, dietary FO significantly decreased the serum PGFM on day 7 post-partum. However, no difference was found on uterine involution, which was complete by day 20 post-partum in either group. Resumption of follicular activity by day 5 post-partum was 87.5% in the FO as compared to 25% in the PO group (p < .05). Similarly, occurrence of behavioural oestrus by day 90 post-partum was 57.1% in goats of the FO group while none of does was in the PO group (p < .01) expressed oestrus. It was concluded that feeding FO-rich diet during -3 to +3 weeks of kidding decreased the PGFM till day 7 post-partum, hastened the expulsion of foetal membranes and reduced the time from kidding to first post-partum oestrus in Rohilkhandi does.


Assuntos
Membranas Extraembrionárias/efeitos dos fármacos , Óleos de Peixe/farmacologia , Cabras , Folículo Ovariano/efeitos dos fármacos , Útero/efeitos dos fármacos , Ração Animal/análise , Fenômenos Fisiológicos da Nutrição Animal , Animais , Dieta/veterinária , Suplementos Nutricionais , Dinoprosta/sangue , Estro/efeitos dos fármacos , Membranas Extraembrionárias/fisiologia , Ácidos Graxos Ômega-3/administração & dosagem , Feminino , Óleos de Peixe/química , Folículo Ovariano/fisiologia , Gravidez , Útero/fisiologia
6.
Anim Reprod Sci ; 206: 11-16, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31103349

RESUMO

This prospective study was conducted to compare two treatment protocols for retained foetal membranes (RFM) in cattle. The routine therapy consisting of intrauterine tetracycline boluses was compared with the administration of ozone foam, both intrauterine therapies without attempts to manually remove the placenta. Cows undergoing caesarean section, suffering from uterine torsion or prolapse of the uterus, and other illnesses than RFM on the first day post-partum were excluded. Values for baseline variables as age, breed, yearly milk yield, parity, pregnancy duration, calving season and number of routine intrauterine treatments were not different between the groups. Number of days with rectal temperature >39.7 °C within the first 10 days in milk (DIM) were greater in the ozone group. Number of escape therapies were recorded when there was imposing of this therapy as a result of cows having a fever (based on rectal temperature >39.7 °C) and a significant reduced food intake. Escape therapies consisted of parenteral administration of tetracyclines and NSAIDS during the first 10 DIM. Number of escape therapies, however, was not different between the groups. Results from use of a logistic regression model with fever as a dichotomous variable indicated there was no effect of breed, studbook, type of stall, pasture, business and no effect of age or milk yield. In a second assessment, cows were evaluated until 200 DIM. Results from a survival analysis indicated group (Te/Oz) did not affect rate of culling from the herd.


Assuntos
Doenças dos Bovinos/tratamento farmacológico , Membranas Extraembrionárias/efeitos dos fármacos , Ozônio/farmacologia , Placenta Retida/tratamento farmacológico , Placenta Retida/veterinária , Tetraciclina/farmacologia , Animais , Antibacterianos/farmacologia , Bovinos , Feminino , Gravidez , Estudos Prospectivos , Útero/efeitos dos fármacos
7.
Endocrinology ; 160(4): 925-937, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30776301

RESUMO

Glucocorticoid use during pregnancy is known to increase the risk of preterm birth and preterm premature rupture of membranes (pPROM). Here, we investigated the mechanism of how glucocorticoids weaken the fetal membranes. The amnion mesenchymal layer was significantly thinner in pregnant women treated with prednisolone and in corticosterone-injected mice than in control groups. Matrix metalloproteinase (MMP)-9 mRNA and its activity, COX2 mRNA levels, and prostaglandin E2 synthesis were increased, whereas type 1 collagen (COL1A1) mRNA levels were decreased in the fetal membranes of corticosterone-injected mice. Unexpectedly, the proliferation and migration of macrophages were observed around the corticosterone-injected amnion, and IL-1ß was released from these macrophages. In human amnion mesenchymal cells, cortisol did not change MMP mRNA expression, whereas IL-1ß treatment robustly increased MMP and COX2 mRNA expression. COL1A1 mRNA level was decreased by both cortisol and IL-1ß. These data suggest that the recruitment of amniotic macrophages by glucocorticoids plays a pivotal role in weakening of the fetal membranes, leading to the pathogenesis of pPROM.


Assuntos
Âmnio/efeitos dos fármacos , Corticosterona/administração & dosagem , Glucocorticoides/administração & dosagem , Macrófagos/efeitos dos fármacos , Prednisolona/administração & dosagem , Adulto , Âmnio/metabolismo , Animais , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ciclo-Oxigenase 2/metabolismo , Membranas Extraembrionárias/efeitos dos fármacos , Membranas Extraembrionárias/metabolismo , Feminino , Humanos , Macrófagos/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Adulto Jovem
8.
Phytomedicine ; 49: 11-22, 2018 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-30217257

RESUMO

BACKGROUND: Preterm birth is the most prominent complication attributing to poor pregnancy and neonatal outcome. Infection is most commonly implicated in preterm birth; it initiates a cascade of inflammatory events that leads to the rupture of fetal membranes and spontaneous uterine contractions. Anti-inflammatory agents may thus be a therapeutic approach to prevent the premature rupture of fetal membranes and block contractions. In non-gestational tissues, the polyphenol honokiol has been shown to possess potent anti-inflammatory properties. PURPOSE: The aim of this study was to investigate the effect of honokiol on pro-inflammatory mediators in human gestational tissues. METHODS: Fetal membranes, myometrium and freshly isolated amnion cells and primary myometrial cells were treated with honokiol in the absence or presence of the products lipopolysaccharide (LPS) and fibroblast-stimulating lipopeptide-1 (fsl-1), the viral dsRNA analogue polyinosinic:polycytidylic acid (poly(I:C)) or the pro-inflammatory cytokines TNF or IL1B. A luciferase assay was used to determine the effect of honokiol on nuclear factor kappa B (NF-κB) RelA transcriptional activity. RESULTS: Honokiol significantly decreased pro-inflammatory cytokine (IL1A, IL6) and chemokine (CXCL8, CXCL1, CCL2) mRNA expression and secretion from fetal membranes (amnion and choriodecidua) and myometrium stimulated with LPS, fsl-1 or poly(I:C). In amnion cells, honokiol also significantly decreased the expression and secretion of the extracellular matrix degrading enzyme MMP9. Moreover, in myometrium, honokiol significantly suppressed the expression of the contraction associated protein PTGFR, the secretion of the uterotonic prostaglandins PGE2 and PGF2α, and blocked TNF-induced myometrial cell contractility. Finally, honokiol significantly suppressed IL1B- and TNF-induced NF-κB RelA transcriptional activity in primary amnion and myometrial cells. CONCLUSIONS: Honokiol reduced the expression of pro-inflammatory and pro-labour mediators in human amnion, choriodecidua and myometrium and that this may be facilitated through the suppression of NF-κB activation. These results indicate that the polyphenol honokiol may be a potent therapeutic for the prevention of preterm birth.


Assuntos
Anti-Inflamatórios/farmacologia , Compostos de Bifenilo/farmacologia , Membranas Extraembrionárias/efeitos dos fármacos , Lignanas/farmacologia , Miométrio/efeitos dos fármacos , Quimiocina CCL2/metabolismo , Quimiocina CXCL1/metabolismo , Dinoprosta/metabolismo , Dinoprostona/metabolismo , Feminino , Humanos , Interleucina-1beta/farmacologia , Interleucina-8/metabolismo , Gravidez , Nascimento Prematuro/prevenção & controle , Cultura Primária de Células , Fator de Transcrição RelA/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
9.
Placenta ; 68: 9-14, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30055672

RESUMO

INTRODUCTION: We established an in-vitro model for the study of human fetal membrane (FM) weakening leading to pPROM. In this model, granulocyte-macrophage colony-stimulating factor (GM-CSF) is a critical intermediate for both tumor necrosis factor-α (TNF; modeling infection/inflammation) and thrombin (modeling decidual bleeding/abruption)-induced weakening. Thus, inhibitors of FM weakening can be categorized as targeting GM-CSF production, GM-CSF downstream action, or both. Most progestogens inhibit both, except 17-α hydroxyprogesterone caproate which inhibits FM weakening at only one point, GM-CSF production. α-lipoic acid (LA), an over-the-counter dietary supplement, has also been previously shown to inhibit TNF and thrombin induced FM weakening. OBJECTIVE: To determine the point of action of LA inhibition of FM weakening. METHODS: FM fragments were mounted in Transwell inserts and preincubated with/without LA/24 h, then with/without addition of TNF, thrombin or GM-CSF. After 48 h, medium was assayed for GM-CSF, and FM fragments were rupture-strength tested. RESULTS: TNF and thrombin both weakened FM and increased GM-CSF levels. GM-CSF also weakened FM. LA inhibited both TNF and thrombin induced FM weakening and concomitantly inhibited the increase in GM-CSF in a concentration-dependent manner. In addition, LA inhibited GM-CSF induced FM weakening in a concentration dependent manner. CONCLUSIONS: LA blocks TNF and thrombin induced FM weakening at two points, inhibiting both GM-CSF production and downstream action. Thus, we speculate that LA may be a potential standalone therapeutic agent, or supplement to current therapy for prevention of pPROM related spontaneous preterm birth, if preclinical studies to examine feasibility and safety during pregnancy are successfully accomplished.


Assuntos
Membranas Extraembrionárias/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Inflamação/metabolismo , Ácido Tióctico/farmacologia , Ruptura Prematura de Membranas Fetais/metabolismo , Humanos , Técnicas In Vitro , Trombina/farmacologia , Fator de Necrose Tumoral alfa/farmacologia
10.
Am J Reprod Immunol ; 80(1): e12861, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29709093

RESUMO

PROBLEM: Chorioamnionitis and infection-associated inflammation are major causes of preterm birth. Magnesium sulfate (MgSO4 ) is widely used in obstetrics as a tocolytic; however, its mechanism of action is unclear. This study sought to investigate how MgSO4 modulates infection-associated inflammation in fetal membranes (FMs), and whether the response was time dependent. METHOD OF STUDY: Human FM explants were treated with or without bacterial lipopolysaccharide (LPS); with or without MgSO4 added either: 1 hour before LPS; at the same time as LPS; 1 hour post-LPS; or 2 hours post-LPS. Explants were also treated with or without viral dsRNA and LPS, alone or in combination; and MgSO4 added 1 hour post-LPS After 24 hours, supernatants were measured for cytokines/chemokines; and tissue lysates measured for caspase-1 activity. RESULTS: Lipopolysaccharide-induced FM inflammation by upregulating the secretion of a number of inflammatory cytokines/chemokines. Magnesium sulfate administered 1-hour post-LPS inhibited FM secretion of IL-1ß, IL-6, G-CSF, RANTES, and TNFα. Magnesium sulfate administered 2 hours post-LPS augmented FM secretion of these factors as well as IL-8, IFNγ, VEGF, GROα and IP-10. Magnesium sulfate delivered 1- hour post-LPS inhibited LPS-induced caspase-1 activity, and inhibited the augmented IL-1ß response triggered by combination viral dsRNA and LPS. CONCLUSION: Magnesium sulfate differentially modulates LPS-induced FM inflammation in a time-dependent manner, in part through its modulation of caspase-1 activity. Thus, the timing of MgSO4 administration may be critical in optimizing its anti-inflammatory effects in the clinical setting. MgSO4 might also be useful at preventing FM inflammation triggered by a polymicrobial viral-bacterial infection.


Assuntos
Membranas Extraembrionárias/efeitos dos fármacos , Inflamação/tratamento farmacológico , Sulfato de Magnésio/farmacologia , Caspase 1/metabolismo , Corioamnionite/metabolismo , Citocinas/metabolismo , Membranas Extraembrionárias/metabolismo , Feminino , Humanos , Inflamação/metabolismo , Interleucina-1beta/metabolismo , Gravidez , Nascimento Prematuro/prevenção & controle , Fatores de Tempo , Regulação para Cima/efeitos dos fármacos
11.
Reprod Sci ; 25(8): 1161-1167, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29463188

RESUMO

OBJECTIVE: Progesterone supplementation prevents preterm birth (PTB) in some high-risk women, but its mechanism of action is unknown. One-third of PTB is associated with preterm premature rupture of membranes (PPROMs). We have previously shown that progesterone inhibits basal and Tumor Necrosis Factor (TNF) α-induced apoptosis in an explant model of human fetal membranes. This study investigates the molecular mechanisms responsible for progesterone-mediated inhibition of apoptosis in fetal membranes. METHODS: Human fetal membranes were collected at elective cesarean at term (no labor, no infection; n = 6), washed, and pretreated with/without progesterone (125 ng/mL) for 24 hours. Thereafter, membranes were treated with/without TNFα (50 ng/mL) and/or progesterone for 48 hours, harvested, and homogenized. Apoptosis was determined by evaluating caspase-3, -8, and -9 activities. Expression of pro- BH3 interacting domain death against, Bc1-2 associated X protein (BID, BAX) and antiapoptotic proteins (X-linked inhibitor of apoptosis protein [XIAP], Bcl-2, FLICE inhibitory protein [FLIP]) were measured by Western blot. RESULTS: TNFα increased apoptosis (measured by caspase-3, -8, and -9 activities) in fetal membranes, and this effect was abrogated by progesterone. Under basal conditions, progesterone suppressed expression of the proapoptotic protein, BID, by 0.45 (0.14)-fold, and increased expression of the antiapoptotic proteins, Bcl-2 and XIAP; no change was seen in BAX or FLIP. In contrast, TNFα increased BID expression by 5.15 (2.92)-fold, which was prevented by pretreatment with progesterone. CONCLUSIONS: Progesterone inhibits apoptosis in fetal membranes by suppressing expression of the proapoptotic protein, BID (for both basal and TNFα-induced apoptosis), and upregulating expression of the antiapoptotic proteins, XIAP and Bcl-2 (under basal conditions only). These data provide a mechanism by which progesterone supplementation may prevent PPROM and PTB in some women at high risk.


Assuntos
Apoptose , Membranas Extraembrionárias/efeitos dos fármacos , Membranas Extraembrionárias/metabolismo , Progesterona/metabolismo , Humanos , Progesterona/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo
12.
Eur J Pharmacol ; 824: 48-56, 2018 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-29409911

RESUMO

Accumulating epidemiological evidence indicates that infection with Porphyromonas gingivalis which is a major periodontal pathogen, causes preterm birth and low birth weight. However, virulence factors of P. gingivalis responsible for preterm birth/low birth weight remain to be elucidated. In this study, using P. gingivalis-infected pregnant mice as an in vivo model, we investigated whether gingipains-cysteine proteinases produced by P. gingivalis-affect preterm birth and low birth weight. We found that intravenous infection of pregnant mice with P. gingivalis induced higher accumulation of the bacterium in the placenta than that in other organs. Compared to infection with P. gingivalis wild-type, infection with a gingipain-deficient P. gingivalis mutant KDP136 led to significant reduction in preterm birth and pregnancy loss. Although repetitive low-level infections of P. gingivalis failed to induce preterm birth and fetal death, it induced suppressive effects on IFN-γ production. Therapeutically, treatment with ginginpain inhibitors prevented fetal death and preterm birth caused by P. gingivalis infection and resulted in recovery of IFN-γ suppression caused by repetitive chronic P. gingivalis infection. These results indicate that gingipains are major virulence factors of P. gingivalis responsible for preterm birth/low birth, and gingipain inhibitors may be useful not only as a therapeutic agent for periodontal diseases, but also as a preventive medicine for preterm birth/low birth weight.


Assuntos
Adesinas Bacterianas/metabolismo , Cisteína Endopeptidases/metabolismo , Inibidores de Cisteína Proteinase/farmacologia , Morte Fetal/etiologia , Morte Fetal/prevenção & controle , Porphyromonas gingivalis/fisiologia , Nascimento Prematuro/microbiologia , Nascimento Prematuro/prevenção & controle , Animais , Citocinas/biossíntese , Membranas Extraembrionárias/efeitos dos fármacos , Membranas Extraembrionárias/microbiologia , Feminino , Cisteína Endopeptidases Gingipaínas , Camundongos , Mutação , Placenta/efeitos dos fármacos , Placenta/microbiologia , Porphyromonas gingivalis/genética , Gravidez , Nascimento Prematuro/metabolismo
13.
Am J Obstet Gynecol ; 217(6): 695.e1-695.e14, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29031893

RESUMO

BACKGROUND: The progestogen 17-α hydroxyprogesterone caproate (17-OHPC) is 1 of only 2 agents recommended for clinical use in the prevention of spontaneous preterm delivery, and studies of its efficacy have been conflicting. We have developed an in-vitro model to study the fetal membrane weakening process that leads to rupture in preterm premature rupture of the fetal membranes (pPROM). Inflammation/infection associated with tumor necrosis factor-α (TNF-α) induction and decidual bleeding/abruption associated thrombin release are leading causes of preterm premature rupture of the fetal membranes. Both agents (TNF-α and thrombin) cause fetal membrane weakening in the model system. Furthermore, granulocyte-macrophage colony-stimulating factor (GM-CSF) is a critical intermediate for both TNF-α and thrombin-induced fetal membrane weakening. In a previous report, we demonstrated that 3 progestogens, progesterone, 17-alpha hydroxyprogesterone (17-OHP), and medroxyprogesterone acetate (MPA), each inhibit both TNF-α- and thrombin-induced fetal membrane weakening at 2 distinct points of the fetal membrane weakening pathway. Each block both the production of and the downstream action of the critical intermediate granulocyte-macrophage colony-stimulating factor. OBJECTIVE: The objective of the study was to characterize the inhibitory effects of 17-OHPC on TNF-α- and thrombin-induced fetal membrane weakening in vitro. STUDY DESIGN: Full-thickness human fetal membrane fragments from uncomplicated term repeat cesarean deliveries were mounted in 2.5 cm Transwell inserts and cultured with/without 17-alpha hydroxyprogesterone caproate (10-9 to 10-7 M). After 24 hours, medium (supernatant) was removed and replaced with/without the addition of tumor necrosis factor-alpha (20 ng/mL) or thrombin (10 U/mL) or granulocyte-macrophage colony-stimulating factor (200 ng/mL). After 48 hours of culture, medium from the maternal side compartment of the model was assayed for granulocyte-macrophage colony-stimulating factor and the fetal membrane fragments were rupture strength tested. RESULTS: Tumor necrosis factor-alpha and thrombin both weakened fetal membranes (43% and 62%, respectively) and increased granulocyte-macrophage colony-stimulating factor levels (3.7- and 5.9-fold, respectively). Pretreatment with 17-alpha hydroxyprogesterone caproate inhibited both tumor necrosis factor-alpha- and thrombin-induced fetal membrane weakening and concomitantly inhibited the induced increase in granulocyte-macrophage colony-stimulating factor in a concentration-dependent manner. However, contrary to our prior reports regarding progesterone and other progestogens, 17-alpha hydroxyprogesterone caproate did not also inhibit granulocyte-macrophage colony-stimulating factor-induced fetal membrane weakening. CONCLUSION: 17-Alpha hydroxyprogesterone caproate blocks tumor necrosis factor-alpha- and thrombin-induced fetal membrane weakening by inhibiting the production of granulocyte-macrophage colony-stimulating factor. However, 17-alpha hydroxyprogesterone caproate did not also inhibit granulocyte-macrophage colony-stimulating factor-induced weakening. We speculate that progestogens other than 17-alpha hydroxyprogesterone caproate may be more efficacious in preventing preterm premature rupture of the fetal membranes-related spontaneous preterm birth.


Assuntos
Membranas Extraembrionárias/efeitos dos fármacos , Ruptura Prematura de Membranas Fetais/prevenção & controle , Hidroxiprogesteronas/farmacologia , Progestinas/farmacologia , Caproato de 17 alfa-Hidroxiprogesterona , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Hemostáticos/farmacologia , Humanos , Técnicas In Vitro , Modelos Biológicos , Gravidez , Nascimento Prematuro/prevenção & controle , Trombina/farmacologia , Fator de Necrose Tumoral alfa/farmacologia
14.
Am J Reprod Immunol ; 78(1)2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28466996

RESUMO

PROBLEM: Intrauterine infection caused by viral infection has been implicated to contribute to preterm birth. Hepatitis A virus cellular receptor 2 (HAVCR2) regulates inflammation in non-gestational tissues in response to viral infection. METHOD OF STUDY: The aims of this study were to determine the effect of: (i) viral dsRNA analogue polyinosinic:polycytidylic acid (poly(I:C)) on HAVCR2 expression; and (ii) HAVCR2 silencing by siRNA (siHAVCR2) in primary amnion and myometrial cells on poly(I:C)-induced inflammation. RESULTS: In human foetal membranes and myometrium, HAVCR2 mRNA and protein expression was decreased when exposed to poly(I:C). Treatment of primary amnion and myometrial cells with poly(I:C) significantly increased the expression and release of pro-inflammatory cytokines TNF, IL1A, IL1B and IL6; the expression of chemokines CXCL8 and CCL2; the expression and secretion of adhesion molecules ICAM1 and VCAM1; and PTGS2 and PTGFR mRNA expression and the release of prostaglandin PGF2α . This increase was significantly augmented in cells transfected with siHAVCR2. Furthermore, mRNA expression of anti-inflammatory cytokines IL4 and IL10 was significantly decreased. CONCLUSION: Collectively, our data suggest that HAVCR2 regulates cytokines, chemokines, prostaglandins and cell adhesion molecules in the presence of viral infection. This suggests a potential for HAVCR2 activators as therapeutics for the management of preterm birth associated with viral infections.


Assuntos
Membranas Extraembrionárias/metabolismo , Receptor Celular 2 do Vírus da Hepatite A/metabolismo , Miométrio/metabolismo , Viroses/metabolismo , Adolescente , Adulto , Ciclo-Oxigenase 2/genética , Citocinas/metabolismo , Dinoprosta/metabolismo , Membranas Extraembrionárias/efeitos dos fármacos , Feminino , Receptor Celular 2 do Vírus da Hepatite A/genética , Humanos , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/metabolismo , Miométrio/efeitos dos fármacos , Trabalho de Parto Prematuro/genética , Trabalho de Parto Prematuro/metabolismo , Poli I-C/farmacologia , Gravidez , Complicações Infecciosas na Gravidez/genética , Complicações Infecciosas na Gravidez/metabolismo , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , Receptores de Prostaglandina/genética , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/metabolismo , Viroses/genética , Adulto Jovem
15.
Mol Hum Reprod ; 23(6): 428-440, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28333279

RESUMO

STUDY QUESTION: Does proviral integration site for Moloney murine leukaemic virus (PIM)1 kinase play a role in regulating the inflammatory processes of human labour and delivery? SUMMARY ANSWER: PIM1 kinase plays a critical role in foetal membranes in regulating pro-inflammatory and pro-labour mediators. WHAT IS KNOWN ALREADY: Infection and inflammation have strong causal links to preterm delivery by stimulating pro-inflammatory cytokines and collagen degrading enzymes, which can lead to rupture of membranes. PIM1 has been shown to have a role in immune regulation and inflammation in non-gestational tissues; however, its role has not been explored in the field of human labour. STUDY DESIGN, SIZE, DURATION: PIM1 expression was analysed in myometrium and/or foetal membranes obtained at term and preterm (n = 8-9 patients per group). Foetal membranes, freshly isolated amnion cells and primary myometrial cells were used to investigate the effect of PIM1 inhibition on pro-labour mediators (n = 5 patients per treatment group). PARTICIPANTS/MATERIALS, SETTING AND METHODS: Foetal membranes, from term and preterm, were obtained from non-labouring and labouring women, and from preterm pre-labour rupture of membranes (PPROM) (n = 9 per group). Amnion was collected from women with and without preterm chorioamnionitis (n = 8 per group). Expression of PIM1 kinase was determined by qRT-PCR and western blotting. To determine the effect of PIM1 kinase inhibition on the expression of pro-inflammatory and pro-labour mediators induced by bacterial products lipopolysaccharide (LPS) (10 µg/ml) and flagellin (1 µg/ml) and pro-inflammatory cytokine tumour necrosis factor (TNF) (10 ng/ml), chemical inhibitors SMI-4a (20 µM) and AZD1208 (50 µM) were used in foetal membrane explants and siRNA against PIM1 was used in primary amnion cells. Statistical significance was set at P < 0.05. MAIN RESULTS AND THE ROLE OF CHANCE: PIM1 expression was significantly increased in foetal membranes after spontaneous term labour compared to no labour at term and in amnion with preterm chorioamnionitis compared to preterm with no chorioamnionitis. There was no change in PIM1 expression with preterm labour or PPROM compared to preterm with no labour or PPROM. In human foetal membranes, PIM1 inhibitors SMI-4a and AZD1208 significantly decreased the expression of pro-inflammatory cytokine interleukin-6 (IL6) and chemokines CXCL8 and CCL2 mRNA and release, prostaglandin prostaglandin F2α (PGF2α) release, adhesion molecule intercellular adhesion molecule 1 mRNA expression and release, and oxidative stress marker 8-isoprostane release after stimulation with either LPS or flagellin. Primary amnion cells transfected with PIM1 siRNA also showed decreased expression of IL6, CXCL8 and CCL2, PTGS2 mRNA and PGF2α release, and matrix metalloproteinase-9 (MMP9) expression, when stimulated with TNF. LARGE SCALE DATA: None. LIMITATIONS, REASONS FOR CAUTION: The conclusions were drawn from in vitro experiments using foetal membrane explants and primary cells isolated from amnion. Animal models are necessary to determine whether PIM1 kinase inhibitors can prevent spontaneous preterm birth in vivo. WIDER IMPLICATIONS OF THE FINDINGS: PIM1 kinase inhibitors may provide a novel therapeutic approach for preventing spontaneous preterm birth. STUDY FUNDING/COMPETING INTEREST(S): Associate Professor Martha Lappas is supported by a Career Development Fellowship from the National Health and Medical Research Council (NHMRC; grant no. 1047025). Funding for this study was provided by the NHMRC (grant no. 1058786), Norman Beischer Medical Research Foundation and the Mercy Research Foundation. The authors have no conflict of interest.


Assuntos
Corioamnionite/genética , Membranas Extraembrionárias/efeitos dos fármacos , Ruptura Prematura de Membranas Fetais/genética , Trabalho de Parto Prematuro/genética , Proteínas Proto-Oncogênicas c-pim-1/genética , Compostos de Benzilideno/farmacologia , Compostos de Bifenilo/farmacologia , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Corioamnionite/metabolismo , Corioamnionite/patologia , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Membranas Extraembrionárias/metabolismo , Membranas Extraembrionárias/patologia , Feminino , Ruptura Prematura de Membranas Fetais/metabolismo , Ruptura Prematura de Membranas Fetais/patologia , Flagelina/antagonistas & inibidores , Flagelina/farmacologia , Regulação da Expressão Gênica , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Trabalho de Parto , Lipopolissacarídeos/antagonistas & inibidores , Lipopolissacarídeos/farmacologia , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Miométrio/metabolismo , Miométrio/patologia , Trabalho de Parto Prematuro/metabolismo , Trabalho de Parto Prematuro/patologia , Gravidez , Proteínas Proto-Oncogênicas c-pim-1/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-pim-1/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Tiazolidinedionas/farmacologia , Tiazolidinas/farmacologia , Técnicas de Cultura de Tecidos , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/farmacologia
16.
Stem Cell Res Ther ; 8(1): 31, 2017 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-28173875

RESUMO

BACKGROUND: The study of lipid metabolism in stem cell physiology has recently raised great interest. The role of lipids goes beyond the mere structural involvement in assembling extra- and intra-cellular compartments. Nevertheless, we are still far from understanding the impact of membrane lipidomics in stemness maintenance and differentiation patterns. In the last years, it has been reported how in vitro cell culturing can modify membrane lipidomics. The aim of the present work was to study the membrane fatty acid profile of mesenchymal stromal cells (MSCs) derived from human fetal membranes (hFM-MSCs) and to correlate this to specific biological properties by using chemically defined tailored lipid supplements (Refeed®). METHODS: Freshly isolated hFM-MSCs were characterized for their membrane fatty acid composition. hFM-MSCs were cultivated in vitro following a classical protocol and their membrane fatty acid profile at different passages was compared to the profile in vivo. A tailored Refeed® lipid supplement was developed with the aim of reducing the differences created by the in vitro cultivation and was tested on cultured hFM-MSCs. Cell morphology, viability, proliferation, angiogenic differentiation, and immunomodulatory properties after in vitro exposure to the tailored Refeed® lipid supplement were investigated. RESULTS: A significant modification of hFM-MSC membrane fatty acid composition occurred during in vitro culture. Using a tailored lipid supplement, the fatty acid composition of cultured cells remained more similar to their in vivo counterparts, being characterized by a higher polyunsaturated and omega-6 fatty acid content. These changes in membrane composition had no effect on cell morphology and viability, but were linked with increased cell proliferation rate, angiogenic differentiation, and immunomodulatory properties. In particular, Refeed®-supplemented hFM-MSCs showed greater ability to express fully functional cell membrane molecules. CONCLUSIONS: Culturing hFM-MSCs alters their fatty acid composition. A tailored lipid supplement is able to improve in vitro hFM-MSC functional properties by recreating a membrane environment more similar to the physiological counterpart. This approach should be considered in cell therapy applications in order to maintain a higher cell quality during in vitro passaging and to influence the outcome of cell-based therapeutic approaches when cells are administered to patients.


Assuntos
Antioxidantes/farmacologia , Membrana Celular/efeitos dos fármacos , Metabolismo dos Lipídeos/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Diferenciação Celular , Membrana Celular/química , Proliferação de Células , Suplementos Nutricionais , Membranas Extraembrionárias/citologia , Membranas Extraembrionárias/efeitos dos fármacos , Membranas Extraembrionárias/metabolismo , Ácidos Graxos/análise , Ácidos Graxos/metabolismo , Ácidos Graxos Monoinsaturados/análise , Ácidos Graxos Monoinsaturados/metabolismo , Ácidos Graxos Insaturados/análise , Ácidos Graxos Insaturados/metabolismo , Feminino , Humanos , Lipídeos de Membrana/análise , Lipídeos de Membrana/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Placenta/citologia , Placenta/efeitos dos fármacos , Placenta/metabolismo , Gravidez , Cultura Primária de Células
17.
Int J Biochem Cell Biol ; 81(Pt A): 10-19, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27769742

RESUMO

Thirty percent of preterm births directly result from preterm premature rupture of fetal membranes (PPROM). Clinical management currently proposes using a collagen plug to mechanically stop loss of amniotic fluid. Vitamin A and its active metabolite (retinoic acid) have well-known pro-healing properties and could thus make good candidates as a proposable adjuvant to this mechanical approach. Here we investigate the molecular mechanisms involved in the pro-healing properties of all-trans retinoic acid (atRA) in fetal membranes via an approach using an in vitro primary amniocyte wound model and transcriptomics. The results demonstrate that atRA promotes migration in primary amniocytes, improving wound healing in vitro by up to 90%. This effect is mediated by the induction of LOXL4, which plays a crucial role in the dynamics of the extracellular matrix by regulating collagen reticulation. This new insight into how atRA exerts its pro-healing properties prompts us to propose using atRA as a candidate strategy to help prevent future PPROM.


Assuntos
Aminoácido Oxirredutases/biossíntese , Feto/citologia , Tretinoína/farmacologia , Cicatrização/efeitos dos fármacos , Aminoácido Oxirredutases/genética , Aminoácido Oxirredutases/metabolismo , Movimento Celular/efeitos dos fármacos , Indução Enzimática/efeitos dos fármacos , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Membranas Extraembrionárias/efeitos dos fármacos , Membranas Extraembrionárias/metabolismo , Feminino , Humanos , Gravidez , Regiões Promotoras Genéticas/genética , Proteína-Lisina 6-Oxidase , Ativação Transcricional/efeitos dos fármacos
18.
Placenta ; 47: 63-72, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27780541

RESUMO

INTRODUCTION: Infection and inflammation stimulate pro-inflammatory cytokines, prostaglandins and matrix metalloproteinase (MMP)-9, which play a central role in myometrial contractions and rupture of fetal membranes. In human and mouse immune cells, activating transcription factor 3 (ATF3) is a negative regulator of inflammation. No studies have examined the role of ATF3 in human labour. METHODS: Primary amnion cells were used to determine the effect of interleukin (IL)-1ß and the bacterial product fibroblast-stimulating lipopeptide (fsl-1) on ATF3 expression, and the effect of ATF3 siRNA on pro-labour mediators. ATF3 expression was assessed in fetal membranes from non-labouring and labouring women at term and preterm, and after preterm pre-labour rupture of membranes (PPROM). RESULTS: IL-1ß and fsl-1 significantly increased ATF3 expression. Silencing ATF3 significantly increased IL-1ß- or fsl-1-induced expression of pro-inflammatory cytokines (TNF-α, IL-1α, IL-1ß, IL-6) and chemokines (IL-8 and monocyte chemoattractant protein-1 (MCP-1)); cyclooxygenase-2 (COX-2) mRNA expression and prostaglandin PGF2α release; and MMP-9 expression. ATF3 expression was decreased in fetal membranes with term labour. There was no effect of preterm labour or PPROM on ATF3 expression. DISCUSSION: ATF3 is a negative regulator of inflammation in human fetal membranes; in primary amnion cells, ATF3 expression is induced by IL-1ß and fsl-1, and ATF3 silencing further exacerbates the inflammatory response when stimulated with these factors. Subsequently, ATF3 expression is decreased in fetal membranes after term labour and with preterm chorioamnionitis, conditions closely associated with inflammation and infection. Our data suggest that ATF3 may play a role in the terminal processes of human labour and delivery.


Assuntos
Fator 3 Ativador da Transcrição/metabolismo , Membranas Extraembrionárias/metabolismo , Ruptura Prematura de Membranas Fetais/metabolismo , Inflamação/metabolismo , Fator 3 Ativador da Transcrição/genética , Linhagem Celular , Corioamnionite/metabolismo , Citocinas/genética , Citocinas/metabolismo , Diglicerídeos/farmacologia , Membranas Extraembrionárias/efeitos dos fármacos , Feminino , Ruptura Prematura de Membranas Fetais/genética , Humanos , Interleucina-1beta/farmacologia , Trabalho de Parto/metabolismo , Oligopeptídeos/farmacologia , Gravidez
19.
Cytokine ; 81: 82-7, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26954343

RESUMO

BACKGROUND: High Mobility Group Box-1 (HMGB1) is considered a prototype alarmin molecule. Upon its extracellular release, HMGB1 engages pattern recognition receptors and the Receptor for Advanced Glycation End-products (RAGE) followed by an outpouring of inflammatory cytokines, including interleukin (IL)-6. METHODS: We assayed the amniotic fluid (AF) levels of HMGB1 and IL-6 in 255 women that either had a normal pregnancy outcome or delivered preterm. Immunohistochemistry on fetal membranes was used for cellular localization and validation of immunoassay findings. HMGB1 also was analyzed in amniochorion tissue explants subjected to endotoxin. RESULTS: AF HMGB1 levels are not gestational age regulated but are increased in women with intra-amniotic inflammation and preterm birth. The likely source is the damaged amniochorion, as demonstrated by immunohistochemistry and explant experiments. CONCLUSIONS: Our research supports a role for HMGB1 in the inflammatory response leading to preterm birth. As a delayed phase cytokine, in utero exposure to elevated AF HMGB1 levels may have an impact on the newborn beyond the time of birth.


Assuntos
Líquido Amniótico/metabolismo , Corioamnionite/metabolismo , Membranas Extraembrionárias/metabolismo , Proteína HMGB1/análise , Nascimento Prematuro/metabolismo , Adulto , Corioamnionite/fisiopatologia , Membranas Extraembrionárias/efeitos dos fármacos , Membranas Extraembrionárias/patologia , Feminino , Idade Gestacional , Proteína HMGB1/metabolismo , Humanos , Imunoensaio , Imuno-Histoquímica , Recém-Nascido , Interleucina-6/análise , Lipopolissacarídeos/farmacologia , Gravidez , Nascimento Prematuro/fisiopatologia , Receptor para Produtos Finais de Glicação Avançada/análise , Adulto Jovem
20.
Reprod Sci ; 23(9): 1168-78, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-26919974

RESUMO

Progesterone receptor membrane component 1 (PGRMC1) is an important novel mediator of progesterone (P4) function in fetal membrane cells. We demonstrated previously that PGRMC1 is differentially expressed in fetal membranes among pregnancy subjects and diminished in preterm premature rupture of membrane subjects. In the current study, we aim to elucidate whether PGRMC1 expression is regulated by P4, tumor necrosis factor α (TNF-α), and H2O2 in fetal membrane cells. Primary cultured membrane cells were serum starved for 24 hours followed by treatments of P4, 17 hydroxyprogesterone caproate, and medroxyprogesterone 17 acetate (MPA) at 10(-7) mol/L with ethanol as vehicle control; TNF-α at 10, 20, and 50 ng/mL with phosphate-buffered saline (PBS) as control; and H2O2 at 10 and 100 µmol/L with culture media as control for 24, 48, and 72 hours. The messenger RNA (mRNA) and protein expression of PGRMC1 was quantified using polymerase chain reaction and Western blotting, respectively. We found that PGRMC1 protein expression was regulated by MPA, TNF-α, and H2O2 in a dose-dependent manner. This regulation is also specific to the type of cell (amnion, chorion, or decidua). The upregulation of PGRMC1 by MPA might be mediated through glucocorticoid receptor (GR) demonstrated using amnion and chorion cells model with GR knockdown by specific small interfering RNA transfection. The mRNA expression of PGRMC1 was decreased by H2O2 (100 µmol/L) treatment in amnion cells, which might ultimately result in downregulation of PGRMC1 protein as our data demonstrated. None of other treatments changed PGRMC1 mRNA level in these cells. We conclude that these stimuli act as regulatory factors of PGRMC1 in a cell-specific manner.


Assuntos
Membranas Extraembrionárias/metabolismo , Proteínas de Membrana/metabolismo , Estresse Oxidativo , Progestinas/farmacologia , Receptores de Progesterona/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , 17-alfa-Hidroxiprogesterona/farmacologia , Membranas Extraembrionárias/citologia , Membranas Extraembrionárias/efeitos dos fármacos , Humanos , Peróxido de Hidrogênio/farmacologia , Medrogestona/farmacologia , Cultura Primária de Células , Progesterona/farmacologia , RNA Mensageiro/metabolismo , Receptores de Glucocorticoides/metabolismo , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA