Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 24(7)2023 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-37047162

RESUMO

Preeclampsia is associated with an increased lifelong risk of cardiovascular disease (CVD). It is not clear whether this is induced by persistent systemic organ and vascular damage following preeclampsia or due to a predisposition to both conditions that share cardiovascular pathophysiology. Common to both CVD and preeclampsia is the dysregulation of corin and its proteolytic product, atrial natriuretic peptide (ANP). ANP, a hypotensive hormone converted from pro-ANP by corin, is involved in blood pressure homeostasis. While corin is predominantly a cardiac enzyme, both corin and pro-ANP are significantly upregulated in the gravid uterus and dysregulated in preeclampsia. Relatively little is known about ANP function in the endothelium during a pregnancy complicated by preeclampsia. Here, we investigated the effect of ANP on endothelial cell proliferation and migration, markers of endothelial dysfunction, and receptor expression in omental arteries exposed to circulating preeclamptic toxins. ANP receptor expression is significantly upregulated in preeclamptic vasculature but not because of exposure to preeclampsia toxins tumour necrosis factor α or soluble fms-like tyrosine kinase-1. The supplementation of endothelial cells with ANP did not promote proliferation or migration, nor did ANP improve markers of endothelial dysfunction. The role of ANP in preeclampsia is unlikely to be via endothelial pathways.


Assuntos
Doenças Cardiovasculares , Pré-Eclâmpsia , Gravidez , Feminino , Humanos , Fator Natriurético Atrial/metabolismo , Células Endoteliais/metabolismo , Endotélio/metabolismo
2.
Placenta ; 132: 20-26, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36623415

RESUMO

INTRODUCTION: Development of a therapeutic that targets the pathophysiological elements of preeclampsia would be a major advance for obstetrics, with potential to save the lives of countless mothers and babies. We recently identified anti-inflammatory drug sulfasalazine as a prospective candidate therapeutic for treatment of preeclampsia. In primary human cells and tissues in vitro, sulfasalazine potently decreased secretion of anti-angiogenic sFlt-1 and sENG, increased production of pro-angiogenic PlGF, mitigated endothelial dysfunction, and promoted whole vessel vasodilation. METHODS: Using nitric oxide synthase antagonist Nω-Nitro-l-arginine methyl ester hydrochloride, a preeclampsia-like phenotype was induced in pregnant mice, including high blood pressure, fetal growth restriction, and elevated circulating sFlt-1. Mice were treated with sulfasalazine or vehicle from gestational day (D)13.5, with blood pressure measurements across gestation, fetal measurements at D17.5, and wire myograph assessment of vasoactivity. RESULTS: Sulfasalazine had a modest effect on blood pressure, decreasing diastolic and mean blood pressure on D13.5, but not later in gestation, or systolic blood pressure. Sulfasalazine was not able to rescue fetal growth, in male or female fetuses. There was a suggestion of improved vasoactivity with sulfasalazine, but further clarification is required. DISCUSSION: In this mouse model of preeclampsia, sulfasalazine did not sustain reductions in blood pressure nor affect fetal parameters of size and weight, both desirable attributes of a viable preeclampsia therapeutic. While these data suggest sulfasalazine might improve vasoactivity, murine toxicity considerations limited the dose range of sulfasalazine that could be tested in the current study.


Assuntos
Hipertensão , Pré-Eclâmpsia , Gravidez , Feminino , Masculino , Camundongos , Animais , Humanos , Pré-Eclâmpsia/tratamento farmacológico , Sulfassalazina/farmacologia , Sulfassalazina/uso terapêutico , Pressão Sanguínea , Modelos Animais de Doenças , Óxido Nítrico Sintase/farmacologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular , Óxido Nítrico/farmacologia
3.
Reproduction ; 165(1): R9-R23, 2023 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-36305709

RESUMO

In brief: Preterm birth is the leading cause of perinatal morbidity and mortality; however, current therapies offer limited efficacy to delay birth and improve neonatal outcomes. This review explores the potential of repurposing drugs with known safety profiles to quench uterine contractions and inflammation, identifying promising agents for clinical trials. Abstract: Preterm birth is the leading cause of neonatal morbidity and mortality globally. Despite extensive research into the underlying pathophysiology, rates of preterm birth have not significantly reduced. Currently, preterm labour management is based on optimising neonatal outcomes. Treatment involves administering drugs (tocolytics) to suppress uterine contractions to allow sufficient time for transfer to an appropriate facility and administration of antenatal corticosteroids for fetal lung maturation. Current tocolytics are limited as they are associated with adverse maternal and fetal effects and only delay delivery for a short period. There has been a serious lack of therapeutic development for preterm birth, and new approaches to protect against or delay preterm birth are urgently needed. Repurposing drugs for the prevention of preterm birth presents as a promising approach by reducing the time and costs associated with pharmaceutical drug development. In this review, we explore the evidence for the potential of therapies, specifically proton pump inhibitors, tumour necrosis factor inhibitors, prostaglandin receptor antagonists, aspirin, and statins, to be repurposed as preventatives and/or treatments for preterm birth. Importantly, many of these innovative approaches being explored have good safety profiles in pregnancy. We also review how delivery of these drugs can be enhanced, either through targeted delivery systems or via combination therapy approaches. We aim to present innovative strategies capable of targeting multiple aspects of the complex pathophysiology that underlie preterm birth. There is an urgent unmet need for preterm birth therapeutic development, and these strategies hold great promise for improving neonatal outcomes.


Assuntos
Trabalho de Parto Prematuro , Nascimento Prematuro , Tocolíticos , Recém-Nascido , Feminino , Gravidez , Humanos , Nascimento Prematuro/prevenção & controle , Tocolíticos/uso terapêutico , Preparações Farmacêuticas , Reposicionamento de Medicamentos , Trabalho de Parto Prematuro/tratamento farmacológico , Trabalho de Parto Prematuro/prevenção & controle
4.
Life Sci Alliance ; 5(12)2022 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-36260752

RESUMO

Preeclampsia affects ∼2-8% of pregnancies worldwide. It is associated with increased long-term maternal cardiovascular disease risk. This study assesses the effect of the vasoconstrictor N(ω)-nitro-L-arginine methyl ester (L-NAME) in modelling preeclampsia in mice, and its long-term effects on maternal cardiovascular health. In this study, we found that L-NAME administration mimicked key characteristics of preeclampsia, including elevated blood pressure, impaired fetal and placental growth, and increased circulating endothelin-1 (vasoconstrictor), soluble fms-like tyrosine kinase-1 (anti-angiogenic factor), and C-reactive protein (inflammatory marker). Post-delivery, mice that received L-NAME in pregnancy recovered, with no discernible changes in measured cardiovascular indices at 1-, 2-, and 4-wk post-delivery, compared with matched controls. At 10-wk post-delivery, arteries collected from the L-NAME mice constricted significantly more to phenylephrine than controls. In addition, these mice had increased kidney Mmp9:Timp1 and heart Tnf mRNA expression, indicating increased inflammation. These findings suggest that though administration of L-NAME in mice certainly models key characteristics of preeclampsia during pregnancy, it does not appear to model the adverse increase in cardiovascular disease risk seen in individuals after preeclampsia.


Assuntos
Doenças Cardiovasculares , Pré-Eclâmpsia , Animais , Feminino , Camundongos , Gravidez , Proteína C-Reativa/metabolismo , Doenças Cardiovasculares/etiologia , Doenças Cardiovasculares/metabolismo , Modelos Animais de Doenças , Endotelina-1/genética , Endotelina-1/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , NG-Nitroarginina Metil Éster/farmacologia , NG-Nitroarginina Metil Éster/metabolismo , Fenilefrina/metabolismo , Placenta/metabolismo , Pré-Eclâmpsia/metabolismo , RNA Mensageiro/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Vasoconstritores/metabolismo
5.
Int J Mol Sci ; 23(17)2022 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-36076929

RESUMO

Previously, we demonstrated that the proton pump inhibitor, esomeprazole magnesium hydrate (MH), could have potential as a repurposed treatment against preeclampsia, a serious obstetric condition. In this study we investigate the difference in the preclinical effectiveness between 100 µM of esomeprazole MH and its hydration isomer, esomeprazole magnesium trihydrate (MTH). Here, we found that both treatments reduced secretion of sFLT-1 (anti-angiogenic factor) from primary cytotrophoblast, but only esomeprazole MH reduced sFLT-1 secretion from primary human umbilical vein endothelial cells (assessed via ELISA). Both drugs could mitigate expression of the endothelial dysfunction markers, vascular cell adhesion molecule-1 and endothelin-1 (via qPCR). Neither esomeprazole MH nor MTH quenched cytotrophoblast reactive oxygen species production in response to sodium azide (ROS assay). Finally, using wire myography, we demonstrated that both compounds were able to induce vasodilation of human omental arteries at 100 µM. Esomeprazole is safe to use in pregnancy and a candidate treatment for preeclampsia. Using primary human tissues and cells, we validated that esomeprazole is effective in enhancing vascular relaxation, and can reduce key factors associated with preeclampsia, including sFLT-1 and endothelial dysfunction. However, esomeprazole MH was more efficacious than esomeprazole MTH in our in vitro studies.


Assuntos
Pré-Eclâmpsia , Biomarcadores/metabolismo , Esomeprazol/metabolismo , Esomeprazol/farmacologia , Esomeprazol/uso terapêutico , Feminino , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Hidróxido de Magnésio , Placenta/metabolismo , Pré-Eclâmpsia/metabolismo , Gravidez , Inibidores da Bomba de Prótons/farmacologia , Inibidores da Bomba de Prótons/uso terapêutico , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo
6.
Pregnancy Hypertens ; 30: 13-20, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35933758

RESUMO

BACKGROUND: Dishevelled Associated Activator Of Morphogenesis 2 (DAAM2) levels are elevated in the maternal circulation and placenta in pregnancies complicated by fetal growth restriction. However, placental DAAM2 levels in cases of preeclampsia have not previously been explored. Here, we examined placental DAAM2 in pregnancies complicated by preterm preeclampsia, and whether candidate preeclampsia therapeutics altered its expression. METHODS: DAAM2 mRNA and protein levels were assessed in placental tissue from cases of preterm preeclampsia and gestation-matched controls (delivering ≤ 34 weeks; qPCR and western blot respectively). Short interfering RNAs were used to silence DAAM2 in isolated primary cytotrophoblast under normoxic (8 % O2) and hypoxic (1 % O2) conditions, and expression of anti-angiogenic sFLT-1, angiogenic PGF, antioxidant, fetal growth, and inflammatory genes assessed. DAAM2 expression was measured in placental explant tissue from pregnancies complicated by preeclampsia, treated with three proton pump inhibitors (100 µM esomeprazole, lansoprazole, and rabeprazole). RESULTS: DAAM2 expression was significantly reduced in preeclamptic placental tissue compared to controls, but protein production was unchanged. Silencing DAAM2 in hypoxic cytotrophoblast increased sFLT-i13 isoform expression, but did not alter sFLT-e15a or PGF expression, or sFLT-1 secretion. DAAM2 knockdown did not alter expression of antioxidant (NQO-1, TXN, GCLC), fetal growth (SPINT1), or inflammasome (NLRP3) genes. Esomeprazole and lansoprazole, but not rabeprazole, increased DAAM2 expression in placental explant tissue from cases of preeclampsia. CONCLUSION: Placental DAAM2 protein is not significantly altered in placental tissue in cases of preeclampsia, and its suppression does not alter sFLT-1 secretion. Hence, placental DAAM2 is unlikely to drive the pathogenesis associated with preeclampsia.


Assuntos
Proteínas dos Microfilamentos , Pré-Eclâmpsia , Proteínas da Gravidez , Inibidores da Bomba de Prótons , Proteínas rho de Ligação ao GTP , Feminino , Humanos , Recém-Nascido , Gravidez , Antioxidantes/metabolismo , Esomeprazol/uso terapêutico , Hipóxia/metabolismo , Lansoprazol/uso terapêutico , Placenta/metabolismo , Pré-Eclâmpsia/genética , Prostaglandinas F/metabolismo , Inibidores da Bomba de Prótons/uso terapêutico , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Proteínas dos Microfilamentos/metabolismo
7.
Sci Rep ; 11(1): 20670, 2021 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-34667209

RESUMO

Nuclear Receptor Subfamily 4 Group A Member 2 (NR4A2) transcripts are elevated in the circulation of individuals whose pregnancies are complicated by preterm fetal growth restriction (FGR). In this paper, we show that the cases with preeclampsia (PE) have increased circulating NR4A2 transcripts compared to those with normotensive FGR. We aimed to establish whether the dysfunctional placenta mirrors the increase in NR4A2 transcripts and further, to uncover the function of placental NR4A2. NR4A2 expression was detected in preterm and term placental tissue; expressed higher at term. NR4A2 mRNA expression and protein were not altered in placentas from preterm FGR or PE pregnancies. Hypoxia (1% O2 compared to 8% O2) significantly reduced cytotrophoblast NR4A2 mRNA expression, but not placental explant NR4A2 expression. Silencing cytotrophoblast NR4A2 expression under hypoxia (via short interfering (si)RNAs) did not alter angiogenic Placental Growth Factor, nor anti-angiogenic sFlt-1 mRNA expression or protein secretion, but increased expression of cellular antioxidant, oxidative stress, inflammatory, and growth genes. NR4A2 expression was also not altered in a model of tumour necrosis factor-α-induced endothelial dysfunction, or with pravastatin treatment. Further studies are required to identify the origin of the circulating transcripts in pathological pregnancies, and investigate the function of placental NR4A2.


Assuntos
Retardo do Crescimento Fetal/metabolismo , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Fator de Crescimento Placentário/metabolismo , Placenta/metabolismo , Pré-Eclâmpsia/metabolismo , Trofoblastos/metabolismo , Adulto , Antioxidantes/metabolismo , Feminino , Humanos , Hipóxia/metabolismo , Inflamação/metabolismo , Masculino , Estresse Oxidativo/fisiologia , Doenças Placentárias/metabolismo , Gravidez , RNA Mensageiro/metabolismo
8.
Placenta ; 102: 10-16, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32980138

RESUMO

Preeclampsia is a complex disease affecting 2-8% of pregnancies worldwide. It poses significant risk of maternal and perinatal morbidity and mortality. Despite the rising research interest to discover new therapeutic approaches to prevent and treat preeclampsia, options remain limited. Identifying the important pathological stages in the progression of this disease allows us to evaluate effective candidate therapeutics. Three important stages in the pathophysiology are: 1) placental hypoxia and oxidative stress, 2) excess release of anti-angiogenic and pro-inflammatory factors, and 3) widespread systemic endothelial dysfunction and vasoconstriction. Repurposing drugs already safe for use in pregnancy is an attractive option for discovery of novel therapeutics. There are many drugs currently being assessed to treat preeclampsia, including proton pump inhibitors (PPIs), metformin, statins, sulfasalazine, sofalcone, resveratrol, melatonin, and sildenafil citrate. These drugs show positive effects in preclinical studies, targeting placental and endothelial dysfunction. However, using novel therapeutics can raise safety concerns for the developing fetus. Therefore, innovative targeted delivery systems are being developed to safely administer these therapeutics directly to the placenta and/or endothelium. These include nanoparticle delivery systems, developed and used by the oncology field, now being adapted for obstetrics. This technology is currently being assessed in animal models and shows promise for treating preeclampsia. Combining effective therapeutics with targeted drug delivery could be the future of preeclampsia treatment.


Assuntos
Terapia de Alvo Molecular , Pré-Eclâmpsia/tratamento farmacológico , Animais , Aspirina/uso terapêutico , Inibidores de Ciclo-Oxigenase/uso terapêutico , Sistemas de Liberação de Medicamentos , Feminino , Humanos , Pré-Eclâmpsia/etiologia , Pré-Eclâmpsia/prevenção & controle , Gravidez
9.
Pregnancy Hypertens ; 22: 86-92, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32758704

RESUMO

Development and repurposing of therapies that show promise in the prevention or treatment of preeclampsia would be a major advance for the obstetrics field. We recently identified esomeprazole and sulfasalazine as potential candidates for the treatment of preeclampsia. Both reduce placental and endothelial secretion of sFlt-1 and sENG and mitigate endothelial dysfunction in vitro. Here we assessed whether esomeprazole and sulfasalazine in combination would additively attenuate the elevated release of anti-angiogenic factors and markers of endothelial dysfunction, key characteristics of preeclampsia. Primary placental tissue and cells, and primary endothelial cells were treated with esomeprazole and sulfasalazine alone and in combination. We assessed secretion of sFlt-1 and sENG and performed in vitro assays of endothelial dysfunction. Combining esomeprazole and sulfasalazine in lower concentrations caused an additive reduction in sFlt-1 secretion in primary cytotrophoblasts, placental explants and endothelial cells. No additive reduction was observed in sENG secretion when esomeprazole and sulfasalazine were combined. Together, esomeprazole and sulfasalazine additively reduced TNF-α-induced VCAM and ET-1 mRNA expression, and monocyte adhesion to endothelial cells. In conclusion, combining esomeprazole and sulfasalazine additively reduced secretion of sFlt-1 and markers of endothelial dysfunction. Combined administration of esomeprazole and sulfasalazine may provide a more effective treatment or prevention for preeclampsia compared to either as single agents.


Assuntos
Esomeprazol/farmacologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Pré-Eclâmpsia/metabolismo , Sulfassalazina/farmacologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/efeitos dos fármacos , Estudos de Casos e Controles , Quimioterapia Combinada , Feminino , Humanos , Placenta/efeitos dos fármacos , Gravidez
10.
Pregnancy Hypertens ; 20: 83-91, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32199147

RESUMO

OBJECTIVES: There is avid interest in pravastatin as a therapeutic intervention for pre-eclampsia, however little is known on statin action on endothelial dysfunction. This study aimed to evaluate the ability of pravastatin, simvastatin and rosuvastatin to reduce pre-eclampsia-associated markers of endothelial dysfunction in human endothelial cells. STUDY DESIGN: Primary human umbilical vein endothelial cells (HUVECs) and uterine microvascular cells (UtMVs) were isolated and treated with 0.2, 2, 20 and 200 µM pravastatin, simvastatin and rosuvastatin for 24 h, either with or without pre-treatment with TNF-α to induce endothelial dysfunction. MAIN OUTCOME MEASURES: Cell viability (MTS) assays were performed and cells were visually inspected. Expression of endothelial dysfunction markers, endothelin-1 (ET-1) and vascular cell adhesion molecule-1 (VCAM-1) were assessed by qPCR (n=3). Intracellular VCAM-1 protein was examined by Western Blotting (n=5). ET-1 and soluble fms-like tyrosine kinase-1 (sFLT-1) protein secretion was assessed by ELISA in HUVEC conditioned media (n=3). RESULTS: High doses of simvastatin and rosuvastatin significantly compromised HUVEC survival. 200 µM simvastatin significantly reduced UtMV survival. Abnormal cell structure was observed with these doses and thus were excluded from further analysis. The statins did not mitigate TNF-α induced ET-1 or VCAM-1 expression in either HUVECs or UtMVs, nor VCAM-1 protein expression in HUVECs. 0.2 µM pravastatin and simvastatin significantly reduced ET-1 and sFLT-1 protein secretion. CONCLUSIONS: Pravastatin significantly reduced secretion of both ET-1 and sFLT-1, key mediators of endothelial dysfunction. Importantly, pravastatin had no toxic effects, in contrast to rosuvastatin and simvastatin. This further supports selection of pravastatin for clinical applications to combat pre-eclampsia.


Assuntos
Endotélio Vascular/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Pravastatina/farmacologia , Pré-Eclâmpsia/tratamento farmacológico , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Endotelina-1/genética , Endotelina-1/metabolismo , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiopatologia , Feminino , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Pravastatina/toxicidade , Pré-Eclâmpsia/genética , Pré-Eclâmpsia/metabolismo , Pré-Eclâmpsia/fisiopatologia , Gravidez , Rosuvastatina Cálcica/farmacologia , Sinvastatina/farmacologia , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo
11.
Pregnancy Hypertens ; 14: 115-120, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30527097

RESUMO

INTRODUCTION: Preeclampsia is a severe complication of pregnancy, and likely arises from abnormal placental development in early pregnancy. Persistent placental hypoxia is thought to trigger the release of anti-angiogenic factors into the maternal circulation leading to widespread endothelial dysfunction. Epidermal growth factor-like domain 7 (EGFL7) is a secreted angiogenic factor that may play a key role in the disrupted angiogenesis seen in response to placental hypoxia that characterizes preeclampsia. METHODS: Primary trophoblasts were isolated and cultured in both normoxic and hypoxic conditions. Under hypoxia HIF1α was silenced and EGFL7 mRNA expression was assessed. EGFL7 mRNA expression was measured in placentas obtained from women with early (<34 weeks) and late onset preeclampsia; and in peripheral whole blood maternal samples from women with preeclampsia and gestation matched controls. EGFL7 plasma levels were assessed in plasma from women with preeclampsia, compared to gestation-matched controls. RESULTS: EGFL7 expression was significantly upregulated in primary human trophoblasts cultured in hypoxia (>2-fold, p < 0.0001), however this was not regulated via a HIF1α dependent manner. EGFL7 mRNA expression was not altered in placenta from women with early or late onset preeclampsia. Circulating EGFL7 protein levels were not different in women with severe preeclampsia. In contrast, EGFL7 mRNA expression was increased in maternal blood in women with early onset preeclampsia (∼1.6-fold, p < 0.05). DISCUSSION: EGFL7 mRNA expression is increased with hypoxia in human trophoblast and is increased in the maternal circulation in women with preeclampsia. Further studies aimed at understanding the role and regulation of EGLF7 in the pathophysiology of preeclampsia are required.


Assuntos
Fatores de Crescimento Endotelial/metabolismo , Pré-Eclâmpsia/metabolismo , Trofoblastos/metabolismo , Adulto , Proteínas de Ligação ao Cálcio , Estudos de Casos e Controles , Família de Proteínas EGF , Fatores de Crescimento Endotelial/sangue , Feminino , Expressão Gênica , Humanos , Hipóxia/metabolismo , Placenta/metabolismo , Pré-Eclâmpsia/sangue , Gravidez , Adulto Jovem
12.
PLoS One ; 13(4): e0187082, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29641523

RESUMO

Preeclampsia is one of the most serious complications of pregnancy. Currently there are no medical treatments. Given placental oxidative stress may be an early trigger in the pathogenesis of preeclampsia, therapies that enhance antioxidant pathways have been proposed as treatments. Melatonin is a direct free-radical scavenger and indirect antioxidant. We performed in vitro assays to assess whether melatonin 1) enhances the antioxidant response element genes (heme-oxygenase 1, (HO-1), glutamate-cysteine ligase (GCLC), NAD(P)H:quinone acceptor oxidoreductase 1 (NQO1), thioredoxin (TXN)) or 2) alters secretion of the anti-angiogenic factors soluble fms-like tyrosine kinase-1 (sFLT) or soluble endoglin (sENG) from human primary trophoblasts, placental explants and human umbilical vein endothelial cells (HUVECs) and 3) can rescue TNF-α induced endothelial dysfunction. In primary trophoblast melatonin treatment increased expression of the antioxidant enzyme TXN. Expression of TXN, GCLC and NQO1 was upregulated in placental tissue with melatonin treatment. HUVECs treated with melatonin showed an increase in both TXN and GCLC. Melatonin did not increase HO-1 expression in any of the tissues examined. Melatonin reduced sFLT secretion from primary trophoblasts, but had no effect on sFLT or sENG secretion from placental explants or HUVECs. Melatonin did not rescue TNF-α induced VCAM-1 and ET-1 expression in endothelial cells. Our findings suggest that melatonin induces antioxidant pathways in placenta and endothelial cells. Furthermore, it may have effects in reducing sFLT secretion from trophoblast, but does not reduce endothelial dysfunction. Given it is likely to be safe in pregnancy, it may have potential as a therapeutic agent to treat or prevent preeclampsia.


Assuntos
Antioxidantes/metabolismo , Melatonina/farmacologia , Placenta/metabolismo , Pré-Eclâmpsia/tratamento farmacológico , Trofoblastos/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Melatonina/uso terapêutico , Placenta/efeitos dos fármacos , Pré-Eclâmpsia/patologia , Gravidez , Solubilidade , Trofoblastos/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/química
13.
PLoS One ; 13(2): e0188845, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29466360

RESUMO

INTRODUCTION: The discovery of new treatments that prevent or treat preeclampsia would be a major advance. Antiangiogenic factors soluble fms-like tyrosine kinase-1 (sFlt-1) and soluble endoglin (sENG) are secreted in excess from the placenta, causing hypertension, endothelial dysfunction, and multiorgan injury. We recently identified metformin and esomeprazole as potential treatments for preeclampsia. Both reduce placental and endothelial secretion of sFlt-1 and soluble endoglin, and reduce endothelial dysfunction. OBJECTIVES: We set out to assess whether combining metformin and esomeprazole would additively reduce sFlt-1 and soluble endoglin secretion and reduce endothelial dysfunction (verses drug alone). Metformin and esomeprazole were added to primary placental cells and tissues, and endothelial cells and their effects on sFlt-1 and soluble endoglin secretion were assessed in vitro. Tumor necrosis factor-α (TNF-α) was added to endothelial cells to induce dysfunction in vitro. We examined the ability of metformin + esomeprazole to rescue TNF-α induced vascular cell adhesion molecule-1 (VCAM-1) and Endothelin-1 (ET-1) expression, leukocyte adhesion (markers of endothelial dysfunction). RESULTS: Combining metformin and esomeprazole was additive at reducing sFlt-1 secretion and expression of sFlt-1 e15a mRNA isoform in primary cytotrophoblast, placental explants and endothelial cells. In contrast, no additive reduction in sENG was observed with combined metformin and esomeprazole. The low-dose combination of metformin + esomeprazole additively reduced TNF-α-induced VCAM-1 mRNA, but not VCAM-1 protein expression. There was no additive reduction when combining metformin and esomeprazole on TNF-α induced PBMC adhesion to endothelial cells. However, combining metformin and esomeprazole additively reduced ET-1 mRNA expression. CONCLUSIONS: In conclusion combining metformin and esomeprazole additively reduced secretion of sFlt-1, and markers of endothelial dysfunction. The combination of metformin and esomeprazole may provide a more effective treatment or prevention for preeclampsia compared to either as single agents.


Assuntos
Endotélio Vascular/efeitos dos fármacos , Esomeprazol/administração & dosagem , Metformina/administração & dosagem , Pré-Eclâmpsia/tratamento farmacológico , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Endotélio Vascular/fisiopatologia , Feminino , Células Endoteliais da Veia Umbilical Humana , Humanos , Gravidez
14.
PLoS One ; 11(10): e0163096, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27711226

RESUMO

Embryo implantation requires synchronized dialogue between the receptive endometrium and activated blastocyst via locally produced soluble mediators. During the mid-secretory (MS) phase of the menstrual cycle, increased glandular secretion into the uterine lumen provides important mediators that modulate the endometrium and support the conceptus during implantation. Previously we demonstrated the importance of vascular endothelial growth factor (VEGF) in the human uterus, particularly with respect to embryo implantation. In the current study, proteomic analysis of human uterine lavage fluid identified the presence of placental growth factor (PlGF) a homolog of VEGF, that binds the VEGF receptor 1 (VEGFR1). Analysis of immunostaining for PlGF in human endometrial tissue across the menstrual cycle (from both fertile and infertile women) revealed PlGF was predominantly localised to glandular and luminal epithelial cells, with staining in the decidualising stromal cells surrounding the maternal spiral arteries in the secretory phase of the menstrual cycle. Immunoreactive PlGF was also detected in subpopulations of endometrial leukocytes. Functional studies demonstrated that culturing mouse embryos with recombinant human (rh)PlGF enhanced blastocyst cell number and outgrowth. Furthermore, treatment of human endometrial epithelial cells (EEC) with rhPlGF enhanced EEC adhesion. Taken together, these data demonstrate that PlGF is abundant in the human endometrium, and secreted into the uterine lumen where it mediates functional changes in cellular adhesion with important roles in implantation.


Assuntos
Implantação do Embrião , Endométrio/metabolismo , Fator de Crescimento Placentário/metabolismo , Animais , Blastocisto/citologia , Blastocisto/efeitos dos fármacos , Blastocisto/fisiologia , Adesão Celular/efeitos dos fármacos , Contagem de Células , Endométrio/citologia , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Feminino , Humanos , Camundongos , Fator de Crescimento Placentário/farmacologia , Transporte Proteico , Útero/metabolismo
15.
Hypertension ; 66(5): 1073-81, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26324507

RESUMO

Elevated placental release of the antiangiogenic factors, soluble fms-like tyrosine kinase-1 (sFlt-1) and soluble endoglin (sENG), is central to the pathophysiology of preeclampsia. It is widely accepted that heme oxygenase-1 (HO-1) is decreased in preeclamptic placenta and negatively regulates sFlt-1 and sENG production. We set out to verify these contentions. There was no difference in HO-1 mRNA or protein levels in preterm preeclamptic placentas (n=17) compared with gestationally matched controls (n=27). In silico analysis of microarray studies did not identify decreased placental HO-1 expression in preeclamptic placenta. Silencing HO-1 in primary trophoblasts did not affect sFlt-1 protein secretion after 24 or 48 hours. Silencing nuclear factor (erythroid-derived 2)-like 2 (transcription factor that upregulates HO-1) in trophoblasts also did not affect sFlt-1 secretion. Administering tin protoporphyrin IX dichloride (HO-1 inhibitor) or cobalt protoporphyrin (HO-1 inducer) into placental explants did not affect sFlt-1 or sENG secretion. Silencing HO-1 in 2 types of primary endothelial cells (human umbilical vein endothelial and uterine microvascular endothelial cells) significantly increased sFlt-1 secretion but not sENG secretion. However, HO-1 silencing selectively increased mRNA expression of sFlt-1 i13 (generically expressed sFlt-1 variant) but not of sFlt-1 e15a (sFlt-1 variant mainly expressed in placenta). Furthermore, adding tin protoporphyrin IX dichloride decreased sFlt-1, whereas adding HO-1 inducers (cobalt protoporphyrin, dimethyl fumarate, and rosiglitazone) either had no effect or increased sFlt-1 or sENG secretion (these trends are opposite to what is expected). We conclude that HO-1 expression is not decreased in preeclamptic placenta and HO-1 does not negatively regulate placental sFlt-1 and sENG secretion in placental or endothelial cells.


Assuntos
Antígenos CD/metabolismo , Heme Oxigenase-1/metabolismo , Placenta/metabolismo , Pré-Eclâmpsia/metabolismo , Receptores de Superfície Celular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Estudos de Casos e Controles , Células Cultivadas , Endoglina , Endotélio/citologia , Endotélio/metabolismo , Feminino , Idade Gestacional , Humanos , Gravidez , RNA Mensageiro/metabolismo , Técnicas de Cultura de Tecidos , Trofoblastos/citologia , Trofoblastos/metabolismo
16.
Hypertension ; 66(3): 687-97; discussion 445, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26222708

RESUMO

UNLABELLED: Preeclampsia is a major pregnancy complication where excess placental release of soluble fms-like tyrosine kinase-1 (sFlt-1) and soluble endoglin causes maternal endothelial and multisystem organ injury. Clinical trials have commenced examining whether pravastatin can be used to treat preeclampsia. However, the preclinical evidence supporting pravastatin as a treatment is limited to animal models, with almost no studies in human tissues. Therefore, we examined whether pravastatin reduced sFlt-1 and soluble endoglin secretion and decreased endothelial dysfunction in primary human tissues. Pravastatin reduced sFlt-1 secretion from primary endothelial cells, purified cytotrophoblast cells, and placental explants obtained from women with preterm preeclampsia. It increased soluble endoglin secretion from endothelial cells but did not change secretion from placental explants. The regulation of sFlt-1 by pravastatin seemed to be mediated via the 3-hydroxy-3-methylglutaryl-coenzyme A reductase cholesterol synthesis pathway. Pravastatin also reduced markers of endothelial dysfunction, including vascular cell adhesion molecule-1 expression and leukocyte adhesion on endothelial cells and increased endothelial cell migration and invasion. We also treated 4 patients with preterm preeclampsia presenting at <30 weeks of gestation with daily pravastatin. Pravastatin seemed to stabilize blood pressure, proteinuria, and serum uric acid levels. Furthermore, serum sFlt-1 levels decreased. We collected the placentas at delivery and found that pravastatin reduced sFlt-1 secretion. These results indicate that pravastatin reduced sFlt-1 and soluble endoglin production and decreased endothelial dysfunction in primary human tissues. We also present pilot data, suggesting that pravastatin can stabilize clinical and biochemical features of preterm preeclampsia. Our data obtained in human tissues support the concept that pravastatin is a candidate therapeutic for preeclampsia. CLINICAL TRIAL REGISTRATION: URL: http://www.anzctr.org.au. Unique identifier: ACTRN12613000268741.


Assuntos
Antígenos CD/metabolismo , Endotélio Vascular/efeitos dos fármacos , Placenta/efeitos dos fármacos , Pravastatina/farmacologia , Pré-Eclâmpsia/tratamento farmacológico , Receptores de Superfície Celular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Adulto , Molécula 1 de Adesão Celular , Moléculas de Adesão Celular/metabolismo , Endoglina , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Feminino , Humanos , Imunoglobulinas/metabolismo , Placenta/metabolismo , Pravastatina/uso terapêutico , Pré-Eclâmpsia/metabolismo , Gravidez , Transdução de Sinais/efeitos dos fármacos , Resultado do Tratamento , Trofoblastos/efeitos dos fármacos , Trofoblastos/metabolismo , Adulto Jovem
17.
Mol Hum Reprod ; 21(5): 424-34, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25731709

RESUMO

The rate of obesity among men of reproductive age has tripled in the last three decades. Previously, we demonstrated that paternal obesity resulted in impaired preimplantation developmental kinetics, compromised post-compaction metabolism and decreased blastocyst cell number when embryos were generated in vivo. Subsequently, using in vitro fertilization we found embryos of obese males to have altered metabolism before compaction, reduced inner cell mass cell number and retarded fetal development--the difference between these two studies being the method of embryo generation and the presence or absence of seminal plasma, respectively. Here, we hypothesize that both sperm and seminal plasma are affected by obesity, compromising embryogenesis and pregnancy health in a cumulative manner. Epididymal sperm and seminal vesicle fluid were collected from normal and obese C57BL/6 mice. RNA and DNA were extracted from spermatozoa for qPCR and global methylation analysis, respectively. Proteomic (Luminex) and metabolomic (GC-MS) techniques were employed to analyse the composition of seminal vesicle fluid. Nuclear encoded cytochrome c oxidase subunit IV isoform 1 (Cox4i1) of the terminal enzyme in the mitochondrial respiratory chain demonstrated significantly increased RNA levels in the sperm of obese males (P< 0.05). Quantitative seminal plasma analysis identified significant changes in levels of the hormones insulin, leptin and estradiol between normal and obese males (P < 0.05). Further, the metabolite composition of seminal vesicle fluid was significantly affected by obesity. Consequently, this study has determined that obesity affects both sperm and seminal plasma composition. The interaction between sperm and seminal plasma warrants further analysis.


Assuntos
Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Infertilidade Masculina/metabolismo , Obesidade/fisiopatologia , RNA Mensageiro/metabolismo , Proteínas de Plasma Seminal/metabolismo , Espermatozoides/metabolismo , Animais , Dieta Hiperlipídica/efeitos adversos , Complexo IV da Cadeia de Transporte de Elétrons/química , Complexo IV da Cadeia de Transporte de Elétrons/genética , Estradiol/sangue , Estradiol/metabolismo , Infertilidade Masculina/sangue , Infertilidade Masculina/etiologia , Insulina/sangue , Insulina/metabolismo , Leptina/sangue , Leptina/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Obesidade/etiologia , Subunidades Proteicas/química , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Distribuição Aleatória , Sêmen/metabolismo , Proteínas de Plasma Seminal/genética , Contagem de Espermatozoides , Motilidade dos Espermatozoides , Espermatozoides/enzimologia , Regulação para Cima
18.
Reproduction ; 149(5): 435-44, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25725082

RESUMO

Fetal growth restriction (FGR) is a major obstetric complication stemming from poor placental development. We have previously demonstrated that paternal obesity in mice is associated with impaired embryo development and significantly reduced fetal and placental weights. We hypothesised that the FGR observed in our rodent model of paternal diet-induced obesity is associated with alterations in metabolic, cell signalling and stress pathways. Male C57BL/6 mice were fed either a normal or high-fat diet for 10 weeks before sperm collection for IVF and subsequent embryo transfer. On embryonic day 14, placentas were collected and RNA extracted from both male and female placentas to assess mRNA expression of 24 target genes using custom RT-qPCR arrays. Peroxisome proliferator-activated receptor alpha (Ppara) and caspase-12 (Casp12) expression were significantly altered in male placentas from obese fathers compared with normal (P<0.05), but not female placentas. PPARA and CASP12 proteins were localised within the placenta to trophoblast giant cells by immunohistochemistry, and relative protein abundance was determined by western blot analysis. DNA was also extracted from the same placentas to determine methylation status. Global DNA methylation was significantly increased in female placentas from obese fathers compared with normal (P<0.05), but not male placentas. In this study, we demonstrate for the first time that paternal obesity is associated with changes in gene expression and methylation status of extraembryonic tissue in a sex-specific manner. These findings reinforce the negative consequences of paternal obesity before conception, and emphasise the need for more lifestyle advice for prospective fathers.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Pai , Retardo do Crescimento Fetal/metabolismo , Obesidade/complicações , Placenta/metabolismo , Placentação/fisiologia , Animais , Blastocisto/citologia , Blastocisto/metabolismo , Western Blotting , Caspase 12/genética , Caspase 12/metabolismo , Células Cultivadas , Metilação de DNA , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Feminino , Fertilização in vitro , Retardo do Crescimento Fetal/etiologia , Retardo do Crescimento Fetal/patologia , Viabilidade Fetal , Técnicas Imunoenzimáticas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/fisiopatologia , PPAR alfa/genética , PPAR alfa/metabolismo , Placenta/citologia , Gravidez , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Aumento de Peso
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA