Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Nutrients ; 14(16)2022 Aug 11.
Article in English | MEDLINE | ID: mdl-36014792

ABSTRACT

Gestational diabetes mellitus (GDM) and maternal obesity are significant metabolic complications increasingly prevalent in pregnancy. Of major concern, both GDM and maternal obesity can have long-term detrimental impacts on the health of both mother and offspring. Recent research has shown that increased inflammation and oxidative stress are two features central to the pathophysiology of these metabolic conditions. Evidence suggests selenium supplementation may be linked to disease prevention in pregnancy; however, the specific effects of selenium on inflammation and oxidative stress associated with GDM and maternal obesity are unknown. Therefore, this study aimed to investigate the effect of selenium supplementation on an in vitro model of GDM and maternal obesity. Human placental tissue, visceral adipose tissue (VAT) and subcutaneous adipose tissue (SAT) were stimulated with either the bacterial product lipopolysaccharide (LPS) or the pro-inflammatory cytokine TNF-α. Selenium pre-treatment blocked LPS and TNF-α induced mRNA expression and secretion of pro-inflammatory cytokines and chemokines, while increasing anti-inflammatory cytokine and antioxidant mRNA expression in placenta, VAT and SAT. Selenium pre-treatment was also found to inhibit LPS- and TNF-α induced phosphorylation of ERK in placenta, VAT and SAT. These findings indicate that selenium may be able to prevent inflammation and oxidative stress associated with GDM and maternal obesity. Additional in vivo studies are required to identify the efficacy of selenium supplementation in preventing inflammatory pathways activated by GDM and maternal obesity and to elucidate the mechanism involved.


Subject(s)
Diabetes, Gestational , Obesity, Maternal , Selenium , Adipose Tissue/metabolism , Cytokines/metabolism , Diabetes, Gestational/metabolism , Female , Humans , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Placenta/metabolism , Pregnancy , RNA, Messenger/metabolism , Selenium/metabolism , Selenium/pharmacology , Tumor Necrosis Factor-alpha/metabolism
2.
Placenta ; 102: 39-48, 2020 12.
Article in English | MEDLINE | ID: mdl-33218577

ABSTRACT

Gestational diabetes mellitus (GDM) is a global health issue with significant short and long-term complications for both mother and baby. There is a strong need to identify an effective therapeutic that can prevent the development of GDM. A better understanding of the pathophysiology of GDM and the relationship between the adipose tissue, the placenta and fetal growth is required. The placenta regulates fetal growth by modulating nutrient transfer of glucose, amino acids and fatty acids. Various factors secreted by the adipose tissue, such as adipokines, adipocytokines and more recently identified extracellular vesicles, can influence inflammation and interact with placental nutrient transport. In this review, the role of the placental nutrient transporters and the adipose-derived factors that can influence their function will be discussed. A better understanding of these factors and their relationship may make a potential target for therapeutic interventions to prevent the development of GDM and its consequences.


Subject(s)
Adipose Tissue/metabolism , Diabetes, Gestational/metabolism , Fetal Development , Diabetes, Gestational/physiopathology , Female , Humans , Placenta/physiopathology , Pregnancy
3.
Placenta ; 100: 1-12, 2020 10.
Article in English | MEDLINE | ID: mdl-32814232

ABSTRACT

INTRODUCTION: The world is witnessing a steady rise in the prevalence of gestational diabetes mellitus (GDM), correlated with the current obesity epidemic. Both GDM and obesity negatively impact both the health of women but also that of the next generation. GDM and maternal obesity are associated with increased maternal and fetal inflammation and oxidative stress. A safe and effective intervention that can prevent these pathological features, and reduce the intergenerational burden, is required. Phenolic acids, such as punicalagin and curcumin, possess anti-inflammatory and antioxidant properties. Thus, the aim of this study was to examine the effects of punicalagin and curcumin on pro-inflammatory cytokines and chemokines, and antioxidant expression in an in vitro model of inflammation. METHODS: Human placenta, visceral adipose tissue (VAT) and subcutaneous adipose tissue (SAT) explants were obtained at term elective Caesarean section and stimulated with TNF alpha (TNF). RESULTS: We found that punicalagin and curcumin significantly supressed TNF-induced pro-inflammatory cytokine (IL1A, IL1B, and IL6) and chemokine (CCL2-4, CXCL1, CXCL5 and CXCL8) expression in human placenta, VAT and SAT. Anti-inflammatory cytokine IL4 and IL13 mRNA expression was also upregulated by punicalagin and curcumin treatment in placenta, VAT and SAT. Punicalagin and curcumin also altered antioxidant (SOD2 and catalase) mRNA expression in placenta, VAT and SAT, with minimal effect on hydrogen peroxide concentrations in tissue lysates. CONCLUSION: These findings suggest that the phenolic acids punicalagin and curcumin possess potent anti-inflammatory capabilities in in vitro human models of inflammation. Further studies are warranted to determine their suitability as therapeutic interventions for pro-inflammatory gestational complications, including GDM and maternal obesity.


Subject(s)
Adipose Tissue/drug effects , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Curcumin/therapeutic use , Hydrolyzable Tannins/therapeutic use , Placenta/drug effects , Pregnancy Complications/drug therapy , Adipose Tissue/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antioxidants/metabolism , Curcumin/pharmacology , Cytokines/metabolism , Drug Evaluation, Preclinical , Female , Humans , Hydrogen Peroxide/metabolism , Hydrolyzable Tannins/pharmacology , Placenta/metabolism , Pregnancy
4.
Reproduction ; 160(4): 561-578, 2020 10.
Article in English | MEDLINE | ID: mdl-32698152

ABSTRACT

Spontaneous preterm birth is the leading cause of neonatal mortality and morbidity globally. Activation of the maternal immune system leads to a downstream cascade of proinflammatory events that culminate in the activation of spontaneous uterine contractions and the rupture of the foetal membranes. Anti-inflammatory agents may be a novel therapeutic approach to prevent inflammation-induced myometrial contractions and premature rupture of foetal membranes. The polyphenol gallic acid has been previously shown to exert potent anti-inflammatory effects. Thus, this study aimed to determine the effect of gallic acid on proinflammatory and pro-labour mediators in cytokine-stimulated gestational tissues in vitro. In primary human cells isolated from myometrium and foetal membranes (decidua, and amnion mesenchymal and epithelial cells), gallic acid treatment suppressed inflammation-induced expression of proinflammatory cytokines and chemokines and extracellular matrix-degrading and matrix-remodelling enzymes. Gallic acid also significantly inhibited inflammation-induced myometrial activation as evidenced by decreased expression of contraction-associated proteins, the uterotonic PGF2α and collagen cell contractility. Using a global proteomic approach, gallic acid may differentially regulate proteins associated with collagen synthesis, cell contractility and protein synthesis in primary myometrial and decidual cells. In summary, gallic acid inhibited inflammation-induced mediators involved in active labour in primary cells isolated from myometrium and foetal membranes. These in vitro studies suggest that the polyphenol gallic acid may be able to suppress the production of proinflammatory and pro-labour mediators involved in myometrial contractions and rupture of foetal membranes. Future preclinical studies may elucidate the efficacy of gallic acid in preventing inflammation-driven preterm birth.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Extraembryonic Membranes/pathology , Gallic Acid/pharmacology , Inflammation/drug therapy , Myometrium/pathology , Premature Birth/prevention & control , Proteome/analysis , Extraembryonic Membranes/metabolism , Female , Gestational Age , Humans , In Vitro Techniques , Inflammation/metabolism , Inflammation/pathology , Myometrium/metabolism , Pregnancy , Premature Birth/etiology , Premature Birth/metabolism , Premature Birth/pathology , Proteome/metabolism
5.
J Mol Endocrinol ; 65(2): 21-34, 2020 08.
Article in English | MEDLINE | ID: mdl-32580157

ABSTRACT

Gestational diabetes mellitus (GDM) affects up to 16% of pregnant women and is associated with significant long-term health detriments for the mother and her offspring. Two central features of GDM are low-grade inflammation and maternal peripheral insulin resistance, therefore therapeutics which target these may be most effective at preventing the development of GDM. Short-chain fatty acids (SCFAs), such as butyrate and propionate, are metabolites produced from the fermentation of dietary fibre by intestinal microbiota. SCFAs possess anti-inflammatory, anti-obesity and anti-diabetic properties. Therefore, this study aimed to investigate the effect of SCFAs on inflammation and insulin signalling defects in an in vitro model of GDM. Human placenta, visceral adipose tissue (VAT) and s.c. adipose tissue (SAT) were stimulated with either the pro-inflammatory cytokine TNF or bacterial product lipopolysaccharide (LPS). The SCFAs butyrate and propionate blocked TNF- and LPS-induced mRNA expression and secretion of pro-inflammatory cytokines and chemokines in placenta, VAT and SAT. Primary human cells isolated from skeletal muscle were stimulated with TNF to assess the effect of SCFAs on inflammation-induced defects in the insulin signalling pathway. Butyrate and propionate were found to reverse TNF-induced increases in IRS-1 serine phosphorylation and decreases in glucose uptake. Butyrate and propionate exerted these effects by preventing ERK activation. Taken together, these results suggest that the SCFAs may be able to improve insulin sensitivity and prevent inflammation induced by sterile or bacterial inflammation. Future in vivo studies are warranted to investigate the efficacy and safety of SCFAs in preventing insulin resistance and inflammation associated with GDM.


Subject(s)
Diabetes, Gestational/drug therapy , Fatty Acids, Volatile/therapeutic use , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Anti-Inflammatory Agents/pharmacology , Butyrates/pharmacology , Chemokines/metabolism , Diabetes, Gestational/metabolism , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Glucose/metabolism , Humans , Inflammation Mediators/metabolism , Muscle Cells/drug effects , Muscle Cells/metabolism , Muscle, Skeletal/cytology , Placenta/drug effects , Placenta/metabolism , Pregnancy , Propionates/pharmacology
6.
Mol Hum Reprod ; 26(6): 452-468, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32236411

ABSTRACT

Spontaneous preterm birth is a global health issue affecting up to 20% of pregnancies and leaves a legacy of neurodevelopmental complications. Inflammation has been implicated in a significant proportion of preterm births, where pro-inflammatory insults trigger production of additional pro-inflammatory and pro-labor mediators. Thus, novel therapeutics that can target inflammation may be a novel avenue for preventing preterm birth and improving adverse fetal outcomes. Short-chain fatty acids (SCFAs), such as butyrate and propionate, are dietary metabolites produced by bacterial fermentation of fiber in the gut. SCFAs are known to possess anti-inflammatory properties and have been found to function through G-coupled-receptors and histone deacetylases. Therefore, this study aimed to investigate the effect of SCFAs on pro-inflammatory and pro-labor mediators in an in vitro model of preterm birth. Primary human cells isolated from myometrium and fetal membranes (decidua, amnion mesenchymal and amnion epithelial cells) were stimulated with the pro-inflammatory cytokines tumor necrosis factor alpha (TNF) or interleukin 1B (IL1B). The SCFAs butyrate and propionate suppressed inflammation-induced expression of pro-inflammatory cytokines and chemokines, adhesion molecules, the uterotonic prostaglandin PGF2alpha and enzymes involved in remodeling of myometrium and degradation of the fetal membranes. Notably, propionate and butyrate also suppressed inflammation-induced prostaglandin signaling and myometrial cell contraction. These effects appear to be mediated through suppression of nuclear factor kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) activation. These results suggest that the SCFAs may be able to prevent myometrial contractions and rupture of membranes. Further in vivo studies are warranted to identify the efficacy of SCFAs as a novel anti-inflammatory therapeutic to prevent inflammation-induced spontaneous preterm birth.


Subject(s)
Butyrates/pharmacology , Butyrates/therapeutic use , Inflammation/drug therapy , Myometrium/metabolism , Propionates/pharmacology , Propionates/therapeutic use , Animals , Fatty Acids, Volatile/metabolism , Female , Histone Deacetylases/metabolism , Humans , Mitogen-Activated Protein Kinases/metabolism , Myometrium/drug effects , NF-kappa B/metabolism , Pregnancy , Premature Birth
7.
Clin Sci (Lond) ; 134(6): 571-592, 2020 03 27.
Article in English | MEDLINE | ID: mdl-32129440

ABSTRACT

Gestational diabetes mellitus (GDM) is a global health issue, whereby pregnant women are afflicted with carbohydrate intolerance with first onset during pregnancy. GDM is characterized by maternal peripheral insulin resistance, thought to be driven by low-grade maternal inflammation. Nobiletin, a polymethoxylated flavonoid, possesses potent glucose-sensitizing and anti-inflammatory properties; however, its effects in GDM have not been assessed. The present study aimed to determine the effects of nobiletin on glucose metabolism and inflammation associated with GDM in both in vitro human tissues and an in vivo animal model of GDM. In vitro, treatment with nobiletin significantly improved TNF-impaired glucose uptake in human skeletal muscle, and suppressed mRNA expression and protein secretion of pro-inflammatory cytokines and chemokines in human placenta and visceral adipose tissue (VAT). Mechanistically, nobiletin significantly inhibited Akt and Erk activation in placenta, and NF-κB activation in VAT. In vivo, GDM mice treated with 50 mg/kg nobiletin daily via oral gavage from gestational day (gd) 1-17 or via i.p. injections from gd 10-17 significantly improved glucose tolerance. Pregnant GDM mice treated with nobiletin from either gd 1-17 or gd 10-17 exhibited significantly suppressed mRNA expression of pro-inflammatory cytokines and chemokines in placenta, VAT and subcutaneous adipose tissue (SAT). Using a quantitative mass spectrometry approach, we identified differentially abundant proteins associated with the effect of nobiletin in vivo. Together, these studies demonstrate that nobiletin improves glucose metabolism and reduces inflammation associated with GDM and may be a novel therapeutic for the prevention of GDM.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Diabetes, Gestational/drug therapy , Flavones/administration & dosage , Hypoglycemic Agents/administration & dosage , Animals , Cytokines/immunology , Diabetes, Gestational/genetics , Diabetes, Gestational/immunology , Diabetes, Gestational/metabolism , Disease Models, Animal , Female , Glucose/metabolism , Humans , Mice , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Placenta/drug effects , Placenta/immunology , Placenta/metabolism , Pregnancy , Subcutaneous Fat/drug effects , Subcutaneous Fat/metabolism
8.
Clin Sci (Lond) ; 133(23): 2379-2400, 2019 12 12.
Article in English | MEDLINE | ID: mdl-31750510

ABSTRACT

Preterm birth is a global healthcare challenge. Spontaneous preterm birth (sPTB) is commonly caused by inflammation, yet there are currently no effective therapies available. The Bromodomain and Extra-Terminal motif (BET) proteins, Bromodomain-containing protein (Brd) 2 (Brd2), Brd3 and Brd4 regulate inflammation in non-gestational tissues. The roles of Brd2-4 in human pregnancy are unknown. Using human and mouse models, the present study has identified the Brd proteins part of the process by which inflammation induces parturition. Using human clinical samples, we demonstrate that labor and infection increase the expression of Brds in the uterus and fetal membranes. In primary human myometrial, amnion and decidual cells, we found that global Brd protein inhibition, as well as selective inhibition of Brds, suppressed inflammation-induced expression of mediators involved in myometrial contractions and rupture of fetal membranes. Importantly, studies in the mouse model demonstrate that the pan-Brd inhibitor JQ1 reduced intrauterine inflammation induced by bacterial endotoxin LPS as well as decreasing the effectiveness of LPS to induce parturition. These results implicate BET proteins as novel therapeutic targets for reducing inflammation associated with spontaneous preterm labor.


Subject(s)
Azepines/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Obstetric Labor, Premature/metabolism , Transcription Factors/antagonists & inhibitors , Triazoles/pharmacology , Animals , Cell Cycle Proteins/metabolism , Female , Humans , Inflammation/physiopathology , Lipopolysaccharides , Mice , Myometrium/metabolism , Obstetric Labor, Premature/prevention & control , Pregnancy , Premature Birth/etiology , Premature Birth/prevention & control , RNA, Small Interfering/metabolism , Transcription Factors/metabolism
9.
J Mol Endocrinol ; 63(3): R51-R72, 2019 10.
Article in English | MEDLINE | ID: mdl-31416051

ABSTRACT

Gestational diabetes mellitus (GDM) imposes serious short- and long-term health problems for mother and baby. An effective therapeutic that can reduce the incidence of GDM and improve long-term maternal and fetal outcomes is a major research priority, crucially important for public health. A lack of knowledge about the underlying pathophysiology of GDM has hampered the development of such therapeutics. What we do know, however, is that maternal insulin resistance, low-grade inflammation and endothelial cell dysfunction are three central features of pregnancies complicated by GDM. Indeed, data generated over the past decade have implicated a number of candidate regulators of insulin resistance, inflammation and endothelial cell dysfunction in placenta, maternal adipose tissue and skeletal muscle. These include nuclear factor-κB (NF-κB), peroxisome proliferator-activated receptors (PPARs), sirtuins (SIRTs), 5' AMP-activated protein kinase (AMPK), glycogen synthase kinase 3 (GSK3), PI3K/mTOR, inflammasome and endoplasmic reticulum (ER) stress. In this review, the identification of these as key modulators of GDM will be discussed. The biochemical pathways involved in the formation of these may represent potential sites for intervention that may translate to therapeutic interventions to prevent the development of GDM.


Subject(s)
Diabetes, Gestational/genetics , Signal Transduction/genetics , Diabetes, Gestational/physiopathology , Female , Humans , Inflammation/pathology , Insulin Resistance/genetics , Models, Biological , Placenta/pathology , Placenta/physiopathology , Pregnancy
10.
Mol Nutr Food Res ; 63(19): e1900224, 2019 10.
Article in English | MEDLINE | ID: mdl-31343820

ABSTRACT

SCOPE: Gestational diabetes mellitus (GDM), which affects up to 20% of pregnant women, is associated with maternal peripheral insulin resistance, low-grade inflammation, and oxidative stress. The flavonoid naringenin has potent anti-diabetic, anti-inflammatory, and anti-oxidative properties; however, its effects in GDM remain unknown. The study aimed to determine the effects of naringenin on glucose metabolism, inflammation, and oxidative stress associated with GDM both in vitro and in vivo. METHODS AND RESULTS: In vitro, human tissue samples obtained at term elective Caesarean section are stimulated with tumour necrosis factor alpha (TNF) to develop a GDM-like environment. Naringenin treatment significantly improves TNF-impaired glucose uptake in skeletal muscle. In placenta and visceral adipose tissue (VAT), naringenin significantly reduces expression of pro-inflammatory cytokines and chemokines and increases antioxidant mRNA expression. Mechanistically, naringenin suppresses nuclear factor κB activation. In vivo, pregnant heterozygous db/+ mice are used to model GDM. Daily intraperitoneal injections of GDM mice with naringenin from gestational day 10-17 significantly improve glucose tolerance, reduces IL1A mRNA expression, and increases antioxidant mRNA expression in placenta, VAT, and subcutaneous adipose tissue. CONCLUSION: Naringenin is shown to improve insulin sensitivity, inflammation, and oxidative stress associated with GDM and shows promise as a novel preventive therapeutic.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Diabetes, Gestational/physiopathology , Flavanones/pharmacology , Hypoglycemic Agents/pharmacology , Animals , Cesarean Section , Diabetes, Gestational/drug therapy , Disease Models, Animal , Female , Flavanones/therapeutic use , Humans , Insulin Resistance , Intra-Abdominal Fat/drug effects , Intra-Abdominal Fat/metabolism , Mice , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Placenta/drug effects , Placenta/metabolism , Pregnancy , Tumor Necrosis Factor-alpha/pharmacology
11.
Eur J Immunol ; 48(8): 1350-1363, 2018 08.
Article in English | MEDLINE | ID: mdl-29883518

ABSTRACT

There are currently no effective treatments to prevent spontaneous preterm labor. The precise upstream biochemical pathways that regulate the transition between uterine quiescence during pregnancy and contractility during labor remain unclear. It is well known however that intrauterine inflammation, including infection, is commonly associated with preterm labor. In this study, we identified the immunoproteasome subunit low-molecular-mass protein (LMP)7 mRNA expression to be significantly upregulated in laboring human myometrium. Silencing LMP7 using siRNA-targeted knockdown of LMP7 and its inhibitor ONX-0914 in human myometrial cells and tissues decreased proinflammatory cytokines (IL-6), cell chemotaxis (CXCL8, CCL2 expression, and THP-1 migration), cell to cell adhesion (ICAM1 expression and myometrial adhesion), contraction-associated proteins (PTGS2, FP, PGE2, and PGF2α), as well as suppressing contractions in myometrial cells and in myometrial tissues obtained from laboring women. In addition, LMP7 silencing reduced NF-κB RelA activity. ONX-0914 alleviated inflammation (CCL3, CXCL1, PTGS2, and IL-6) in myometrium, placenta, fetal brain, amniotic fluid, and maternal serum induced by LPS in pregnant mice. Collectively, our data suggest a novel role for ONX-014 to suppress uterine activation and contractility associated with preterm labor.


Subject(s)
Myometrium/metabolism , Obstetric Labor, Premature/prevention & control , Oligopeptides/pharmacology , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Uterine Contraction/drug effects , Animals , Cell Adhesion/drug effects , Cell Line, Tumor , Chemokine CCL2/metabolism , Female , Humans , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Interleukin-6/metabolism , Interleukin-8/metabolism , Lipopolysaccharides , Mice , Mice, Inbred C57BL , Models, Animal , Pregnancy , Proteasome Endopeptidase Complex/genetics , RNA Interference , RNA, Small Interfering/genetics , THP-1 Cells , Transcription Factor RelA/metabolism
12.
Placenta ; 48: 38-48, 2016 12.
Article in English | MEDLINE | ID: mdl-27871471

ABSTRACT

INTRODUCTION: Endocan, a member of the proteoglycan family, is involved in a wide range of diseases including obesity and diabetes. The aim of this study was to determine the effect of (i) maternal obesity and gestational diabetes mellitus (GDM) on placental endocan expression; and (ii) endocan knockdown on markers of inflammation. METHODS: Endocan mRNA and protein expression was determined in human placenta from (i) lean and obese and normal glucose tolerant (NGT) pregnant women (n = 10 patients per group); and (ii) women with GDM and BMI-matched NGT women (n = 40 patients per group). Primary villous trophoblast cells and HUVECs were used to assess the effect of endocan siRNA knockdown on pro-inflammatory cytokines. RESULTS: There was no effect of maternal obesity on placental endocan expression. Further, endocan expression was not different between lean NGT and BMI-matched women with GDM. However, endocan mRNA and protein expression was significantly higher in placenta from obese women with GDM when compared to BMI-matched NGT women. Knockdown of endocan in villous trophoblast cells and HUVECs had no effect on infection- or inflammation-induced expression and secretion of IL-6, IL-8 and MCP-1. DISCUSSION: Endocan expression is increased in the human placenta from obese women with GDM, and in response to pro-inflammatory stimuli. Loss-of-function studies in villous trophoblast cells and HUVECs revealed that endocan is not directly involved in the genesis or in the expression of pro-inflammatory cytokines induced by LPS or IL-1ß. Further studies are required to elucidate the functional consequences of increased placental endocan expression in obese GDM pregnancies.


Subject(s)
Diabetes, Gestational/metabolism , Neoplasm Proteins/metabolism , Obesity/metabolism , Placenta/metabolism , Proteoglycans/metabolism , Adult , Cytokines/metabolism , Diabetes, Gestational/genetics , Female , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Neoplasm Proteins/genetics , Obesity/genetics , Pregnancy , Proteoglycans/genetics , RNA, Small Interfering
13.
Placenta ; 47: 63-72, 2016 11.
Article in English | MEDLINE | ID: mdl-27780541

ABSTRACT

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.


Subject(s)
Activating Transcription Factor 3/metabolism , Extraembryonic Membranes/metabolism , Fetal Membranes, Premature Rupture/metabolism , Inflammation/metabolism , Activating Transcription Factor 3/genetics , Cell Line , Chorioamnionitis/metabolism , Cytokines/genetics , Cytokines/metabolism , Diglycerides/pharmacology , Extraembryonic Membranes/drug effects , Female , Fetal Membranes, Premature Rupture/genetics , Humans , Interleukin-1beta/pharmacology , Labor, Obstetric/metabolism , Oligopeptides/pharmacology , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL
...