Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 289
Filter
1.
Pediatr Int ; 65(1): e15604, 2023.
Article in English | MEDLINE | ID: mdl-37551666

ABSTRACT

BACKGROUND: We recently reported on a late preterm infant born at 36 weeks' gestation with serious arrhythmia due to hyperkalemia associated with long-term maternal ritodrine administration. It is unknown whether ritodrine alone increases the risk of neonatal hyperkalemia in infants born at 34-36 weeks' gestation. METHODS: This single-center, retrospective, cohort study enrolled late preterm infants (34-36 gestational weeks) born between 2004 and 2018. Cases with maternal magnesium sulfate use were not sufficient for statistical analysis and so were excluded from the study. Risk factors for the occurrence of hyperkalemia were determined based on clinical relevance and previous reports. RESULTS: In all, 212 late preterm infants with maternal ritodrine use and 400 infants without tocolysis were included in the study. Neonatal hyperkalemia occurred in 5.7% (12/212) in the ritodrine group and 1.8% (7/400) in the control group. The risk of neonatal hyperkalemia was significantly increased by maternal ritodrine administration with a crude odds ratio (OR) of 3.37 (95% confidence interval [CI]: 1.30-8.69; p < 0.01) and an adjusted OR of 3.71 (95% CI: 1.41-9.74; p < 0.01) on multivariable analysis. Long-term tocolysis (≥28 days) with ritodrine increased the risk of neonatal hyperkalemia with 9.3% (11/118) of infants developing hyperkalemia (adjusted OR 4.86; 95% CI: 1.59-14.83; p < 0.01). Neonatal hyperkalemia was not found within 2 weeks of ritodrine administration. CONCLUSION: This research suggests that late preterm infants born after long-term ritodrine administration are at risk of neonatal hyperkalemia and require special attention.


Subject(s)
Hyperkalemia , Obstetric Labor, Premature , Ritodrine , Pregnancy , Infant , Female , Infant, Newborn , Humans , Ritodrine/adverse effects , Obstetric Labor, Premature/chemically induced , Retrospective Studies , Cohort Studies , Hyperkalemia/chemically induced , Hyperkalemia/epidemiology , Infant, Premature
2.
Cytokine ; 160: 156054, 2022 12.
Article in English | MEDLINE | ID: mdl-36191359

ABSTRACT

BACKGROUND: Preterm birth is a global public health threat. Inflammatory reaction is thought to mediate preterm birth. The role of nicotine, an anti-inflammatory agent that is mediated by cholinergic anti-inflammatory pathways (CAP), remains unclear in the pathogenesis. METHODS: Pregnant rats were randomly divided into four groups (20 rats each): pregnant control group (P), RU486-treated group (PTL), RU486 and nicotine-treated group (PTL + N), RU486, nicotine and α-BGT treated group (PTL + N + A). Rats were administered RU486 (1.0 mg/kg) by subcutaneous injection on gestational day (GD) 18 to establish PTL model. Subcutaneous injection of nicotine (1 mg/kg) was administered daily from GD 16 to 18. α-BGT (1 µg/kg) was administrated subcutaneously in two sessions and each session was 30 min prior to nicotine. TNF-α, IL-1ß, IL-4, IL-6, IL-10 in myometrium and serum were detected by Luminex. Macrophage infiltration and α7nAChR were detected by IHC. RESULTS: We established a RU486-induced preterm labor rat model. Preterm labor was associated with a striking upregulation inflammatory mediators and increased macrophage infiltration. Nicotine significantly prolonged gestation (P < 0.05) and α-BGT treatment reversed the prolonged interval (P < 0.05). The cytokines all markedly elevated at 12 h, but deceased after delivery (P < 0.05). The IL-1ß and TNF-α in serum were significantly increased in PTL group vs P group (P < 0.05), and decreased after nicotine treatment (P < 0.05). The cytokines IL-1ß, IL-4, IL-6, IL-10 and TNF-α in myometrium increased as the same trend as in serum. Nicotine treatment also downregulated the expression of α7nAChR in pregnant tissue. CONCLUSION: We confirmed the increased inflammation in preterm birth. Nicotine was able to down-regulate the inflammatory mediates and prolong the pregnant duration in PTL model, which might be induced by activating α7nAChR through CAP. This study provides a novel evidence supporting the future development of therapeutic target for preterm birth.


Subject(s)
Obstetric Labor, Premature , Animals , Anti-Inflammatory Agents , Cytokines/metabolism , Female , Inflammation/metabolism , Inflammation Mediators , Interleukin-10/metabolism , Interleukin-4/metabolism , Interleukin-6/metabolism , Mifepristone , Neuroimmunomodulation , Nicotine , Obstetric Labor, Premature/chemically induced , Obstetric Labor, Premature/drug therapy , Pregnancy , Premature Birth , Rats , Tumor Necrosis Factor-alpha/metabolism , alpha7 Nicotinic Acetylcholine Receptor
3.
J Clin Pharm Ther ; 47(7): 1036-1048, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35304748

ABSTRACT

WHAT IS KNOWN AND OBJECTIVE: Premature birth affects more than 15 million infants, as well as mothers and families around the world. With the relaxation of the two-child policy, the problem of premature birth has become relatively prominent in China. According to statistics, China had a birth population of 15.23 million in 2018, with a considerably large number of premature births. This study aims to evaluate the efficacy and safety of tocolysis in the treatment of preterm delivery, provide clinical evidence for medical staff and promote the self-management of patients with premature births. METHODS: Four English databases (PubMed, Embase, Cochrane Library and Web of Science) were retrieved by computer, the retrieval time was from the establishment of each database to November 2021, and the randomized controlled trials for the treatment of preterm delivery were screened according to the pre-set natriuretic exclusion criteria. After literature screening, data selection and risk of bias evaluation were independently conducted by two researchers. R 4.1.1 and Stata 17.0 software were used for statistical analysis. RESULTS AND DISCUSSION: A total of 44 RCTs were included, including 6939 patients. The results of network meta-analysis reveal that in terms of effectiveness, indomethacin was the most effective intervention measure, followed by nifedipine, and the difference was statistically significant; regarding safety, nifedipine was the safest intervention measure, followed by indomethacin, and the difference was statistically significant; and in respect of adverse reactions, ritodrine had the highest probability, and the difference was statistically significant. WHAT IS NEW AND CONCLUSION: Nifedipine may be better for delayed delivery and less likely to produce adverse pregnancy outcomes, followed by indomethacin. Limited by the number and quality of recipient studies, the aforementioned conclusions need to be verified through more high-quality studies. At the same time, the focus should be on patients with twin pregnancy and patients with clinical manifestations of extreme preterm delivery.


Subject(s)
Obstetric Labor, Premature , Premature Birth , Tocolytic Agents , Female , Humans , Indomethacin/therapeutic use , Infant , Infant, Newborn , Network Meta-Analysis , Nifedipine/therapeutic use , Obstetric Labor, Premature/chemically induced , Obstetric Labor, Premature/drug therapy , Obstetric Labor, Premature/prevention & control , Pregnancy , Premature Birth/chemically induced , Premature Birth/drug therapy , Premature Birth/prevention & control , Tocolysis/methods , Tocolytic Agents/adverse effects
4.
Environ Res ; 196: 110894, 2021 05.
Article in English | MEDLINE | ID: mdl-33609551

ABSTRACT

BACKGROUND: Previous reports indicate an association between ambient temperature (Ta) and air pollution exposure during pregnancy and preterm birth (PTB). Nevertheless, information regarding the association between environmental factors and specific precursors of spontaneous preterm birth is lacking. We aimed to determine the association between Ta and air pollution during gestation and the precursors of spontaneous preterm parturition, i.e. preterm labor (PTL) and preterm prelabor rupture of membranes (PPROM). METHODS: From 2003 to 2013 there were 84,476 deliveries of singleton gestation that comprised the study cohort. Exposure data during pregnancy included daily measurements of temperature and particulate matter <2.5 µm and <10 µm, PM2.5 and PM10, respectively. Deliveries were grouped into PPROM, PTL and non-spontaneous preterm and term deliveries. Exposure effect was tested in windows of a week and two days prior to admission for delivery and adjusted to gestational age and socio-economic status. Poisson regression models were used for analyses. RESULTS: There is an association of environmental exposure with the precursors of spontaneous preterm parturition; PPROM was more sensitive to Ta fluctuations than PTL. This effect was modified by the ethnicity, Bedouin-Arabs were susceptible to elevated Ta, especially within the last day prior to admission with PPROM (Relative Risk (RR) =1.19 [95% CI, 1.03; 1.37]). Jews, on the other hand, were susceptible to ambient pollutants, two (RR=1.025 [1.010; 1.040]) and one (RR= 1.017 [1.002; 1.033]) days prior to spontaneous PTL with intact membranes resulting in preterm birth. CONCLUSION: High temperature is an independent risk factor for PPROM among Bedouin-Arabs; ambient pollution is an independent risk factor for spontaneous PTL resulting in preterm birth. Thus, the precursors of spontaneous preterm parturition differ in their association with environmental factors.


Subject(s)
Fetal Membranes, Premature Rupture , Obstetric Labor, Premature , Premature Birth , Female , Gestational Age , Humans , Infant, Newborn , Obstetric Labor, Premature/chemically induced , Obstetric Labor, Premature/epidemiology , Particulate Matter , Pregnancy , Premature Birth/chemically induced , Premature Birth/epidemiology
5.
Biol Reprod ; 102(2): 445-455, 2020 02 14.
Article in English | MEDLINE | ID: mdl-31599921

ABSTRACT

In our earlier work, we found that intrauterine (i.u.) and intraperitoneal (i.p.) injection of LPS (10-µg serotype 0111:B4) induced preterm labor (PTL) with high pup mortality, marked systemic inflammatory response and hypotension. Here, we used both i.u. and i.p. LPS models in pregnant wild-type (wt) and CCR2 knockout (CCR2-/-) mice on E16 to investigate the role played by the CCL2/CCR2 system in the response to LPS. Basally, lower numbers of monocytes and macrophages and higher numbers of neutrophils were found in the myometrium, placenta, and blood of CCR2-/- vs. wt mice. After i.u. LPS, parturition occurred at 14 h in both groups of mice. At 7 h post-injection, 70% of wt pups were dead vs. 10% of CCR2-/- pups, but at delivery 100% of wt and 90% of CCR2-/- pups were dead. Myometrial and placental monocytes and macrophages were generally lower in CCR2-/- mice, but this was less consistent in the circulation, lung, and liver. At 7 h post-LPS, myometrial ERK activation was greater and JNK and p65 lower and the mRNA levels of chemokines were higher and of inflammatory cytokines lower in CCR2-/- vs. wt mice. Pup brain and placental inflammation were similar. Using the IP LPS model, we found that all measures of arterial pressure increased in CCR2-/- but declined in wt mice. These data suggest that the CCL2/CCR2 system plays a critical role in the cardiovascular response to LPS and contributes to pup death but does not influence the onset of inflammation-induced PTL.


Subject(s)
Arterial Pressure/physiology , Lipopolysaccharides/adverse effects , Myometrium/metabolism , Obstetric Labor, Premature/chemically induced , Placenta/metabolism , Receptors, CCR2/metabolism , Animals , Arterial Pressure/drug effects , Disease Models, Animal , Female , Inflammation/genetics , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Mice, Knockout , Monocytes/drug effects , Monocytes/metabolism , Myometrium/drug effects , Obstetric Labor, Premature/genetics , Obstetric Labor, Premature/metabolism , Parturition/drug effects , Parturition/genetics , Parturition/metabolism , Placenta/drug effects , Pregnancy , Receptors, CCR2/genetics
6.
FASEB J ; 33(2): 2743-2758, 2019 02.
Article in English | MEDLINE | ID: mdl-30312114

ABSTRACT

Preterm birth (PTB), the leading cause of neonatal morbidity and mortality, urgently requires novel therapeutic agents. Spontaneous PTB, resulting from preterm labor, is commonly caused by intrauterine infection/inflammation. Statins are well-established, cholesterol-lowering drugs that can reduce inflammation and inhibit vascular smooth muscle contraction. We show that simvastatin reduced the incidence of PTB in a validated intrauterine LPS-induced PTB mouse model, decreased uterine proinflammatory mRNA concentrations (IL-6, Cxcl1, and Ccl2), and reduced serum IL-6 concentration. In human myometrial cells, simvastatin reduced proinflammatory mediator mRNA and protein expression (IL-6 and IL-8) and increased anti-inflammatory cytokine mRNA expression (IL-10 and IL-13). Critically, simvastatin inhibited myometrial cell contraction, basally and during inflammation, and reduced phosphorylated myosin light chain concentration. Supplementation with mevalonate and geranylgeranyl pyrophosphate, but not farnesyl pyrophosphate, abolished these anticontractile effects, indicating that the Rho/Rho-associated protein kinase pathway is critically involved. Thus, simvastatin reduces PTB incidence in mice, inhibits myometrial contractions, and exhibits key anti-inflammatory effects, providing a rationale for investigation into the repurposing of statins to treat preterm labor in women.-Boyle, A. K., Rinaldi, S. F., Rossi, A. G., Saunders, P. T. K., Norman, J. E. Repurposing simvastatin as a therapy for preterm labor: evidence from preclinical models.


Subject(s)
Anticholesteremic Agents/pharmacology , Drug Repositioning , Inflammation/prevention & control , Myometrium , Obstetric Labor, Premature/drug therapy , Simvastatin/pharmacology , Uterine Contraction/drug effects , Animals , Cells, Cultured , Disease Models, Animal , Female , Humans , Inflammation/etiology , Inflammation/pathology , Lipopolysaccharides/toxicity , Mice , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Myometrium/cytology , Myometrium/drug effects , Myometrium/metabolism , Obstetric Labor, Premature/chemically induced , Obstetric Labor, Premature/pathology , Pregnancy , Signal Transduction/drug effects
7.
Adv Exp Med Biol ; 1252: 107-114, 2020.
Article in English | MEDLINE | ID: mdl-32816269

ABSTRACT

Non-obstetric surgery is needed in 0.75-2% of pregnant women, and safety of anesthesia for mother and child are key points at this time. Some breast diseases need to be approached in a short time interval, and surgery must be performed during pregnancy . In these cases, the technique of anesthesia regarding local, regional or general anesthesia and type of anesthetic medicine are selected based on the extent of the procedure, gestational age, and condition of the mother and child. The ideal timing for any surgery during pregnancy is in the second trimester because the risk of fetal adverse effects as well as preterm labor are lower. However, surgery of breast cancer during pregnancy is performed in any trimester as guided by treatment guidelines and is not deferred based on anesthesia preferences. Various types of anesthesia for breast surgery during pregnancy , preoperative and postoperative considerations are discussed in this chapter.


Subject(s)
Anesthesia/adverse effects , Anesthesia/methods , Anesthetics , Breast/surgery , Pregnancy Complications , Breast Neoplasms/surgery , Female , Fetus/drug effects , Humans , Obstetric Labor, Premature/chemically induced , Obstetric Labor, Premature/etiology , Obstetric Labor, Premature/prevention & control , Pregnancy , Pregnancy Complications/chemically induced , Pregnancy Complications/etiology , Pregnancy Complications/prevention & control
8.
Biol Reprod ; 100(5): 1306-1318, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30596885

ABSTRACT

Sterile intra-amniotic inflammation is commonly observed in patients with spontaneous preterm labor, a syndrome that commonly precedes preterm birth, the leading cause of perinatal morbidity and mortality worldwide. However, the mechanisms leading to sterile intra-amniotic inflammation are poorly understood and no treatment exists for this clinical condition. Herein, we investigated whether the alarmin S100B could induce sterile intra-amniotic inflammation by activating the NLRP3 inflammasome, and whether the inhibition of this pathway could prevent preterm labor/birth and adverse neonatal outcomes. We found that the ultrasound-guided intra-amniotic administration of S100B induced a 50% rate of preterm labor/birth and a high rate of neonatal mortality (59.7%) without altering the fetal and placental weights. Using a multiplex cytokine array and immunoblotting, we reported that S100B caused a proinflammatory response in the amniotic cavity and induced the activation of the NLRP3 inflammasome in the fetal membranes, indicated by the upregulation of the NLRP3 protein and increased release of active caspase-1 and mature IL-1ß. Inhibition of the NLRP3 inflammasome via the specific inhibitor MCC950 prevented preterm labor/birth by 35.7% and reduced neonatal mortality by 26.7%. Yet, inhibition of the NLRP3 inflammasome at term did not drastically obstruct the physiological process of parturition. In conclusion, the data presented herein indicate that the alarmin S100B can induce sterile intra-amniotic inflammation, preterm labor/birth, and adverse neonatal outcomes by activating the NLRP3 inflammasome, which can be prevented by inhibiting such a pathway. These findings provide evidence that sterile intra-amniotic inflammation could be treated by targeting the NLRP3 inflammasome.


Subject(s)
Furans/pharmacology , Inflammation/prevention & control , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Obstetric Labor, Premature/prevention & control , Premature Birth/prevention & control , S100 Calcium Binding Protein beta Subunit/pharmacology , Sulfonamides/pharmacology , Animals , Animals, Newborn , Cytokines/genetics , Cytokines/metabolism , Female , Fetus/drug effects , Furans/administration & dosage , Gene Expression Regulation/drug effects , Heterocyclic Compounds, 4 or More Rings , Indenes , Inflammation/chemically induced , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Obstetric Labor, Premature/chemically induced , Placenta/drug effects , Pregnancy , Premature Birth/chemically induced , S100 Calcium Binding Protein beta Subunit/administration & dosage , Sulfonamides/administration & dosage , Sulfones
9.
Mol Hum Reprod ; 25(5): 274-282, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30892651

ABSTRACT

Preterm labour is a common pregnancy complication contributing to major maternal and fetal morbidity and mortality. We have found microRNA (miR)-212-3p, a potential infection-associated molecule, was significantly over-expressed during human preterm labour. However, the mechanism remains unknown. In this study, we have adopted a lipopolysaccharide (LPS)-induced Institute of Cancer Research murine preterm model to examine the role of miR-212-3p in the infection-induced preterm labour. Myometrial miR-212-3p expression was increased by nearly 4-fold in the term labour group (P = 0.10) and 12-fold (P = 0.03) in the LPS-induced preterm labour group compared with the non-labour group. In vitro cellular experiments confirmed that a series of pro-inflammatory cytokines, including interleukin (IL)1B (P = 0.02) and IL-6 (P = 0.01), rather than LPS (P = 0.08) itself could significantly upregulate miR-212-3p expression in human myometrial smooth muscle cells. Methyl-CpG-binding protein 2 (MeCP2), as a target gene of miR-212-3p confirmed by our dual luciferase assay, influenced myocyte contractility and connexin 43 expression which is an important contraction-associated protein. Therefore, we conclude that miR-212-3p may be involved in infection-induced preterm labour through MeCP2 and it is a promoting molecule and novel target for the diagnosis and treatment of preterm labour in the future.


Subject(s)
Lipopolysaccharides/pharmacology , Methyl-CpG-Binding Protein 2/genetics , MicroRNAs/genetics , Myocytes, Smooth Muscle/drug effects , Obstetric Labor, Premature/genetics , Animals , Base Sequence , Connexin 43/genetics , Connexin 43/metabolism , Female , Gene Expression Regulation , Genes, Reporter , Humans , Infant, Newborn , Interleukin-1beta/pharmacology , Interleukin-6/pharmacology , Luciferases/genetics , Luciferases/metabolism , Methyl-CpG-Binding Protein 2/metabolism , Mice , Mice, Inbred ICR , MicroRNAs/agonists , MicroRNAs/metabolism , Models, Animal , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Myometrium/cytology , Myometrium/drug effects , Myometrium/metabolism , Obstetric Labor, Premature/chemically induced , Obstetric Labor, Premature/metabolism , Obstetric Labor, Premature/pathology , Pregnancy
10.
Biol Reprod ; 98(3): 299-308, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29315356

ABSTRACT

Preterm birth is widespread and causes 35% of all neonatal deaths. Infants who survive face potential long-term complications. A major contributing factor of preterm birth is infection. We investigated the role of interleukin 22 (IL22) as a potential clinically relevant cytokine during gestational infection. IL22 is an effector molecule secreted by immune cells. While the expression of IL22 was reported in normal nonpregnant endometrium and early pregnancy decidua, little is known about uterine IL22 expression during mid or late gestational stages of pregnancy. Since IL22 has been shown to be an essential mediator in epithelial regeneration and wound repair, we investigated the potential role of IL22 during defense against an inflammatory response at the maternal-fetal interface. We used a well-established model to study infection and infection-associated inflammation during preterm birth in the mouse. We have shown that IL22 is upregulated to respond to an intrauterine lipopolysaccharide administration and plays an important role in controlling the risk of inflammation-induced preterm birth. This paper proposes IL22 as a treatment method to combat infection and prevent preterm birth in susceptible patients.


Subject(s)
Interleukins/metabolism , Lipopolysaccharides/pharmacology , Obstetric Labor, Premature/metabolism , Obstetric Labor, Premature/prevention & control , Up-Regulation/physiology , Uterus/metabolism , Animals , Caspases/metabolism , Fas Ligand Protein/metabolism , Female , Interleukins/genetics , Mice , Obstetric Labor, Premature/chemically induced , Pregnancy , Up-Regulation/drug effects , Uterus/drug effects , Interleukin-22
11.
Ann Neurol ; 82(3): 457-465, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28856694

ABSTRACT

OBJECTIVE: To evaluate the effects of epilepsy and antiepileptic drugs (AEDs) used during pregnancy on fetal growth and preterm delivery. METHODS: This study included singleton liveborn infants born to women enrolled in the North American Antiepileptic Drug Pregnancy Registry between 1997 and 2016. Data were collected prospectively through telephone interviews. The prevalence of preterm birth (<37 weeks) and small for gestational age status (SGA) among infants exposed prenatally to AEDs when used by women with epilepsy (WWE) or women without epilepsy (WWOE) was compared with that among infants unexposed to AEDs and born to WWOE. Multivariate log-binomial regression models were used to estimate relative risks (RRs) and 95% confidence intervals (CIs). RESULTS: The study population included infants born to 6,777 AED-WWE, 696 AED-WWOE, and 486 no-AED-WWOE. The risk of prematurity was 6.2% for no-AED-WWOE, 9.3% for AED-WWE (RR = 1.5, 95% CI = 1.0-2.1), and 10.5% for AED-WWOE (RR = 1.5, 95% CI = 1.0-2.4). Prenatal exposure to AEDs in WWE and WWOE was associated with a mean lower birth weight of 110 and 136g, respectively, as compared to no-AED-WWOE. The prevalence of SGA was 5.0% for no-AED-WWOE, 10.9% for AED-WWE (RR = 2.0, 95% CI = 1.3-3.0), and 11.0% for AED-WWOE (RR = 1.9, 95% CI = 1.2-2.9). Within users of AEDs in monotherapy, the prevalence of SGA ranged from 7.3% for lamotrigine to 18.5% for topiramate. INTERPRETATION: Women on AEDs during pregnancy, whether for epilepsy or for other neuropsychiatric indications, are at a higher risk of delivering prematurely and giving birth to SGA newborns. The risk may vary by drug. Ann Neurol 2017;82:457-465.


Subject(s)
Anticonvulsants/adverse effects , Epilepsy/drug therapy , Fetal Development/drug effects , Obstetric Labor, Premature/epidemiology , Adult , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Female , Humans , Infant, Newborn , Infant, Premature , Infant, Small for Gestational Age , Male , Obstetric Labor, Premature/chemically induced , Pregnancy , Prevalence , Prospective Studies , Registries , Risk , Young Adult
12.
Toxicol Appl Pharmacol ; 356: 114-119, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30075163

ABSTRACT

Arsenic is an environmental toxicant. The association of gestational arsenic exposure with adverse pregnant outcomes remains controversial. This study was to investigate the association of serum As level with adverse pregnant outcomes in a large Chinese cohort population. Total 3194 mother-and-infant pairs were recruited from the China-Anhui Birth Cohort Study. Maternal serum arsenic (As) concentration was measured using hydride generation-atomic fluorescence spectrometry. Subjects were divided into L-As group and H-As group in accordance to the 75th percentile of serum As concentration. The associations of serum As level during gestation with adverse pregnant outcomes were analyzed. The incidence of small-for-gestational-age (SGA) newborns was elevated in H-As group compared to L-As group (9.9% vs 7.6%, P = .044). After controlling confounders and stratified analysis, the adjusted OR for SGA was significant only in girls with H-As but not in boys. Moreover, the incidence of preterm delivery (PTD) was elevated in H-As group compared to L-As group (7.0% vs 4.8%, P = .016). Further analysis found that the adjusted OR for moderate-to-late PTD was 1.47 (95%CI: 1.03, 2.09; P = .034) in H-As group as compared with L-As group. These results indicate that maternal serum As level during gestation is positively associated with adverse pregnant outcomes.


Subject(s)
Arsenic/blood , Maternal Exposure/adverse effects , Pregnancy Outcome , Adult , Asian People , Birth Weight , China/epidemiology , Cohort Studies , Dose-Response Relationship, Drug , Female , Humans , Infant, Newborn , Infant, Small for Gestational Age , Obstetric Labor, Premature/chemically induced , Obstetric Labor, Premature/epidemiology , Pregnancy , Sex Factors , Spectrometry, Fluorescence , Young Adult
13.
J Obstet Gynaecol ; 38(2): 185-188, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28816562

ABSTRACT

The relationship between Behcet's disease (BD) and pregnancy is only reported in limited number of studies. We retrospectively collected data of 26 women with BD diagnosis and their 66 pregnancies. We analysed patients according to disease activity, age at BD diagnosis, age at first/last pregnancy, obstetric history, obstetric complications, neonatal birthweight, associated foetal abnormalities and pregnancy-related complications. Sixteen miscarriages (24.2%), two intrauterine deaths (3%) and 48 live births (72.8%) were identified. Preterm labour was observed in 12 (24%) of 50 deliveries. Colchicine was used in six pregnancies, however, there was no drug treatment for BD in the remaining 59. There was a higher rate of preterm labour and low birthweight in patients using colchicine. BD was in remission in 60 (90.9%) of 66 pregnancies, and disease flared up only in six cases. In conclusion, BD patients with altered symptoms during pregnancy carry an increased risk of obstetric complications. IMPACT STATEMENT What is already known on this subject: There are limited and conflicting data about the interaction between BD and gestation. What the results of this study add: Our findings indicated that patients who were in an active symptomatic phase of BD and were being treated with colchicine had an increased risk of preterm delivery and low birthweight. What the implications are of these findings for clinical practice and/or further research: Clinicians should consider increased obstetric complication risk among patients with active BD.


Subject(s)
Behcet Syndrome/drug therapy , Colchicine/administration & dosage , Pregnancy Complications, Cardiovascular/drug therapy , Pregnancy Outcome , Abortion, Spontaneous , Adult , Behcet Syndrome/complications , Behcet Syndrome/physiopathology , Colchicine/adverse effects , Female , Gestational Age , Humans , Infant, Low Birth Weight , Infant, Newborn , Obstetric Labor, Premature/chemically induced , Placenta/immunology , Pregnancy , Pregnancy Complications, Cardiovascular/immunology , Premature Birth , Retrospective Studies , Young Adult
14.
Therapie ; 73(3): 199-207, 2018.
Article in English | MEDLINE | ID: mdl-29100610

ABSTRACT

AIM OF THE STUDY: To evaluate whether azathioprine exposure during pregnancy increases the risk of birth defects and prematurity. METHOD: Prospective comparative observational study using the French pregnancy database TERAPPEL. To evaluate birth defects, outcomes of pregnancies exposed to azathioprine during the 1st trimester were prospectively assessed and compared to that of pregnancies exposed to another drug used for the same indications. Secondly, the rate of preterm births was compared between fetuses exposed to azathioprine at least during the third trimester and those exposed during the first trimester only. RESULTS: From 447 requests for a risk assessment for women receiving azathioprine during pregnancy, 193 pregnancies meet inclusion criteria. One hundred and twenty-four of them were exposed to azathioprine during the 1st trimester and were compared to that of 124 pregnancies exposed to another drug used for the same indication. Azathioprine use during the first trimester was not statistically associated with the risk of all birth defects ([7.3% vs. 5.4%]; [OR=1.36; 95%CI: 0.44-4.20]) nor with major birth defects (5.2% vs. 1.8% [OR=2.96; 95%CI: 0.56-15.64]). The rate of preterm births (22.5% vs. 27.3%, P=0.579) was similar regardless of the exposure period to azathioprine (at least during the third trimester or during the first trimester only). CONCLUSIONS: This study confirms that first trimester exposure to azathioprine is not associated with an elevated rate of birth defects and that the high rate of preterm births among women exposed to azathioprine is probably explained by the underlying maternal disease.


Subject(s)
Azathioprine/adverse effects , Immunosuppressive Agents/adverse effects , Pregnancy Outcome , Abnormalities, Drug-Induced/epidemiology , Adult , Female , France , Humans , Infant, Low Birth Weight , Infant, Newborn , Obstetric Labor, Premature/chemically induced , Obstetric Labor, Premature/epidemiology , Pregnancy , Pregnancy Trimesters
15.
J Pineal Res ; 63(4)2017 Nov.
Article in English | MEDLINE | ID: mdl-28776755

ABSTRACT

Preterm birth is a major contributor to early and delayed physical and cognitive impairment. Epidemiological and experimental data indicate that maternal infections are a significant and preventable cause of preterm birth. Recently, melatonin has been suggested to exert neuroprotective effects in several models of brain injury. Here, we sought to investigate whether the administration of melatonin is able to prevent lipopolysaccharide (LPS)-induced fetal brain damage in a model of LPS-induced preterm labor. For this purpose, 15-day pregnant BALB/c mice received intraperitoneally 2 doses of LPS or vehicle: the first one at 10:00 hours (0.26 mg/kg) and the second at 13:00 hours (0.52 mg/kg). On day 14 of pregnancy, a group of mice was subcutaneously implanted with a pellet of 25 mg melatonin. This experimental protocol resulted in 100% of preterm birth and pup death in the LPS group and a 50% of term birth and pup survival in the melatonin + LPS group. In the absence of melatonin, fetuses from LPS-treated mothers showed histological signs of brain damage, microglial/macrophage activation, and higher levels of IL-1ß, inducible nitric oxide synthase (NOS), and neuronal NOS mRNAs as well as increased histone acetyltransferase activity and histone H3 hyperacetylation. In contrast, antenatal administration of melatonin prevented LPS-induced fetal brain damage. Moreover, when behavioral traits were analyzed in the offspring from control, melatonin, and melatonin + LPS, no significant differences were found, suggesting that melatonin prevented LPS-induced long-term neurodevelopmental impairments. Collectively, our results suggest that melatonin could be a new therapeutic tool to prevent fetal brain damage and its long-term consequences induced by maternal inflammation.


Subject(s)
Birth Injuries/prevention & control , Brain Injuries/prevention & control , Melatonin/pharmacology , Neuroprotective Agents/pharmacology , Premature Birth , Animals , Birth Injuries/etiology , Brain Injuries/etiology , Female , Inflammation/chemically induced , Lipopolysaccharides/toxicity , Mice , Mice, Inbred BALB C , Obstetric Labor, Premature/chemically induced , Pregnancy , Premature Birth/chemically induced
16.
BMC Med ; 14(1): 86, 2016 06 13.
Article in English | MEDLINE | ID: mdl-27291689

ABSTRACT

BACKGROUND: Preterm birth is now recognized as the primary cause of infant mortality worldwide. Interplay between hormonal and inflammatory signaling in the uterus modulates the onset of contractions; however, the relative contribution of each remains unclear. In this study we aimed to characterize temporal transcriptome changes in the uterus preceding term labor and preterm labor (PTL) induced by progesterone withdrawal or inflammation in the mouse and compare these findings with human data. METHODS: Myometrium was collected at multiple time points during gestation and labor from three murine models of parturition: (1) term gestation; (2) PTL induced by RU486; and (3) PTL induced by lipopolysaccharide (LPS). RNA was extracted and cDNA libraries were prepared and sequenced using the Illumina HiSeq 2000 system. Resulting RNA-Seq data were analyzed using multivariate modeling approaches as well as pathway and causal network analyses and compared against human myometrial transcriptome data. RESULTS: We identified a core set of temporal myometrial gene changes associated with term labor and PTL in the mouse induced by either inflammation or progesterone withdrawal. Progesterone withdrawal initiated labor without inflammatory gene activation, yet LPS activation of uterine inflammation was sufficient to override the repressive effects of progesterone and induce a laboring phenotype. Comparison of human and mouse uterine transcriptomic datasets revealed that human labor more closely resembles inflammation-induced PTL in the mouse. CONCLUSIONS: Labor in the mouse can be achieved through inflammatory gene activation yet these changes are not a requisite for labor itself. Human labor more closely resembles LPS-induced PTL in the mouse, supporting an essential role for inflammatory mediators in human "functional progesterone withdrawal." This improved understanding of inflammatory and progesterone influence on the uterine transcriptome has important implications for the development of PTL prevention strategies.


Subject(s)
Inflammation Mediators/metabolism , Labor, Obstetric/metabolism , Obstetric Labor, Premature/metabolism , Progesterone/metabolism , Transcriptome/physiology , Uterus/physiology , Animals , Female , Humans , Inflammation/chemically induced , Inflammation/genetics , Inflammation/physiopathology , Labor, Obstetric/drug effects , Labor, Obstetric/genetics , Lipopolysaccharides/toxicity , Mice , Models, Animal , Myometrium/drug effects , Myometrium/physiology , Obstetric Labor, Premature/chemically induced , Obstetric Labor, Premature/genetics , Parturition/drug effects , Parturition/genetics , Parturition/metabolism , Pregnancy , Progesterone/genetics , Transcriptome/drug effects , Uterus/drug effects
17.
Biol Reprod ; 95(6): 125, 2016 12.
Article in English | MEDLINE | ID: mdl-27760748

ABSTRACT

Inflammation plays a key role in human term and preterm labor (PTL). Intrauterine LPS has been widely used to model inflammation-induced complications of pregnancy, including PTL. It has been shown to induce an intense myometrial inflammatory cell infiltration, but the role of LPS-induced inflammatory cell activation in labor onset and fetal demise is unclear. We investigated this using a mouse model of PTL, where an intrauterine injection of 10 µg of LPS (serotype 0111:B4) was given at E16 of CD1 mouse pregnancy. This dose induced PTL at an average of 12.7 h postinjection in association with 85% fetal demise. Flow cytometry showed that LPS induced a dramatic systemic inflammatory response provoking a rapid and marked leucocyte infiltration into the maternal lung and liver in association with increased cytokine levels. Although there was acute placental inflammatory gene expression, there was no corresponding increase in fetal brain inflammatory gene expression until after fetal demise. There was marked myometrial activation of NFκB and MAPK/AP-1 systems in association with increased chemokine and cytokine levels, both of which peaked with the onset of parturition. Myometrial macrophage and neutrophil numbers were greater in the LPS-injected mice with labor onset only; prior to labor, myometrial neutrophils and monocytes numbers were greater in PBS-injected mice, but this was not associated with an earlier onset of labor. These data suggest that intrauterine LPS induces parturition directly, independent of myometrial inflammatory cell infiltration, and that fetal demise occurs without fetal inflammation. Intrauterine LPS provokes a marked local and systemic inflammatory response but with limited inflammatory cell infiltration into the myometrium or placenta.


Subject(s)
Inflammation/immunology , Leukocytes/immunology , Lipopolysaccharides/pharmacology , Myometrium/immunology , Obstetric Labor, Premature/immunology , Uterus/drug effects , Animals , Chemokines/metabolism , Cytokines/metabolism , Female , Gene Expression , Inflammation/chemically induced , Inflammation/metabolism , Leukocytes/drug effects , Leukocytes/metabolism , Lung/drug effects , Lung/immunology , Lung/metabolism , Mice , Myometrium/drug effects , Myometrium/metabolism , NF-kappa B/metabolism , Obstetric Labor, Premature/chemically induced , Obstetric Labor, Premature/metabolism , Pregnancy , Signal Transduction/physiology , Uterus/immunology , Uterus/metabolism
18.
Am J Pathol ; 185(9): 2390-401, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26212908

ABSTRACT

Intrauterine inflammation is recognized as a key mediator of both normal and preterm birth but is also associated with neonatal neurological injury. Lipopolysaccharide (LPS) is often used to stimulate inflammatory pathways in animal models of infection/inflammation-induced preterm labor; however, inconsistencies in maternal and neonatal responses to LPS are frequently reported. We hypothesized that LPS serotype-specific responses may account for a portion of these inconsistencies. Four different Escherichia coli LPS serotypes (O111:B4, O55:B5, O127:B8, and O128:B12) were administered to CD1 mice via intrauterine injection at gestational day 16. Although control animals delivered at term 60 ± 15 hours postinjection (p.i.), those administered with O111:B4 delivered 7 ± 2 hours p.i., O55:B5 delivered 10 ± 3 hours p.i., O127:B8 delivered 16 ± 10 hours p.i., and O128:B12 delivered 17 ± 2 hours p.i. (means ± SD). A correlation between the onset of preterm labor and myometrial activation of the inflammatory transcription factor, activator protein 1, but not NF-κB was observed. Specific LPS serotypes induced differential activation of downstream contractile and inflammatory pathways in myometrium and neonatal pup brain. Our findings demonstrate functional disparity in inflammatory pathway activation in response to differing LPS serotypes. Selective use of LPS serotypes may represent a useful tool for targeting specific inflammatory response mechanisms in these models.


Subject(s)
Lipopolysaccharides/pharmacology , Myometrium/drug effects , Obstetric Labor, Premature/chemically induced , Premature Birth/metabolism , Animals , Disease Models, Animal , Escherichia coli/drug effects , Female , Inflammation/chemically induced , Inflammation/complications , Mice , Myometrium/metabolism , Pregnancy , Pregnancy Complications/chemically induced , Transcription Factor AP-1/metabolism
19.
FASEB J ; 29(6): 2653-66, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25757563

ABSTRACT

Preterm birth is an inflammatory process resulting from the massive infiltration of innate immune cells and the production of proinflammatory cytokines in the myometrium. However, proinflammatory cytokines, which induce labor in vivo, fail to induce labor-associated features in human myometrial cells (MCs). We thus aimed to investigate if reactive oxygen species (ROS) production could be the missing step between immune cell activation and MC response. Indeed, we found that ROS production is increased in the human preterm laboring myometrium (27% ROS producing cells, respectively, versus 2% in nonlaboring controls), with 90% ROS production in macrophages. Using LPS-stimulated myometrial samples and cell coculture experiments, we demonstrated that ROS production is required for labor onset. Furthermore, we showed that ROS are required first in the NADPH oxidase (NADPHox)-2/NF-κB-dependent macrophage response to inflammatory stimuli but, more importantly, to trigger macrophage-induced MCs transactivation. Remarkably, in a murine model of LPS-induced preterm labor (inducing delivery within 17 hours, with no pup survival), cotreatment with glutathione delayed labor onset up to 94 hours and prevented in utero fetal distress, allowing 46% pups to survive. These results suggest that targeting ROS production with the macrophage-permeable antioxidant glutathione could constitute a promising strategy to prevent preterm birth.


Subject(s)
Fetal Death/prevention & control , Glutathione/pharmacology , Macrophages/metabolism , Myometrium/drug effects , Obstetric Labor, Premature/prevention & control , Reactive Oxygen Species/metabolism , Adult , Animals , Animals, Newborn , Blotting, Western , Cell Line, Tumor , Cells, Cultured , Female , Gene Expression , Glutathione/administration & dosage , Humans , Infant, Newborn , Lipopolysaccharides , Male , Mice, Inbred C57BL , Myometrium/cytology , Myometrium/metabolism , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , NF-kappa B/metabolism , Obstetric Labor, Premature/chemically induced , Obstetric Labor, Premature/metabolism , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Young Adult
20.
Pediatr Res ; 80(3): 433-9, 2016 09.
Article in English | MEDLINE | ID: mdl-27100048

ABSTRACT

BACKGROUND: Maternal intrauterine infection/inflammation represents the major etiology of preterm delivery and the leading cause of neonatal mortality and morbidity. The aim of this study was to investigate the anti-inflammatory properties of thioredoxin-1 in vivo and its potential ability to attenuate the rate of inflammation-induced preterm delivery. METHODS: Two intraperitoneal injections of lipopolysaccharide from Escherichia coli were administered in pregnant mice on gestational day 15, with a 3-h interval between the injections. From either 1 h before or 1 h after the first lipopolysaccharide injection, mice received three intravenous injections of either recombinant human thioredoxin-1, ovalbumin, or vehicle, with a 3-h interval between injections. RESULTS: Intraperitoneal injection of lipopolysaccharide induced a rise of tumor necrosis factor-α, interferon-γ, monocyte chemotactic protein 1, and interleukin-6 in maternal serum levels and provoked preterm delivery. Recombinant human thoredoxin-1 prevented the rise in these proinflammatory cytokine levels. After the inflammatory challenge, placentas exhibited severe maternal vascular dilatation and congestion and a marked decidual neutrophil activation. These placental pathological findings were ameliorated by recombinant human thioredoxin-1, and the rate of inflammation-induced preterm delivery was attenuated. CONCLUSION: Thioredoxin-1 may thus represent a novel effective treatment to delay inflammation-induced preterm delivery.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Obstetric Labor, Premature/drug therapy , Thioredoxins/pharmacology , Animals , Animals, Newborn , Chemokine CCL2/blood , Cytokines/blood , Female , Humans , Inflammation , Interferon-gamma/blood , Interleukin-6/blood , Lipopolysaccharides , Macrophages/metabolism , Male , Mice , Mice, Inbred C3H , Obstetric Labor, Premature/chemically induced , Placenta/metabolism , Pregnancy , Reactive Oxygen Species/metabolism , Recombinant Proteins/pharmacology , Thioredoxins/physiology , Tumor Necrosis Factor-alpha/blood
SELECTION OF CITATIONS
SEARCH DETAIL