Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
1.
BMC Pregnancy Childbirth ; 22(1): 87, 2022 Jan 31.
Article in English | MEDLINE | ID: mdl-35100981

ABSTRACT

BACKGROUND: The accumulation of reactive oxygen species (ROS) resulting from upregulated levels of oxidative stress is commonly implicated in preeclampsia (PE). Ferroptosis is a novel form of iron-dependent cell death instigated by lipid peroxidation that likely plays an important role in PE pathogenesis. This study aimed to investigate the expression profiles and functions of ferroptosis-related genes (FRGs) in early-onset preeclampsia (EOPE) and late-onset preeclampsia (LOPE). METHODS: Gene expression data and clinical information were downloaded from the Gene Expression Omnibus (GEO) database. The "limma" R package was used to screen differentially expressed genes. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and protein-protein interaction (PPI) network analyses were conducted to investigate the bioinformatics functions and molecular interactions of significantly different FRGs. Quantitative reverse transcription polymerase chain reaction (RT-qPCR) was used to verify the expression of hub FRGs in PE. RESULTS: A total of 4215 differentially expressed genes (DEGs) were identified between EOPE and preterm cases while 556 DEGs were found between LOPE and term controls. Twenty significantly different FRGs were identified in EOPE subtypes, while only 3 FRGs were identified in LOPE subtypes. Functional enrichment analysis revealed that the differentially expressed FRGs were mainly involved in EOPE and enriched in hypoxia- and iron-related pathways, such as the response to hypoxia, iron homeostasis and iron ion binding process. PPI network analysis and verification by RT-qPCR resulted in the identification of the following five FRGs of interest: FTH1, HIF1A, FTL, MAPK8 and PLIN2. CONCLUSIONS: EOPE and LOPE have distinct underlying molecular mechanisms, and ferroptosis may be mainly implicated in the pathogenesis of EOPE. Further studies are necessary for deeper inquiry into placental ferroptosis and its role in the pathogenesis of EOPE.


Subject(s)
Ferroptosis/genetics , Gene Expression Profiling , Pre-Eclampsia/genetics , Adult , Apoferritins/genetics , Down-Regulation , Female , Ferritins/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mitogen-Activated Protein Kinase 8/genetics , Oxidoreductases/genetics , Perilipin-2/genetics , Placenta/metabolism , Pregnancy , Pregnancy Trimesters/metabolism , Principal Component Analysis , Protein Interaction Maps , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
2.
Drug Metab Dispos ; 49(10): 929-937, 2021 10.
Article in English | MEDLINE | ID: mdl-34315779

ABSTRACT

Pregnancy can significantly change the pharmacokinetics of drugs, including those renally secreted by organic anion transporters (OATs). Quantifying these changes in pregnant women is logistically and ethically challenging. Hence, predicting the in vivo plasma renal secretory clearance (CLsec) and renal CL (CLrenal) of OAT drugs in pregnancy is important to design correct dosing regimens of OAT drugs. Here, we first quantified the fold-change in renal OAT activity in pregnant versus nonpregnant individual using available selective OAT probe drug CLrenal data (training dataset; OAT1: tenofovir, OAT2: acyclovir, OAT3: oseltamivir carboxylate). The fold-change in OAT1 activity during the 2nd and 3rd trimester was 2.9 and 1.0 compared with nonpregnant individual, respectively. OAT2 activity increased 3.1-fold during the 3rd trimester. OAT3 activity increased 2.2, 1.7 and 1.3-fold during the 1st, 2nd, and 3rd trimester, respectively. Based on these data, we predicted the CLsec, CLrenal and total clearance ((CLtotal) of drugs in pregnancy, which are secreted by multiple OATs (verification dataset; amoxicillin, pravastatin, cefazolin and ketorolac, R-ketorolac, S-ketorolac). Then, the predicted clearances (CLs) were compared with the observed values. The predicted/observed CLsec, CLrenal, and CLtotal of drugs in pregnancy of all verification drugs were within 0.80-1.25 fold except for CLsec of amoxicillin in the 3rd trimester (0.76-fold) and cefazolin in the 2nd trimester (1.27-fold). Overall, we successfully predicted the CLsec, CLrenal, and CLtotal of drugs in pregnancy that are renally secreted by multiple OATs. This approach could be used in the future to adjust dosing regimens of renally secreted OAT drugs which are administered to pregnant women. SIGNIFICANCE STATEMENT: To the authors' knowledge, this is the first report to successfully predict renal secretory clearance and renal clearance of multiple OAT substrate drugs during pregnancy. The data presented here could be used in the future to adjust dosing regimens of renally secreted OAT drugs in pregnancy. In addition, the mechanistic approach used here could be extended to drugs transported by other renal transporters.


Subject(s)
Biological Transport, Active/physiology , Dose-Response Relationship, Drug , Organic Anion Transporters , Pharmacokinetics , Renal Elimination/physiology , Biotransformation/physiology , Drug Dosage Calculations , Female , HEK293 Cells , Humans , Metabolic Clearance Rate , Organic Anion Transporters/classification , Organic Anion Transporters/metabolism , Pharmaceutical Preparations/classification , Pharmaceutical Preparations/metabolism , Pregnancy , Pregnancy Trimesters/drug effects , Pregnancy Trimesters/metabolism , Reproducibility of Results
3.
Psychoneuroendocrinology ; 122: 104904, 2020 12.
Article in English | MEDLINE | ID: mdl-33080521

ABSTRACT

The present study systematically reviewed 56 articles that assessed hair cortisol concentrations during pregnancy collected from PubMed, Scopus, and Web of Science on 8/9/19 and updated on 6/29/20. Our goals were to establish reference ranges by trimester based on published studies. The majority of any given sample (e.g., 70 %, the range of -1SD to +1SD) is expected to fall between 0 and 34.15 pg/mg in trimester 1 and 2, and between 8.59 and 44 pg/mg in trimester 3, with very wide ranges (e.g., values of >250 pg/mg) and substantially higher values (e.g., averages of 200's-300's reaching as high as 768 pg/mg) coming out of one specific lab. Delineating a reference range for hair cortisol concentrations across pregnancy is challenging because of known factors like differences in values returned by different laboratories and assay types. We observed inconsistency in descriptions of the data and data preparation steps post-assay. Key findings include that only half of the studies examining all three trimesters showed a constant increase in mean levels (most retrospectively assessed via segmenting), with considerable variability in patterns of change. None of the studies reported individual patterns of change. Examining within-person changes are an important next step for the field. We conclude that researchers should more clearly report decisions around outliers, units, and specifics of data transformations in the future in order to improve our ability to compare findings across studies, to understand differences in HCC values reported, and potentially to understand differences in reported associations of HCC with other phenotypes in the literature.


Subject(s)
Cortisone/standards , Stress, Psychological/metabolism , Cortisone/analysis , Female , Hair/chemistry , Humans , Pregnancy , Pregnancy Trimesters/metabolism , Reference Values , Retrospective Studies
4.
J Pharmacokinet Pharmacodyn ; 47(4): 361-383, 2020 08.
Article in English | MEDLINE | ID: mdl-32840724

ABSTRACT

Concerns over gestational effects on the disposition of drugs has highlighted the need for a better understanding of drug distribution and elimination during pregnancy. This study aimed at predicting maternal drug kinetics using a physiologically based pharmacokinetic (PBPK) modelling approach focusing on the observed gestational changes in three important Cytochrome P450 metabolizing enzymes, namely, CYP1A2, CYP2D6 and CYP3A4 at different gestational weeks (GWs). The Pregnancy PBPK model within the Simcyp Simulator V19 was used to predict the pharmacokinetics of sensitive probes to these enzymes; namely caffeine, theophylline, metoprolol, propranolol, paroxetine, midazolam, nifedipine and rilpivirine. PBPK model predictions were compared against clinical data collated from multiple studies for each compound to cover a wide spectrum of gestational ages. Pregnancy PBPK model predictions were within 2-fold error and indicated that CYP1A2 activity is approximately 0.70, 0.44 and 0.30 fold of the non-pregnant level at the end of the first, second and third trimesters, respectively. On the other hand, CYP2D6 activity increases by 1.36, 2.16 and 3.10 fold of the non-pregnant level at the end of the first, second and third trimesters, respectively. Likewise, CYP3A4 activity increases by 1.25, 1.75 and 2.32 fold of the non-pregnant level at the end of the first, second and third trimesters, respectively. The enzymes activity have been qualified throughout pregnancy. Quantified changes in drug dosing are most relevant during the third trimester, especially for drugs that are mainly eliminated by CYP1A2, CYP2D6 and CYP3A4 enzymes. The provided functions describing the continuous changes to the activity of these enzymes during pregnancy are important when modelling long term pharmacokinetic studies where longitudinal modelling or time-varying covariates are used.


Subject(s)
Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP2D6/metabolism , Cytochrome P-450 CYP3A/metabolism , Models, Biological , Prescription Drugs/pharmacokinetics , Administration, Intravenous , Administration, Oral , Adolescent , Adult , Biological Variation, Population , Dose-Response Relationship, Drug , Female , Gestational Age , Humans , Maternal Age , Metabolic Clearance Rate , Middle Aged , Pregnancy , Pregnancy Complications/drug therapy , Pregnancy Complications/metabolism , Pregnancy Trimesters/metabolism , Prescription Drugs/administration & dosage , Stochastic Processes , Tissue Distribution , Young Adult
5.
Drug Metab Dispos ; 48(9): 735-741, 2020 09.
Article in English | MEDLINE | ID: mdl-32591415

ABSTRACT

Some women take medication during pregnancy to address a variety of clinical conditions. Because of ethical and logistical concerns, it is impossible to determine fetal drug exposure, and therefore fetal risk, during pregnancy. Hence, alternative approaches need to be developed to predict maternal-fetal drug exposure throughout pregnancy. To do so, we previously developed and verified a maternal-fetal physiologically based pharmacokinetic model, which can predict fetal exposure to drugs that passively cross the placenta. However, many drugs are actively transported by the placenta (e.g., human immunodeficiency virus protease inhibitors). To extend our maternal-fetal physiologically based pharmacokinetic model to these actively transported drugs, we determined the gestational age-dependent changes in the protein abundance of placental transporters. Total cellular membrane fractions from first trimester (T1; n = 15), second trimester (T2; n = 19), and term (n = 15) human placentae obtained from uncomplicated pregnancies were isolated by ultracentrifugation. Transporter protein abundance was determined by targeted quantitative proteomics using liquid chromatography tandem mass specrometry. We observed that breast cancer resistance protein and P-glycoprotein abundance significantly decreased from T1 to term by 55% and 69%, respectively (per gram of tissue). Organic anion-transporting polypeptide (OATP) 2B1 abundance significantly decreased from T1 to T2 by 32%. In contrast, organic cation transporter (OCT) 3 and organic anion transporter 4 abundance significantly increased with gestational age (2-fold from T1 to term, 1.6-fold from T2 to term). Serotonin transporter and norepinephrine transporter did not change with gestational age. The abundance of bile salt export pump, multidrug resistance-associated protein 1-5, Na+-taurocholate cotransporting polypeptide, OATP1B1, OATP1B3, OCTN1-2, concentrative nucleoside transporter 1-3, equilibrative nucleoside transporter 2, and multidrug and toxin extrusion 1 could not be quantified. These data can be incorporated into our maternal-fetal physiologically based pharmacokinetic model to predict fetal exposure to drugs that are actively transported across the placenta. SIGNIFICANCE STATEMENT: We quantified the protein abundance of key placental uptake and efflux transporters [organic cation transporter (OCT) 3, P-glycoprotein (P-gp), breast cancer resistance protein (BCRP)] across gestational ages (first trimester, second trimester, and term) using quantitative targeted proteomics. We observed that the protein abundance of P-gp and BCRP decreased, whereas that of OCT3 increased with gestational age. Incorporating the protein abundance determined in this study into maternal-fetal physiologically based pharmacokinetic model can help us better predict fetal drug exposure to substrates of these transporters.


Subject(s)
Maternal-Fetal Exchange , Membrane Transport Proteins/metabolism , Placenta/metabolism , Pregnancy Complications/drug therapy , Pregnancy Trimesters/metabolism , Female , Gestational Age , Humans , Membrane Transport Proteins/analysis , Models, Biological , Pregnancy , Proteomics/methods
6.
Hypertension ; 75(6): 1513-1522, 2020 06.
Article in English | MEDLINE | ID: mdl-32336238

ABSTRACT

Preeclampsia, a hypertensive pregnancy disorder, links to increased long-term maternal cardiovascular disease (CVD). The risk is further increased with early-onset preeclampsia (EPE) and delivery of a growth-restricted child. We hypothesized that circulating biomarkers associated with CVD risk differed between preeclampsia subtypes and controls. We compared EPE; n=37, delivery

Subject(s)
Atherosclerosis/metabolism , Cardiovascular Diseases/epidemiology , Fetal Growth Retardation/metabolism , Matrix Metalloproteinase 1/blood , Placenta Growth Factor/blood , Pre-Eclampsia , Adult , Biomarkers/blood , Blood Pressure/physiology , Blood Pressure Determination/methods , Blood Pressure Determination/statistics & numerical data , Chemokine CX3CL1/blood , Correlation of Data , Female , Heart Disease Risk Factors , Humans , Placental Circulation/physiology , Pre-Eclampsia/blood , Pre-Eclampsia/classification , Pre-Eclampsia/diagnosis , Pre-Eclampsia/therapy , Pregnancy , Pregnancy Trimesters/metabolism
7.
Article in English | MEDLINE | ID: mdl-32120190

ABSTRACT

Fatty acids are essential for feto-placental growth and development. Maternal fatty acids and their metabolites are involved in every stage of pregnancy by supporting cell growth and development, cell signaling, and modulating other critical aspects of structural and functional processes. Early placentation process is critical for placental growth and function. Several fatty acids modulate angiogenesis as observed by increased tube formation and secretion of angiogenic growth factors in first-trimester human placental trophoblasts. Long-chain fatty acids stimulate angiogenesis in these cells via vascular endothelium growth factor (VEGF), angiopoietin-like protein 4 (ANGPTL4), fatty acid-binding proteins (FABPs), or eicosanoids. Inadequate placental angiogenesis and trophoblast invasion of the maternal decidua and uterine spiral arterioles leads to structural and functional deficiency of placenta, which contributes to preeclampsia, pre-term intrauterine growth restriction, and spontaneous abortion and also affects overall fetal growth and development. During the third trimester of pregnancy, placental preferential transport of maternal plasma long-chain polyunsaturated fatty acids is of critical importance for fetal growth and development. Fatty acids cross the placental microvillous and basal membranes by mainly via plasma membrane fatty acid transport system (FAT, FATP, p-FABPpm, & FFARs) and cytoplasmic FABPs. Besides, a member of the major facilitator superfamily-MFSD2a, present in the placenta is involved in the supply of DHA to the fetus. Maternal factors such as diet, obesity, endocrine, inflammation can modulate the expression and activity of the placental fatty acid transport activity and thereby impact feto-placental growth and development. In this review, we discuss the maternal dietary fatty acids, and placental transport and metabolism, and their roles in placental growth and development.


Subject(s)
Dietary Fats/metabolism , Fatty Acids/metabolism , Placenta/metabolism , Placentation/physiology , Angiopoietin-Like Protein 4/metabolism , Cell Membrane/metabolism , Fatty Acid-Binding Proteins/metabolism , Female , Fetal Development/physiology , Humans , Neovascularization, Physiologic/physiology , Pregnancy , Pregnancy Trimesters/metabolism , Trophoblasts/metabolism , Vascular Endothelial Growth Factor A/metabolism
8.
Ther Drug Monit ; 42(2): 264-270, 2020 04.
Article in English | MEDLINE | ID: mdl-31929398

ABSTRACT

BACKGROUND: In the United States, drug addiction has become a nationwide health crisis. Recently, buprenorphine (BUP), a maintenance therapy approved by the Food and Drug Administration, has been increasingly used in pregnant women for the treatment of opioid use disorder. Pregnancy is associated with various anatomic and physiological changes, which may result in altered drug pharmacokinetics (PKs). Previously, we reported that dose-adjusted plasma concentrations of BUP are lower during pregnancy than after pregnancy. The mechanism(s) responsible for this difference has not yet been defined. Our study aimed to evaluate alterations in cytochromes P450 (CYP)- and uridine diphosphate glucunosyltransferases (UGT)-mediated metabolism of BUP during pregnancy to determine the mechanism(s) responsible for this observation. METHODS: Data from 2 clinical studies were included in the current analysis. Study 1 was a prospective, open-labeled, nonrandomized longitudinal BUP PK study in pregnant women with a singleton gestation, stabilized on twice-daily sublingual BUP opioid substitution therapy. Each subject participated in up to 3 studies during and after pregnancy (the second, third trimester, and postpartum). The design of study 2 was similar to study 1, with patients evaluated at different time points during the pregnancy (first, second-half of pregnancy), as well as during the postpartum period. In addition, the dosing frequency of BUP study 2 participants was not restricted to twice-daily dosing. At each study visit, blood samples were collected before a BUP dose, followed by multiple collection times (10-12) after the dose, for up to 12 hours or till the end of the dosing interval. Plasma concentrations of BUP and 3 metabolites were quantified using validated ultraperformance liquid chromatography-tandem mass spectrometric assays. RESULTS: In total, 19, 18, and 14 subjects completed the PK study during 1/2 trimester, third trimester, and postpartum, respectively. The AUC ratios of norbuprenorphine and norbuprenorphine glucuronide to buprenorphine, a measure of CYP3A mediated N-demethylation, were 1.89, 1.84, and 1.33 during the first and second, third trimesters, and postpartum, respectively. The AUC ratios of buprenorphine glucuronide to BUP, indicative of UGT activity, were 0.71, 2.07, and 0.3 at first/second trimesters, third trimester, and postpartum, respectively. Linear mixed-effect modeling analysis indicated that the AUC ratios of CYP- and UGT-mediated metabolism of BUP were significantly higher during pregnancy compared with postpartum. CONCLUSIONS: The CYP and UGT activities were significantly increased as determined by the metabolic ratios of BUP during pregnancy compared with the postpartum period. The increased UGT activity appeared to account for a substantial part of the observed change in metabolic activity during pregnancy. This is in agreement with the need for BUP dose increment in pregnant women to reach similar BUP exposure and therapeutic effect as in nonpregnant subjects.


Subject(s)
Buprenorphine/pharmacokinetics , Cytochrome P-450 Enzyme System/metabolism , Glucuronosyltransferase/metabolism , Narcotic Antagonists/pharmacokinetics , Adult , Buprenorphine/analogs & derivatives , Buprenorphine/blood , Cytochrome P-450 CYP3A/metabolism , Female , Humans , Longitudinal Studies , Narcotic Antagonists/therapeutic use , Opiate Substitution Treatment/methods , Opioid-Related Disorders/drug therapy , Postpartum Period/metabolism , Pregnancy , Pregnancy Trimesters/metabolism , Young Adult
9.
J Clin Pharmacol ; 60(2): 240-255, 2020 02.
Article in English | MEDLINE | ID: mdl-31489678

ABSTRACT

Pregnancy is associated with physiological changes that may impact drug pharmacokinetics (PK). The goals of this study were to build maternal-fetal physiologically based pharmacokinetic (PBPK) models for acyclovir and emtricitabine, 2 anti(retro)viral drugs with active renal net secretion, and to (1) evaluate the predicted maternal PK at different stages of pregnancy; (2) predict the changes in PK target parameters following the current dosing regimen of these drugs throughout pregnancy; (3) evaluate the predicted concentrations of these drugs in the umbilical vein at delivery; (4) compare the model performance for predicting maternal PK of emtricitabine in the third trimester with that of previously published PBPK models; and (5) compare different previously published approaches for estimating the placental permeability of these 2 drugs. Results showed that the pregnancy PBPK model for acyclovir predicted all maternal concentrations within a 2-fold error range, whereas the model for emtricitabine predicted 79% of the maternal concentrations values within that range. Extrapolation of these models to earlier stages of pregnancy indicated that the change in the median PK target parameters remained well above the target threshold. Concentrations of acyclovir and emtricitabine in the umbilical vein were overall adequately predicted. The comparison of different emtricitabine PBPK models suggested an overall similar predictive performance in the third trimester, but the comparison of different approaches for estimating placental drug permeability revealed large differences. These models can enhance the understanding of the PK behavior of renally excreted drugs, which may ultimately inform pharmacotherapeutic decision making in pregnant women and their fetuses.


Subject(s)
Acyclovir/pharmacokinetics , Anti-HIV Agents/pharmacokinetics , Antiviral Agents/pharmacokinetics , Emtricitabine/pharmacokinetics , Pregnancy Complications, Infectious/metabolism , Acyclovir/blood , Adult , Anti-HIV Agents/blood , Antiviral Agents/blood , Clinical Trials as Topic , Computer Simulation , Drug Administration Schedule , Emtricitabine/blood , Female , Fetus/metabolism , Humans , Maternal-Fetal Exchange , Models, Biological , Placenta/metabolism , Pregnancy/blood , Pregnancy Complications, Infectious/drug therapy , Pregnancy Trimesters/metabolism , Renal Elimination , Umbilical Veins/metabolism
10.
Proteomics Clin Appl ; 13(5): e1800060, 2019 09.
Article in English | MEDLINE | ID: mdl-31162828

ABSTRACT

PURPOSE: This study is designed to screen serum proteins that may serve as biomarkers for gestational diabetes mellitus (GDM), and clarify the mechanisms of disease. EXPERIMENTAL DESIGN: By using isobaric tags for relative and absolute quantitation proteomics analysis, serum proteins levels were quantified in pregnant women who subsequently developed GDM (12-16 weeks), GDM patients (24-28 weeks), and their corresponding controls. The strategy of mixing samples is used in proteomic analysis. RESULTS: Thirty-one and 27 differentially expressed proteins are identified in the serum of pregnant women with developed GDM at 12-16 weeks and GDM patients during 24-28 weeks, respectively. Thirty eight and 28 proteins are identified in 24-28 weeks versus 12-16 weeks controls (24/12 CTR group), and 24-28 weeks GDM patients versus 12-16 weeks women with subsequently developed GDM (24/12 GDM group), respectively. Most of these proteins in the case and control subjects are associated with diabetes and maternal and perinatal short- and long-term complications. CONCLUSIONS AND CLINICAL RELEVANCE: The results highlight the roles of complement system and the blood clotting cascade in the pathogenesis of GDM. Differentially expressed proteins may serve as potential biomarkers for GDM prediction and diagnosis in the future.


Subject(s)
Blood Proteins/metabolism , Diabetes, Gestational/blood , Diabetes, Gestational/metabolism , Pregnancy Trimesters/blood , Proteomics , Adult , Biomarkers/blood , Biomarkers/metabolism , Female , Gene Expression Profiling , Humans , Pregnancy , Pregnancy Trimesters/metabolism , Protein Interaction Maps
11.
Acta Med Port ; 32(4): 305-312, 2019 Apr 30.
Article in English | MEDLINE | ID: mdl-31067425

ABSTRACT

INTRODUCTION: Inflammatory bowel disease activity is associated with adverse pregnancy outcomes. Anti-tumor necrosis factor α therapy is often required to treat flares and to maintain disease remission. However, there are concerns regarding treatment with these agents during pregnancy, as they actively cross the placental barrier. MATERIAL AND METHODS: Studies regarding anti-tumor necrosis factor α therapy during pregnancy were identified from PubMed from 1958 to January 2018. The reference lists of the selected studies were reviewed to identify complementary publications. RESULTS AND DISCUSSION: Anti-tumor necrosis factor α agents are efficient treatments for moderate-to-severe inflammatory bowel disease and may ensure remission during pregnancy. Although these drugs cross the placenta, they are considered safe for both the mother and the fetus. Furthermore, up-to-date guidelines support therapy continuation during pregnancy aiming for disease control. The same guidelines also consider stopping treatment during the third trimester to limit maternal-fetal drug transfer. However, data shows that this strategy does not completely prevent fetus exposure. In addition, stopping treatment incurs in risk of disease flare and threatens subsequent therapy response. Fetus drug exposure has not showed an association with adverse childhood development. However, as infant drug levels could be detected up to seven months after birth, postponement of live virus vaccination is recommended. CONCLUSION: There should be no disagreement among the medical community as to the need to maintain therapy aiming for disease remission during gestation in inflammatory bowel disease. Anti-tumor necrosis factor α agents are safe for both the mother and the fetus.


Introdução: O melhor preditor de complicações durante a gravidez, na doença inflamatória intestinal, é a atividade da doença. A terapêutica com agentes anti-tumor necrosis factor α atravessa a barreira placentária o que levanta questões relativamente à sua segurança durante a gravidez. Material e Métodos: Revisão bibliográfica suportada a partir de artigos indexados na PubMed (1958 a 01/2018) sobre a terapêutica anti-tumor necrosis factor α durante a gravidez na doença inflamatória intestinal. Resultados e Discussão: Os agentes anti-tumor necrosis factor α são eficazes na doença inflamatória intestinal e podem garantir a remissão clínica durante a gravidez. Estes fármacos atravessam a barreira placentária, mas são seguros para a mãe e feto. Neste sentido, as orientações atuais defendem a manutenção terapêutica durante a gravidez para assegurar a remissão clínica. Paralelamente, as mesmas orientações consideram a suspensão terapêutica durante o terceiro trimestre para limitar a exposição fetal ao fármaco. No entanto, esta estratégia não só não previne totalmente a exposição fetal, como aumenta o risco de agudização da doença e da perda de resposta à terapêutica após o seu reinício. Esta exposição fetal não está associada a alterações do desenvolvimento in utero ou neonatal. Ainda assim, uma vez que é possível dosear fármaco no recém-nascido até aos sete meses de vida, recomenda-se adiar a administração de vacinas vivas em recém-nascidos expostos. Conclusão: Não deve haver discordância na comunidade médica quanto à necessidade de garantir a remissão da doença inflamatória intestinal durante a gestação. Os agentes anti-tumor necrosis factor α devem ser vistos como opções terapêuticas seguras para mãe e feto durante a gravidez.


Subject(s)
Anti-Inflammatory Agents/pharmacokinetics , Inflammatory Bowel Diseases/drug therapy , Maternal-Fetal Exchange , Pregnancy Complications/drug therapy , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Adalimumab/pharmacokinetics , Adalimumab/therapeutic use , Adrenal Cortex Hormones/adverse effects , Adrenal Cortex Hormones/pharmacokinetics , Adrenal Cortex Hormones/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/therapeutic use , Certolizumab Pegol/pharmacokinetics , Certolizumab Pegol/therapeutic use , Female , Humans , Infliximab/pharmacokinetics , Infliximab/therapeutic use , Placenta/metabolism , Practice Guidelines as Topic , Pregnancy , Pregnancy Trimesters/metabolism
12.
BMJ Open ; 9(4): e025395, 2019 05 01.
Article in English | MEDLINE | ID: mdl-31048433

ABSTRACT

INTRODUCTION: Pregnancy is characterised by a high rate of metabolic shifts from early to late phases of gestation in order to meet the raised physiological and metabolic needs. This change in levels of metabolites is influenced by gestational weight gain (GWG), which is an important characteristic of healthy pregnancy. Inadequate/excessive GWG has short-term and long-term implications on maternal and child health. Exploration of gestational metabolism is required for understanding the quantitative changes in metabolite levels during the course of pregnancy. Therefore, our aim is to study trimester-specific variation in levels of metabolites in relation to GWG and its influence on fetal growth and newborn anthropometric traits at birth. METHODS AND ANALYSIS: A prospective longitudinal study is planned (start date: February 2018; end date: March 2023) on pregnant women that are being recruited in the first trimester and followed in subsequent trimesters and at the time of delivery (total 3 follow-ups). The study is being conducted in a hospital located in Bikaner district (66% rural population), Rajasthan, India. The estimated sample size is of 1000 mother-offspring pairs. Information on gynaecological and obstetric history, socioeconomic position, diet, physical activity, tobacco and alcohol consumption, depression, anthropometric measurements and blood samples is being collected for metabolic assays in each trimester using standardised methods. Mixed effects regression models will be used to assess the role of gestational weight in influencing metabolite levels in each trimester. The association of maternal levels of metabolites with fetal growth, offspring's weight and body composition at birth will be investigated using regression modelling. ETHICS AND DISSEMINATION: The study has been approved by the ethics committees of the Department of Anthropology, University of Delhi and Sardar Patel Medical College, Rajasthan. We are taking written informed consent after discussing the various aspects of the study with the participants in the local language.


Subject(s)
Metabolome/physiology , Pregnancy Trimesters/metabolism , Adult , Birth Weight , Female , Gestational Weight Gain/physiology , Humans , Pregnancy , Prospective Studies , Regression Analysis , Young Adult
13.
Am J Clin Nutr ; 109(4): 1064-1070, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30982859

ABSTRACT

BACKGROUND: The 2007 World Health Organization/Food and Agriculture Organization/United Nations University (WHO/FAO/UNU) recommendation for the Estimated Average Requirement (EAR) of additional protein during pregnancy for a gestational weight gain (GWG) of 12 kg (recalculated from a GWG of 13.8 kg) is 6.7 and 21.7 g/d in the second and the third trimester, respectively. This EAR is based on measurements of potassium accretion in high-income country (HIC) pregnant women. It is not known if low- to middle-income country, but well-nourished, pregnant women have comparable requirements. OBJECTIVE: We aimed to estimate total body potassium (TBK) accretion during pregnancy in Indian pregnant women, using a whole-body potassium counter (WBKC), to measure their additional protein EAR. METHODS: Well-nourished pregnant women (20-40 y, n = 38, middle socioeconomic stratum) were recruited in the first trimester of pregnancy. Anthropometric, dietary, and physical activity measurements, and measurements of TBK using a WBKC, were performed at each trimester and at birth. RESULTS: The mid-trimester weight gain was 2.7 kg and 8.0 kg in the second and the third trimester, respectively, for an average 37-wk GWG of 10.7 kg and a mean birth weight of 3.0 kg. Protein accretion was 2.7 and 5.7 g/d, for an EAR of 8.2 and 18.9 g/d in the second and the third trimester, respectively. The additional protein EAR, calculated for a GWG of 12 kg, was 9.1 and 21.2 g/d in the second and the third trimester, respectively. CONCLUSION: The additional protein requirements of well-nourished Indian pregnant women for a GWG of 12 kg in the second and third trimesters were similar to the recalculated 2007 WHO/FAO/UNU requirements for 12 kg.


Subject(s)
Dietary Proteins/metabolism , Potassium/metabolism , Pregnancy/metabolism , Adolescent , Adult , Body Mass Index , Diet , Dietary Proteins/analysis , Female , Gestational Weight Gain , Humans , India , Potassium/analysis , Pregnancy Trimesters/metabolism , Prenatal Nutritional Physiological Phenomena , Young Adult
14.
J Proteome Res ; 18(1): 292-300, 2019 01 04.
Article in English | MEDLINE | ID: mdl-30488697

ABSTRACT

Despite the increasing research attention paid to gestational diabetes mellitus (GDM) due to its high prevalence, limited knowledge is available about its pathogenesis. In this study, 428 serum samples were collected from 107 pregnant women suffering from GDM and 107 matched healthy controls. The nontargeted metabolomics data of maternal serum samples from the first (T1, n = 214) and second trimesters (T2, n = 214) were acquired by using ultrahigh performance liquid chromatography coupled with Orbitrap mass spectrometry (MS). A total of 93 differential metabolites were identified on the basis of the accurate mass and MS/MS fragmentation. After false discovery rate correction, the levels of 31 metabolites in GDM group were significantly altered in the first trimester. The differential metabolites were mainly attributed to purine metabolism, fatty acid ß-oxidation, urea cycle, and tricarboxylic acid cycle pathways. The fold changes across pregnancy (T2/T1) of six amino acids (serine, proline, leucine/isoleucine, glutamic acid, tyrosine, and ornithine), a lysophosphatidylcholine (LysoPC(20:4)), and uric acid in GDM group were significantly different from those in the control groups, suggesting that these 8 metabolites might have contributed to the occurrence and progression of GDM. The findings revealed that the amino acid metabolism, lipid metabolism, and other pathways might be disturbed prior to GDM onset and during the period from the first to the second trimester of pregnancy.


Subject(s)
Diabetes, Gestational/metabolism , Metabolomics/methods , Pregnancy Trimesters/metabolism , Adult , Amino Acids/metabolism , Blood Specimen Collection , Case-Control Studies , Diabetes, Gestational/blood , Female , Humans , Lipid Metabolism , Metabolic Networks and Pathways , Pregnancy , Young Adult
15.
PLoS One ; 13(9): e0204807, 2018.
Article in English | MEDLINE | ID: mdl-30265722

ABSTRACT

BACKGROUND: Hair cortisol concentration (HCC), as a novel promising method to retrospectively measure hypothalamic-pituitary-adrenal (HPA) axis activation, is being increasingly studied. We tested the relationships between HCC and a range of possible confounding variables in a Spanish sample of healthy adults and pregnant women. METHODS: The number of healthy adults who participated in the study was 529, being 270 males and 259 females, with a combined mean age of 37.88 years (SD = 15.66). Additionally, a separate sample of 62 pregnant women was also recruited with a mean age of 32.95 (SD = 3.67), and in the first trimester of pregnancy. Each participant was interviewed before the study to obtain sociodemographic and lifestyle variables, and a hair sample was taken from the posterior vertex of the head, cut as close to the scalp as possible. Assuming the average growth rate of head hair is 1 cm per month, a 3-cm segment was analysed, in order to measure the cortisol concentrations from a three-month period. For the pregnant women, hair samples for each trimester of pregnancy were analysed. RESULTS: The mean hair cortisol concentration was 127.91 (111.52) pg/mg for the general sample. The variables of age, education, employment status, use of hair dyes, use of oral contraceptives, and physical exercise had a significant relation to HCC. When adjusted for further variables, only education and physical exercise remained statistically significant. When including the use of oral contraceptives and only with respect to females, only physical exercise remains statistically significant. For the subsample of pregnant woman, the mean hair cortisol concentration was 334.51 (409.77) pg/mg for the first trimester, 302.18 (270.24) pg/mg for the second trimester, and 331.31 (295.46) pg/mg for the third trimester of pregnancy. None of the assessed confounding variables (age, body mass index, previous miscarriages, employment status, hair dyes, dependent children and physical exercise), except education level, was related to HCC. CONCLUSIONS: In this sample of healthy Spaniards, results suggested an association between HCC and physical exercise and educational level. In pregnant women, the prevalence of HCC was higher than in non-pregnant woman, and was related to educational level. This study emphasises the need to determine the relationship between HCC and confounders such as sociodemographic and lifestyle variables in the general population and specific groups formed by individuals such as pregnant women.


Subject(s)
Hair/metabolism , Hydrocortisone/metabolism , Pregnancy Trimesters/metabolism , Pregnancy/metabolism , Adult , Female , Humans , Male , Middle Aged , Spain
16.
Mol Hum Reprod ; 24(10): 495-509, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30124911

ABSTRACT

STUDY QUESTION: Does A Disintegrin And Metalloproteinase 8 (ADAM8) control extravillous trophoblast (EVT) differentiation and migration in early human placental development? SUMMARY ANSWER: ADAM8 mRNA preferentially localizes to invasive HLA-G-positive trophoblasts, associates with the acquirement of an EVT phenotype and promotes trophoblast migration through a mechanism requiring ß1-integrin. WHAT IS KNOWN ALREADY: Placental establishment in the first trimester of pregnancy requires the differentiation of progenitor trophoblasts into invasive EVTs that produce a diverse repertoire of proteases that facilitate matrix remodeling and activation of signaling pathways important in controlling cell migration. While multiple ADAM proteases, including ADAM8, are highly expressed by invasive trophoblasts, the role of ADAM8 in controlling EVT-related processes is unknown. STUDY DESIGN, SIZE, DURATION: First trimester placental villi and decidua (6-12 weeks' gestation), primary trophoblasts and trophoblastic cell lines (JEG3, JAR, Bewo, HTR8/SVNeo) were used to examine ADAM8 expression, localization and function. All experiments were performed on at least three independent occasions (n = 3). PARTICIPANTS/MATERIALS, SETTING, METHODS: Placental villi and primary trophoblasts derived from IRB approved first trimester placental (n = 24) and decidual (n = 4) were used to examine ADAM8 localization and expression by in situ RNAScope hybridization, flow cytometry, quantitative PCR and immunoblot analyses. Primary trophoblasts were differentiated into EVT-like cells by plating on fibronectin and were assessed by immunofluorescence microscopy and immunoblot analysis of keratin-7, vimentin, epidermal growth factor receptor (EGFR), HLA-G and ADAM8. ADAM8 function was examined in primary EVTs and trophoblastic cell lines utilizing siRNA-directed silencing and over-expression strategies. Trophoblast migration was assessed using Transwell chambers, cell-matrix binding was tested using fibronectin-adhesion assays, and ADAM8-ß1-integrin interactions were determined by immunofluorescence microscopy, co-immunoprecipitation experiments and function-promoting/inhibiting antibodies. MAIN RESULTS AND THE ROLE OF CHANCE: Within first trimester placental tissues, ADAM8 preferentially localized to HLA-G+ trophoblasts residing within anchoring columns and decidua. Functional experiments in primary trophoblasts and trophoblastic cell lines show that ADAM8 promotes trophoblast migration through a mechanism independent of intrinsic protease activity. We show that ADAM8 localizes to peri-nuclear and cell-membrane actin-rich structures during cell-matrix attachment and promotes trophoblast binding to fibronectin matrix. Moreover, ADAM8 potentiates ß1-integrin activation and promotes cell migration through a mechanism dependent on ß1-integrin function. LIMITATIONS, REASONS FOR CAUTION: The primary limitation of this study was the use of in vitro experiments in examining ADAM8 function, as well as the implementation of immortalized trophoblastic cell lines. Histological localization of ADAM8 within placental and decidual tissue sections was limited to mRNA level analysis. Further, patient information corresponding to tissues obtained by elective terminations was not available. WIDER IMPLICATIONS OF THE FINDINGS: The novel non-proteolytic pro-migratory role for ADAM8 in controlling trophoblast migration revealed by this study sheds insight into the importance of ADAM8 in EVT biology and placental development. STUDY FUNDING/COMPETING INTEREST(S): This work was supported by grants from the Natural Sciences and Engineering Research Council of Canada (NSERC-Discovery Grant) and the Canadian Institutes of Health Research (CIHR-Open Operating Grant). There are no conflicts or competing interests. TRIAL REGISTRATION NUMBER: NA.


Subject(s)
ADAM Proteins/metabolism , Integrin beta1/metabolism , Membrane Proteins/metabolism , Placenta/metabolism , Trophoblasts/metabolism , Cell Line , Cell Movement/physiology , Female , Humans , Pregnancy , Pregnancy Trimesters/metabolism
17.
Mol Ther ; 26(9): 2189-2205, 2018 09 05.
Article in English | MEDLINE | ID: mdl-30061037

ABSTRACT

Preeclampsia (PE) is the leading cause of maternal and neonatal morbidity and mortality. Defects in trophoblast invasion, differentiation of endovascular extravillous trophoblasts (enEVTs), and spiral artery remodeling are key factors in PE development. There are no markers clinically available to predict PE, leaving expedited delivery as the only effective therapy. Dysregulation of miRNA in clinical tissues and maternal circulation have opened a new avenue for biomarker discovery. In this study, we investigated the role of miR-218-5p in PE development. miR-218-5p was highly expressed in EVTs and significantly downregulated in PE placentas. Using first-trimester trophoblast cell lines and human placental explants, we found that miR-218-5p overexpression promoted, whereas anti-miR-218-5p suppressed, trophoblast invasion, EVT outgrowth, and enEVT differentiation. Furthermore, miR-218-5p accelerated spiral artery remodeling in a decidua-placenta co-culture. The effect of miR-218-5p was mediated by the suppression of transforming growth factor (TGF)-ß2 signaling. Silencing of TGFB2 mimicked, whereas treatment with TGF-ß2 partially reversed, the effects of miR-218-5p. Taken together, these findings demonstrate that miR-218-5p promotes trophoblast invasion and enEVT differentiation through a novel miR-218-5p-TGF-ß2 pathway. This study elucidates the role of an miRNA in enEVT differentiation and spiral artery remodeling and suggests that downregulation of miR-218-5p contributes to PE development.


Subject(s)
MicroRNAs/metabolism , Placenta/cytology , Placenta/metabolism , Pregnancy Trimesters/metabolism , Trophoblasts/cytology , Trophoblasts/metabolism , Cell Line , Female , Human Umbilical Vein Endothelial Cells , Humans , Immunohistochemistry , In Vitro Techniques , Pre-Eclampsia/metabolism , Pregnancy , Transforming Growth Factor beta2/genetics , Transforming Growth Factor beta2/metabolism , Transforming Growth Factor beta2/pharmacology , Trophoblasts/drug effects
18.
Biol Res Nurs ; 20(3): 292-299, 2018 05.
Article in English | MEDLINE | ID: mdl-29490472

ABSTRACT

OBJECTIVE: To evaluate relationships between hair cortisol levels and perceived stress in mothers who deliver preterm and term. We hypothesized that the rate of change in cortisol levels would be greater in the preterm delivery group. METHODS: This preliminary study compared hair cortisol levels and Perceived Stress Scale (PSS) scores in predominately Caucasian mothers who delivered preterm ( n = 22) and term ( n = 30). We collected PSS and hair samples of ≥10 cm in length from mothers after delivery. Hair was segmented into three sections, and cortisol was measured using enzyme-linked immunosorbent assay. RESULTS: The mean gestational age was 31.45 ( SD = 4.2) weeks for preterm deliveries and 39.45 ( SD = 1.1) for term. Cortisol differed significantly in the third trimester between mothers delivering term and preterm ( t = 2.16, df = 48, p = .04) and trended toward significance in the second trimester ( t = 1.88, df = 48, p = .06). PSS differed significantly between the two groups ( t = -2.96, df = 50, p = .05). Our data did not provide support for our hypothesis. CONCLUSION: There appeared to be a blunted, flattened pattern of change in cortisol levels across gestation in the women who delivered preterm, suggesting diminished hypothalamic-pituitary-adrenal axis responsiveness in mechanisms that promote preterm labor. Future studies are needed to further evaluate best strategies for measuring the mechanisms of allostatic load during pregnancy along with the psychoneuroendocrine and immune triggers and placental responses that lead to premature birth.


Subject(s)
Hair/metabolism , Hydrocortisone/metabolism , Mothers , Premature Birth/metabolism , Stress, Psychological/metabolism , Adult , Female , Gestational Age , Humans , Hypothalamo-Hypophyseal System , Infant, Newborn , Pituitary-Adrenal System , Pregnancy , Pregnancy Trimester, Third/metabolism , Pregnancy Trimesters/metabolism , Young Adult
19.
BJOG ; 125(11): 1379-1387, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29460466

ABSTRACT

The transcription factor nuclear factor kappa B (NFκB) controls the expression of over 400 genes, some of which are associated with reproductive events. During implantation, immune cells accumulate in the maternal-fetal interface; they secrete inflammatory mediators under the control of NFĸB, the level of which also rises. NFĸB is then downregulated to maintain gestation, but its level rises again before birth to manage prostaglandin, cytokine, and chemokine synthesis, and to stimulate uterine contraction. This review summarises the current state of knowledge about NFκB and its role in the molecular regulation of processes related to pregnancy development. TWEETABLE ABSTRACT: This review examines the current state of knowledge about role of NFκB in the development of pregnancy.


Subject(s)
Embryo Implantation/physiology , Labor, Obstetric/metabolism , NF-kappa B/physiology , Pregnancy Trimesters/metabolism , Chemokines/metabolism , Cytokines/metabolism , Female , Humans , Inflammation Mediators/metabolism , Pregnancy , Prostaglandins/metabolism , Uterine Contraction/metabolism
20.
Mediators Inflamm ; 2018: 8476217, 2018.
Article in English | MEDLINE | ID: mdl-30622436

ABSTRACT

The development of the fetal nervous system mirrors general fetal development, comprising a combination of genetic resources and effects of the intrauterine environment. Our aim was to assess the 2nd trimester amniotic fluid levels of brain-derived neurotrophic factor (BDNF) and to investigate its association with fetal growth. In accordance with our study design, samples of amniotic fluid were collected from women who had undergone amniocentesis early in the 2nd trimester. All pregnancies were followed up until delivery and fetal growth patterns and birth weights were recorded, following which pregnancies were divided into three groups based on fetal weight: (1) AGA (appropriate for gestational age), (2) SGA (small for gestational age), and (3) LGA (large for gestational age). We focused on these three groups representing a reflection of the intrauterine growth spectrum. Our results revealed the presence of notably higher BDNF levels in the amniotic fluid of impaired growth fetuses by comparison with those of normal growth. Both SGA and macrosomic fetuses are characterized by notably higher amniotic fluid levels of BDNF (mean values of 36,300 pg/ml and 35,700 pg/ml, respectively) compared to normal-growth fetuses (mean value of 32,700 pg/ml). Though apparently small, this difference is, nevertheless, statistically significant (p value < 0.05) in SGA fetuses in the extremes of the distribution, i.e., below the 3rd centile. In conclusion, there is clear evidence that severe impairment of fetal growth induces the increased production of fetal brain growth factor as an adaptive mechanism in reaction to a hostile intrauterine environment, thereby accelerating fetal brain development and maturation.


Subject(s)
Amniotic Fluid/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Pregnancy Trimesters/metabolism , Birth Weight/genetics , Birth Weight/physiology , Female , Fetal Development/genetics , Fetal Development/physiology , Gestational Age , Humans , Infant, Newborn , Infant, Small for Gestational Age , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL
...