Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.975
Filter
1.
Int J Mol Sci ; 25(13)2024 Jul 07.
Article in English | MEDLINE | ID: mdl-39000556

ABSTRACT

Obesity is an important risk factor for the development of pregnancy complications. We investigated the effects of pregestational overweight and obesity on maternal lipidome during pregnancy and on newborns' characteristics. The study encompassed 131 pregnant women, 99 with pre-pregnancy body mass index (BMI) < 25 kg/m2 and 32 with BMI ≥ 25 kg/m2. Maternal lipid status parameters, plasma markers of cholesterol synthesis and absorption and sphingolipids were determined in each trimester. Data on neonatal height, weight and APGAR scores were assessed. The results showed a higher prevalence (p < 0.05) of pregnancy and childbirth complications among the participants with elevated pregestational BMI. Levels of total cholesterol, HDL-cholesterol (p < 0.05) and LDL-cholesterol (p < 0.01) were significantly lower, and concentrations of triglycerides were higher (p < 0.05) in women with increased pre-gestational BMI. Lower concentrations of the cholesterol synthesis marker, desmosterol, in the 2nd trimester (p < 0.01) and the cholesterol absorption marker, campesterol, in each trimester (p < 0.01, p < 0.05, p < 0.01, respectively) were also found in this group. Markers of maternal cholesterol synthesis were in positive correlation with neonatal APGAR scores in the group of mothers with healthy pre-pregnancy weight but in negative correlation in the overweight/obese group. Our results indicate that gestational adaptations of maternal lipidome depend on her pregestational nutritional status and that such changes may affect neonatal outcomes.


Subject(s)
Body Mass Index , Lipidomics , Obesity , Overweight , Pregnancy Complications , Humans , Female , Pregnancy , Infant, Newborn , Adult , Obesity/metabolism , Obesity/blood , Lipidomics/methods , Overweight/metabolism , Pregnancy Complications/metabolism , Pregnancy Complications/blood , Lipids/blood , Cholesterol/blood
2.
Int J Mol Sci ; 25(12)2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38928446

ABSTRACT

Multiple sclerosis (MS) is a common disease in young women of reproductive age, characterized by demyelination of the central nervous system (CNS). Understanding how genes related to MS are expressed during pregnancy can provide insights into the potential mechanisms by which pregnancy affects the course of this disease. This review article presents evidence-based studies on these patients' gene expression patterns. In addition, it constructs interaction networks using bioinformatics tools, such as STRING and KEGG pathways, to understand the molecular role of each of these genes. Bioinformatics research identified 25 genes and 21 signaling pathways, which allows us to understand pregnancy patients' genetic and biological phenomena and formulate new questions about MS during pregnancy.


Subject(s)
Computational Biology , Multiple Sclerosis , Humans , Multiple Sclerosis/genetics , Multiple Sclerosis/metabolism , Female , Pregnancy , Computational Biology/methods , Gene Regulatory Networks , Pregnancy Complications/genetics , Pregnancy Complications/metabolism , Gene Expression Profiling , Signal Transduction/genetics , Gene Expression Regulation
3.
Front Endocrinol (Lausanne) ; 15: 1381180, 2024.
Article in English | MEDLINE | ID: mdl-38752179

ABSTRACT

Background: The prevalence of autism spectrum disorder (ASD) has significantly risen in the past three decades, prompting researchers to explore the potential contributions of environmental factors during pregnancy to ASD development. One such factor of interest is gestational hypothyroxinemia (HTX), a frequent condition in pregnancy associated with cognitive impairments in the offspring. While retrospective human studies have linked gestational HTX to autistic traits, the cellular and molecular mechanisms underlying the development of ASD-like phenotypes remain poorly understood. This study used a mouse model of gestational HTX to evaluate ASD-like phenotypes in the offspring. Methods: To induce gestational HTX, pregnant mice were treated with 2-mercapto-1-methylimidazole (MMI), a thyroid hormones synthesis inhibitor, in the tap-drinking water from embryonic days (E) 10 to E14. A separate group received MMI along with a daily subcutaneous injection of T4, while the control group received regular tap water during the entire pregnancy. Female and male offspring underwent assessments for repetitive, anxious, and social behaviors from postnatal day (P) 55 to P64. On P65, mice were euthanized for the evaluation of ASD-related inflammatory markers in blood, spleen, and specific brain regions. Additionally, the expression of glutamatergic proteins (NLGN3 and HOMER1) was analyzed in the prefrontal cortex and hippocampus. Results: The HTX-offspring exhibited anxious-like behavior, a subordinate state, and impaired social interactions. Subsequently, both female and male HTX-offspring displayed elevated proinflammatory cytokines in blood, including IL-1ß, IL-6, IL-17A, and TNF-α, while only males showed reduced levels of IL-10. The spleen of HTX-offspring of both sexes showed increased Th17/Treg ratio and M1-like macrophages. In the prefrontal cortex and hippocampus of male HTX-offspring, elevated levels of IL-17A and reduced IL-10 were observed, accompanied by increased expression of hippocampal NLGN3 and HOMER1. All these observations were compared to those observed in the Control-offspring. Notably, the supplementation with T4 during the MMI treatment prevents the development of the observed phenotypes. Correlation analysis revealed an association between maternal T4 levels and specific ASD-like outcomes. Discussion: This study validates human observations, demonstrating for the first time that gestational HTX induces ASD-like phenotypes in the offspring, highlighting the need of monitoring thyroid function during pregnancy.


Subject(s)
Autism Spectrum Disorder , Prenatal Exposure Delayed Effects , Animals , Female , Pregnancy , Autism Spectrum Disorder/etiology , Autism Spectrum Disorder/metabolism , Mice , Male , Prenatal Exposure Delayed Effects/metabolism , Phenotype , Behavior, Animal , Hypothyroidism/metabolism , Thyroxine/blood , Biomarkers/metabolism , Mice, Inbred C57BL , Pregnancy Complications/metabolism , Disease Models, Animal , Inflammation/metabolism , Social Behavior
4.
Biomed Pharmacother ; 176: 116815, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38788598

ABSTRACT

Programmed cell death is intricately linked to various physiological phenomena such as growth, development, and metabolism, as well as the proper function of the pancreatic ß cell and the migration and invasion of trophoblast cells in the placenta during pregnancy. Traditional and recently identified programmed cell death include apoptosis, autophagy, pyroptosis, necroptosis, and ferroptosis. In addition to cancer and degenerative diseases, abnormal activation of cell death has also been implicated in pregnancy related diseases like preeclampsia, gestational diabetes mellitus, intrahepatic cholestasis of pregnancy, fetal growth restriction, and recurrent miscarriage. Excessive or insufficient cell death and pregnancy related diseases may be mutually determined, ultimately resulting in adverse pregnancy outcomes. In this review, we systematically describe the characteristics and mechanisms underlying several types of cell death and their roles in pregnancy related diseases. Moreover, we discuss potential therapeutic strategies that target cell death signaling pathways for pregnancy related diseases, hoping that more meaningful treatments will be applied in clinical practice in the future.


Subject(s)
Cell Death , Pregnancy Complications , Humans , Pregnancy , Female , Cell Death/physiology , Animals , Pregnancy Complications/metabolism , Pregnancy Complications/pathology , Signal Transduction , Apoptosis/physiology , Autophagy/physiology
5.
Acta Cir Bras ; 39: e391524, 2024.
Article in English | MEDLINE | ID: mdl-38629649

ABSTRACT

PURPOSE: Pre-eclampsia (PE) is a pregnancy-related complication. Eucommia is effective in the treatment of hypertensive disorders in pregnancy, but the specific effects and possible mechanisms of Eucommia granules (EG) in PE remain unknown. The aim of this study was to investigate the effects and possible mechanisms of EG in PE rats. METHODS: Pregnant Sprague Dawley rats were divided into five groups (n = 6): the control group, the model group, the low-dose group, the medium-dose group, and the high-dose group of EG. The PE model was established by subcutaneous injection of levonitroarginine methyl ester. Saline was given to the blank and model groups, and the Eucommia granules were given by gavage to the remaining groups. Blood pressure and urinary protein were detected. The body length and weight of the pups and the weight of the placenta were recorded. Superoxide dismutase (SOD) activity and levels of malondialdehyde (MDA), placental growth factor (PIGF), and soluble vascular endothelial growth factor receptor-1 (sFIt-1) were measured in the placenta. Pathological changes were observed by hematoxylin-eosin staining. Wnt/ß-catenin pathway-related protein expression was detected using Western blot. RESULTS: Compared with the model group, the PE rats treated with EG had lower blood pressure and urinary protein. The length and weight of the pups and placental weight were increased. Inflammation and necrosis in the placental tissue was improved. SOD level increased, MDA content and sFIt-1/PIGF ratio decreased, and Wnt/ß-catenin pathway-related protein expression level increased. Moreover, the results of EG on PE rats increased with higher doses of EG. CONCLUSIONS: EG may activate the Wnt/ß-catenin pathway and inhibit oxidative stress, inflammation, and vascular endothelial injury in PE rats, thereby improving the perinatal prognosis of preeclamptic rats. EG may inhibit oxidative stress, inflammation, and vascular endothelial injury through activation of the Wnt/ß-catenin pathway in preeclampsia rats, thereby improving perinatal outcomes in PE rats.


Subject(s)
Pre-Eclampsia , Pregnancy Complications , Humans , Rats , Female , Pregnancy , Animals , Pre-Eclampsia/drug therapy , Pre-Eclampsia/metabolism , Pre-Eclampsia/pathology , Placenta , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor A/metabolism , beta Catenin/metabolism , Placenta Growth Factor/metabolism , Placenta Growth Factor/pharmacology , Placenta Growth Factor/therapeutic use , Oxidative Stress , Pregnancy Complications/metabolism , Inflammation/pathology , Superoxide Dismutase/metabolism
6.
J Obstet Gynaecol ; 44(1): 2345276, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38685831

ABSTRACT

BACKGROUND: In order to contribute new insights for future prevention and treatment of intrahepatic cholestasis of pregnancy (ICP), and to promote positive pregnancy outcomes, we evaluated serum Ca2+ levels and inositol 1,4,5-trisphosphate receptor (InsP3R) expression in the liver tissue of a rat ICP model. METHODS: After establishing the model by injection of oestradiol benzoate and progesterone into pregnant rats, animals were divided into normal control (n = 5) and ICP model groups (n = 5). The expression of InsP3R protein in the liver, and serum levels of Ca2+, glycocholic acid and bile acid were detected. RESULTS: InsP3R mRNA and protein were significantly lower in the ICP model group compared to the normal group, as determined by qPCR and immunohistochemistry, respectively. Serum enzyme-linked immunosorbent assay results revealed significantly higher levels of glycocholic acid and bile acid in the ICP model group compared to the normal group, while Ca2+ levels were significantly lower. The levers of Ca2+ were significantly and negatively correlated with the levels of glycocholic acid. The observed decrease in Ca2+ was associated with an increase in total bile acids, but there was no significant correlation. CONCLUSIONS: Our results revealed that the expression of InsP3R and serum Ca2+ levels was significantly decreased in the liver tissue of ICP model rats. Additionally, Ca2+ levels were found to be negatively correlated with the level of glycocholic acid.


This study investigated the relationship between serum Ca2+ levels, inositol 1,4,5-trisphosphate receptor (InsP3R) expression and intrahepatic cholestasis of pregnancy (ICP) in a rat model. The results indicated a significant decrease in InsP3R expression and Ca2+ in the disease group compared to the control group, alongside elevated levels of glycocholic acid and bile acid. The levels of Ca2+ exhibited a negative correlation with the levels of glycocholic acid. These findings indicated that the decrease of InsP3R expression and Ca2+ levels may be related to the pathogenesis of ICP. The study provides further insight into the treatment of this disease.


Subject(s)
Bile Acids and Salts , Calcium , Cholestasis, Intrahepatic , Disease Models, Animal , Estradiol , Inositol 1,4,5-Trisphosphate Receptors , Liver , Pregnancy Complications , Animals , Female , Pregnancy , Rats , Bile Acids and Salts/metabolism , Bile Acids and Salts/blood , Calcium/metabolism , Calcium/blood , Calcium Signaling , Cholestasis, Intrahepatic/metabolism , Cholestasis, Intrahepatic/blood , Estradiol/blood , Estradiol/analogs & derivatives , Glycocholic Acid/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Liver/metabolism , Pregnancy Complications/metabolism , Progesterone/blood , Rats, Sprague-Dawley , Male
7.
Psychoneuroendocrinology ; 166: 107060, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38677195

ABSTRACT

BACKGROUND: The placenta acts as a buffer to regulate the degree of fetal exposure to maternal cortisol through the 11-Beta Hydroxysteroid Dehydrogenase isoenzyme type 2 (11-ß HSD2) enzyme. We conducted a systematic review and meta-analysis to assess the effect of prenatal psychological distress (PPD) on placental 11-ß HSD2 gene expression and explore the related mechanistic pathways involved in fetal neurodevelopment. METHODS: We searched PubMed, Embase, Scopus, APA PsycInfo®, and ProQuest Dissertations for observational studies assessing the association between PPD and 11-ß HSD2 expression in human placentas. Adjusted regression coefficients (ß) and corresponding 95% confidence intervals (CIs) were pooled based on three contextual PPD exposure groups: prenatal depression, anxiety symptoms, and perceived stress. RESULTS: Of 3159 retrieved records, sixteen longitudinal studies involving 1869 participants across seven countries were included. Overall, exposure to PPD disorders showed weak negative associations with the placental 11-ß HSD2 gene expression as follows: prenatal depression (ß -0.01, 95% CI 0.05-0.02, I2=0%), anxiety symptoms (ß -0.02, 95% CI 0.06-0.01, I2=0%), and perceived stress (ß -0.01 95% CI 0.06-0.04, I2=62.8%). Third-trimester PPD exposure was more frequently associated with lower placental 11-ß HSD2 levels. PPD and placental 11-ß HSD2 were associated with changes in cortisol reactivity and the development of adverse health outcomes in mothers and children. Female-offspring were more vulnerable to PPD exposures. CONCLUSION: The study presents evidence of a modest role of prenatal psychological distress in regulating placental 11-ß HSD2 gene expression. Future prospective cohorts utilizing larger sample sizes or advanced statistical methods to enhance the detection of small effect sizes should be planned. Additionally, controlling for key predictors such as the mother's ethnicity, trimester of PPD exposure, mode of delivery, and infant sex is crucial for valid exploration of PPD effects on fetal programming.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 2 , Placenta , Pregnancy Complications , Psychological Distress , Stress, Psychological , Humans , Pregnancy , 11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 2/metabolism , Female , Placenta/metabolism , Stress, Psychological/metabolism , Stress, Psychological/genetics , Pregnancy Complications/genetics , Pregnancy Complications/metabolism , Pregnancy Complications/psychology , Depression/genetics , Depression/metabolism , Gene Expression/genetics , Anxiety/genetics , Anxiety/metabolism , Hydrocortisone/metabolism , Prenatal Exposure Delayed Effects/genetics , Prenatal Exposure Delayed Effects/metabolism
8.
Psychoneuroendocrinology ; 165: 107044, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38657342

ABSTRACT

BACKGROUND: Prenatal glucocorticoids are one of the most widely proposed prenatal programming mechanisms, yet few studies exist that measure fetal cortisol via neonatal hair. Neonatal hair provides a window into the fetal experience and represents cortisol accumulation in the third trimester of pregnancy. In the current study, we test the links between two types of anxiety over the course of gestation (pregnancy-related anxiety and general anxiety) with neonatal hair cortisol. METHOD: Pregnant individuals (N = 107) and their neonates (59.8% female) participated in the current study. Prenatal pregnancy-related anxiety and general anxiety were measured using the Pregnancy Related Anxiety Scale (PRAS) and the State-Trait Anxiety Inventory (STAI), in each trimester of pregnancy. Hierarchical linear modeling was used to model the intercept and slope of each type of anxiety over gestation. Neonatal hair samples were collected shortly after birth (Median days = 1.17, IQR = 0.75-2.00). RESULTS: Both higher pregnancy-related anxiety and general anxiety at the beginning of pregnancy and a flatter decline of pregnancy-related anxiety over gestation were associated with lower neonatal hair cortisol. After inclusion of gestational age at birth and parity as covariates, pregnancy-related anxiety (intercept: ß = -0.614, p =.012; slope: ß = -0.681, p =.006), but not general anxiety (intercept: ß = -0.389, p =.114; slope: ß = -0.302, p =.217) remained a significant predictor. Further, when both general and pregnancy-related anxiety were entered into the same model, only pregnancy-related anxiety (intercept and slope) were significant predictors of neonatal hair cortisol, indicating an association with pregnancy-related anxiety above and beyond general anxiety. CONCLUSION: Cortisol plays a central role in maturation of fetal organ systems, and at the end of gestation, higher cortisol has beneficial effects such as promoting fetal lung maturation. Further, lower maternal cortisol is linked to less optimal cognitive development and altered brain development. As maternal higher anxiety in early pregnancy and a flatter decrease over time are both associated with lower neonatal hair cortisol, maternal pregnancy-related anxiety could be a target of future intervention efforts.


Subject(s)
Anxiety , Hair , Hydrocortisone , Humans , Female , Hair/chemistry , Pregnancy , Hydrocortisone/analysis , Hydrocortisone/metabolism , Anxiety/metabolism , Infant, Newborn , Adult , Gestational Age , Pregnancy Complications/metabolism , Male , Pregnancy Trimester, Third/metabolism
9.
Front Endocrinol (Lausanne) ; 15: 1314214, 2024.
Article in English | MEDLINE | ID: mdl-38495790

ABSTRACT

Successful pregnancy requires the tolerance of the maternal immune system for the semi-allogeneic embryo, as well as a synchrony between the receptive endometrium and the competent embryo. The annexin family belongs to calcium-regulated phospholipid-binding protein, which functions as a membrane skeleton to stabilize the lipid bilayer and participate in various biological processes in humans. There is an abundance of the annexin family at the maternal-fetal interface, and it exerts a crucial role in embryo implantation and the subsequent development of the placenta. Altered expression of the annexin family and dysfunction of annexin proteins or polymorphisms of the ANXA gene are involved in a range of pregnancy complications. In this review, we summarize the current knowledge of the annexin A protein family at the maternal-fetal interface and its association with female reproductive disorders, suggesting the use of ANXA as the potential therapeutic target in the clinical diagnosis and treatment of pregnancy complications.


Subject(s)
Embryo Implantation , Pregnancy Complications , Pregnancy , Female , Humans , Embryo Implantation/genetics , Placenta/metabolism , Endometrium/metabolism , Pregnancy Complications/genetics , Pregnancy Complications/metabolism , Annexins/genetics , Annexins/metabolism
10.
Nature ; 628(8006): 130-138, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38448586

ABSTRACT

Genome-wide association analyses using high-throughput metabolomics platforms have led to novel insights into the biology of human metabolism1-7. This detailed knowledge of the genetic determinants of systemic metabolism has been pivotal for uncovering how genetic pathways influence biological mechanisms and complex diseases8-11. Here we present a genome-wide association study for 233 circulating metabolic traits quantified by nuclear magnetic resonance spectroscopy in up to 136,016 participants from 33 cohorts. We identify more than 400 independent loci and assign probable causal genes at two-thirds of these using manual curation of plausible biological candidates. We highlight the importance of sample and participant characteristics that can have significant effects on genetic associations. We use detailed metabolic profiling of lipoprotein- and lipid-associated variants to better characterize how known lipid loci and novel loci affect lipoprotein metabolism at a granular level. We demonstrate the translational utility of comprehensively phenotyped molecular data, characterizing the metabolic associations of intrahepatic cholestasis of pregnancy. Finally, we observe substantial genetic pleiotropy for multiple metabolic pathways and illustrate the importance of careful instrument selection in Mendelian randomization analysis, revealing a putative causal relationship between acetone and hypertension. Our publicly available results provide a foundational resource for the community to examine the role of metabolism across diverse diseases.


Subject(s)
Biomarkers , Genome-Wide Association Study , Metabolomics , Female , Humans , Pregnancy , Acetone/blood , Acetone/metabolism , Biomarkers/blood , Biomarkers/metabolism , Cholestasis, Intrahepatic/blood , Cholestasis, Intrahepatic/genetics , Cholestasis, Intrahepatic/metabolism , Cohort Studies , Genome-Wide Association Study/methods , Hypertension/blood , Hypertension/genetics , Hypertension/metabolism , Lipoproteins/genetics , Lipoproteins/metabolism , Magnetic Resonance Spectroscopy , Mendelian Randomization Analysis , Metabolic Networks and Pathways/genetics , Phenotype , Polymorphism, Single Nucleotide/genetics , Pregnancy Complications/blood , Pregnancy Complications/genetics , Pregnancy Complications/metabolism
11.
Front Immunol ; 15: 1362784, 2024.
Article in English | MEDLINE | ID: mdl-38545107

ABSTRACT

Due to the physiological alteration during pregnancy, maternal gut microbiota changes following the metabolic processes. Recent studies have revealed that maternal gut microbiota is closely associated with the immune microenvironment in utero during pregnancy and plays a vital role in specific pregnancy complications, including preeclampsia, gestational diabetes, preterm birth and recurrent miscarriages. Some other evidence has also shown that aberrant maternal gut microbiota increases the risk of various diseases in the offspring, such as allergic and neurodevelopmental disorders, through the immune alignment between mother and fetus and the possible intrauterine microbiota. Probiotics and the high-fiber diet are effective inventions to prevent mothers and fetuses from diseases. In this review, we summarize the role of maternal gut microbiota in the development of pregnancy complications and the health condition of future generations from the perspective of immunology, which may provide new therapeutic strategies for the health management of mothers and offspring.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Pregnancy Complications , Premature Birth , Pregnancy , Female , Humans , Infant, Newborn , Mothers , Pregnancy Complications/metabolism
12.
Arch Gynecol Obstet ; 309(6): 2279-2288, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38494514

ABSTRACT

The prevalence of maternal obesity rapidly increases, which represents a major public health concern worldwide. Maternal obesity is characteristic by metabolic dysfunction and chronic inflammation. It is associated with health problems in both mother and offspring. Increasing evidence indicates that the placenta is an axis connecting maternal obesity with poor outcomes in the offspring. In this brief review, we have summarized the current data regarding deregulated placental function in maternal obesity. The data show that maternal obesity induces numerous placental defects, including lipid and glucose metabolism, stress response, inflammation, immune regulation and epigenetics. These placental defects affect each other and result in a stressful intrauterine environment, which transduces and mediates the adverse effects of maternal obesity to the fetus. Further investigations are required to explore the exact molecular alterations in the placenta in maternal obesity, which may pave the way to develop specific interventions for preventing epigenetic and metabolic programming in the fetus.


Subject(s)
Obesity, Maternal , Placenta , Humans , Pregnancy , Female , Placenta/metabolism , Obesity, Maternal/metabolism , Epigenesis, Genetic , Maternal-Fetal Exchange , Inflammation/metabolism , Placenta Diseases/physiopathology , Placenta Diseases/metabolism , Pregnancy Complications/metabolism , Pregnancy Complications/physiopathology , Obesity/metabolism , Obesity/physiopathology
13.
Brain Behav Immun ; 119: 146-153, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38555986

ABSTRACT

BACKGROUND: Perinatal depression (including antenatal-, postnatal-, and depression that spans both timepoints) is a prevalent disorder with high morbidity that affects both mother and child. Even though the full biological blueprints of perinatal depression remain incomplete, multiple studies indicate that, at least for antenatal depression, the disorder has an inflammatory component likely linked to a dysregulation of the enzymatic kynurenine pathway. The production of neuroactive metabolites in this pathway, including quinolinic acid (QUIN), is upregulated in the placenta due to the multiple immunological roles of the metabolites during pregnancy. Since neuroactive metabolites produced by the pathway also may affect mood by directly affecting glutamate neurotransmission, we sought to investigate whether the placental expression of kynurenine pathway enzymes controlling QUIN production was associated with both peripheral inflammation and depressive symptoms during pregnancy. METHODS: 68 placentas obtained at birth were analyzed using qPCR to determine the expression of kynurenine pathway enzymes. Cytokines and metabolites were quantified in plasma using high-sensitivity electroluminescence and ultra-performance liquid chromatography, respectively. Maternal depressive symptoms were assessed using the Edinburgh Postnatal Depression Scale (EPDS) throughout pregnancy and the post-partum. Associations between these factors were assessed using robust linear regression with ranked enzymes. RESULTS: Low placental quinolinate phosphoribosyl transferase (QPRT), the enzyme responsible for degrading QUIN, was associated with higher IL-6 and higher QUIN/kynurenic acid ratios at the 3rd trimester. Moreover, women with severe depressive symptoms in the 3rd trimester had significantly lower placental expression of both QPRT and 2-amino-3-carboxymuconate-6-semialdehyde decarboxylase (ACMSD); impaired activity of these two enzymes leads to QUIN accumulation. CONCLUSION: Overall, our data support that a compromised placental environment, featuring low expression of critical kynurenine pathway enzymes is associated with increased levels of plasma cytokines and the dysregulated kynurenine metabolite pattern observed in depressed women during pregnancy.


Subject(s)
Depression , Inflammation , Kynurenine , Placenta , Quinolinic Acid , Humans , Female , Pregnancy , Kynurenine/metabolism , Kynurenine/blood , Placenta/metabolism , Adult , Inflammation/metabolism , Depression/metabolism , Quinolinic Acid/metabolism , Quinolinic Acid/blood , Cytokines/metabolism , Pregnancy Complications/metabolism , Carboxy-Lyases/metabolism , Pentosyltransferases
14.
Placenta ; 148: 20-30, 2024 Mar 25.
Article in English | MEDLINE | ID: mdl-38346375

ABSTRACT

BACKGROUND: Abnormal bile acid metabolism leading to changes in placental function during pregnancy. To determine whether endoplasmic reticulum protein 29 (ERp29) can mediate the pregnancy effects of cholestasis by altering the level of trophoblast cell apoptosis. METHODS: ERp29 in serum of 66 intrahepatic cholestasis of pregnancy (ICP) pregnant women and 74 healthy were detected by ELISA. Subcutaneous injection of ethinyl estradiol (E2) was used to induce ICP in pregnant rats. Taurocholic acid (TCA) was used to simulate the ICP environment, and TGF-ß1 was added to induce the epithelial mesenchymal transformation (EMT) process. The scratch, migration, and invasion test were used to detect the EMT process. ERp29 overexpression/knockdown vector were constructed and transfected to verify the role of ERp29 in the EMT process. Downstream gene was obtained through RNA-seq. RESULTS: Compared with the healthy pregnant women, the expression levels of ERp29 in serum of ICP pregnancy women were significantly increased (P < 0.001). ERp29 in the placenta tissue of the ICP pregnant rats increased significantly, and the level of apoptosis increased. The placental tissues of the ICP had high expression of E-cadherin and low expression of N-cadherin, snail1, vimentin. After HTR-8/SVneo cells were induced by TCA, EMT was inhibited, while the ERp29 increased. Cell and animal experiments showed that, knockdown of ERp29 reduced the inhibition of EMT, the ICP progress was alleviated. Overexpression of FOS salvaged the inhibitory effects of ERp29 on cell EMT. DISCUSSION: The high level of ERp29 in placental trophoblast cells reduced FOS mRNA levels, inhibited the EMT process and aggravated the occurrence and development of ICP.


Subject(s)
Cholestasis, Intrahepatic , Pregnancy Complications , Female , Pregnancy , Humans , Rats , Animals , Placenta/metabolism , Trophoblasts/metabolism , Pregnancy Complications/metabolism , Cholestasis, Intrahepatic/genetics , Cholestasis, Intrahepatic/metabolism , Taurocholic Acid/metabolism , Taurocholic Acid/pharmacology , Apoptosis/physiology , Epithelial-Mesenchymal Transition/physiology
15.
Commun Biol ; 7(1): 227, 2024 Feb 24.
Article in English | MEDLINE | ID: mdl-38402336

ABSTRACT

Endometriosis is linked to increased infertility and pregnancy complications due to defective endometrial decidualization. We hypothesized that identification of altered signaling pathways during decidualization could identify the underlying cause of infertility and pregnancy complications. Our study reveals that transforming growth factor ß (TGFß) pathways are impaired in the endometrium of individuals with endometriosis, leading to defective decidualization. Through detailed transcriptomic analyses, we discovered abnormalities in TGFß signaling pathways and key regulators, such as SMAD4, in the endometrium of affected individuals. We also observed compromised activity of bone morphogenetic proteins (BMP), a subset of the TGFß family, that control endometrial receptivity. Using 3-dimensional models of endometrial stromal and epithelial assembloids, we showed that exogenous BMP2 improved decidual marker expression in individuals with endometriosis. Our findings reveal dysfunction of BMP/SMAD signaling in the endometrium of individuals with endometriosis, explaining decidualization defects and subsequent pregnancy complications in these individuals.


Subject(s)
Endometriosis , Infertility , Pregnancy Complications , Pregnancy , Female , Humans , Endometriosis/genetics , Endometriosis/metabolism , Decidua/metabolism , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Transforming Growth Factor beta/metabolism , Signal Transduction , Infertility/metabolism , Pregnancy Complications/metabolism
16.
Curr Opin Lipidol ; 35(3): 133-140, 2024 06 01.
Article in English | MEDLINE | ID: mdl-38408036

ABSTRACT

PURPOSE OF REVIEW: Accommodating fetal growth and development, women undergo multiple physiological changes during pregnancy. In recent years, several studies contributed to the accumulating evidence about the impact of gestational hyperlipidemia on cardiovascular risk for mother and child. This review aims to provide a comprehensive overview of the current research on lipid profile alterations during pregnancy and its associated (cardiovascular) outcomes for mother and child from a clinical perspective. RECENT FINDINGS: In a normal pregnancy, total and LDL-cholesterol levels increase by approximately 30-50%, HDL-cholesterol by 20-40%, and triglycerides by 50-100%. In some women, for example, with familial hypercholesterolemia (FH), a more atherogenic lipid profile is observed. Dyslipidemia during pregnancy is found to be associated with adverse (cardiovascular) outcomes for the mother (e.g. preeclampsia, gestational diabetes, metabolic syndrome, unfavorable lipid profile) and for the child (e.g. preterm birth, large for gestational age, preatherosclerotic lesions, unfavorable lipid profile). SUMMARY: The lipid profile of women during pregnancy provides a unique window of opportunity into the potential future cardiovascular risk for mother and child. Better knowledge about adverse outcomes and specific risk groups could lead to better risk assessment and earlier cardiovascular prevention. Future research should investigate implementation of gestational screening possibilities.


Subject(s)
Lipid Metabolism , Humans , Pregnancy , Female , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/blood , Pregnancy Complications/metabolism , Pregnancy Complications/blood , Child , Lipids/blood
17.
Dig Dis Sci ; 69(4): 1253-1262, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38361148

ABSTRACT

BACKGROUND: Intrahepatic cholestasis of pregnancy (ICP) is associated with adverse pregnancy outcomes; however, the underlying mechanisms are not fully understood. AIMS: This study aimed to determine the role of exosomal miR-6891-5p in placental trophoblast dysfunction in ICP and identify new biomarkers for ICP diagnosis. METHODS: Serum samples were collected from ICP patients and healthy pregnant women, and serum exosomes were extracted and identified. Fluorescent dye labeling of exosomes and cell-verified cell phagocytosis were performed. In vitro experiments were conducted by adding taurocholic acid to simulate the ICP environment. Cell proliferation and apoptosis levels were detected using flow cytometry and the cell counting kit-8 assay. Mimics were constructed to overexpress miR-6891-5p in cells, and the binding site between miR-6891-5p and YWHAE was verified using luciferase reporter genes. RESULTS: miR-6891-5p expression was significantly decreased in serum exosomes of ICP patients. Co-culturing with exosomes derived from ICP patients' serum (ICP-Exos) decreased HTR-8/SVeno cell proliferation and increased apoptosis levels. miR-6891-5p upregulation in HTR-8/SVeno cells significantly increased cell viability and reduced cell apoptosis levels, as determined by the cell counting kit-8 assay and flow cytometry. A double luciferase assay confirmed that miR-6891-5p affected the expression of the downstream YWHAE protein. CONCLUSIONS: This study indicates that serum exosomes from ICP patients can impact the apoptosis of placental trophoblast HTR-8/SVeno cells through the miR-6891-5P/YWHAE pathway and can serve as specific molecular markers for ICP diagnosis.


Subject(s)
Cholestasis, Intrahepatic , Exosomes , MicroRNAs , Pregnancy Complications , Female , Humans , Pregnancy , 14-3-3 Proteins/metabolism , Apoptosis , Cell Proliferation , Cholestasis, Intrahepatic/genetics , Cholestasis, Intrahepatic/metabolism , Exosomes/genetics , Luciferases/metabolism , MicroRNAs/blood , MicroRNAs/genetics , MicroRNAs/metabolism , Placenta/metabolism , Pregnancy Complications/genetics , Pregnancy Complications/metabolism
18.
Arch Womens Ment Health ; 27(4): 577-584, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38308143

ABSTRACT

PURPOSE: Maternal cortisol levels in pregnancy may support the growth of or adversely affect fetal organs, including the brain. While moderate cortisol levels are essential for fetal development, excessive or prolonged elevations may have negative health consequences for both the mother and the offspring. Little is known about predictors of altered hypothalamic-pituitary-adrenal (HPA) axis activity during pregnancy. This study examined maternal hair cortisol concentration (HCC) in the 3rd trimester of pregnancy in relation to severe psychopathology. METHODS: Hair samples were collected from 69 women, 32 with a lifetime diagnosis of severe mental disorders (bipolar I or II disorder, moderate or severe depressive disorder, schizophrenic spectrum disorder), and 37 non-clinical controls. Hair samples were collected during the 3rd trimester, and liquid chromatography tandem mass spectrometry was used for cortisol assessment. Psychiatric diagnosis and current level of symptomatic functioning were assessed using the structured clinical interview from the DSM-5 and the global assessment of functioning scale. RESULTS: Women with a lifetime diagnosis of severe mental illness had significantly elevated HCC compared to controls. Poorer current symptomatic functioning was also significantly associated with elevated HCC in pregnancy. CONCLUSIONS: The implications of alterations in HCC on both maternal and infant health need further study.


Subject(s)
Bipolar Disorder , Hair , Hydrocortisone , Schizophrenia , Humans , Female , Hydrocortisone/analysis , Hydrocortisone/metabolism , Pregnancy , Hair/chemistry , Adult , Schizophrenia/metabolism , Bipolar Disorder/metabolism , Bipolar Disorder/psychology , Pregnancy Trimester, Third , Pregnancy Complications/psychology , Pregnancy Complications/metabolism , Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/metabolism , Case-Control Studies , Depressive Disorder/metabolism , Depressive Disorder/psychology , Young Adult
19.
Trends Endocrinol Metab ; 35(7): 638-647, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38418281

ABSTRACT

The obesity epidemic has led to a growing body of research investigating the consequences of maternal obesity on pregnancy and offspring health. The placenta, traditionally viewed as a passive intermediary between mother and fetus, is known to play a critical role in modulating the intrauterine environment and fetal development, and we now know that maternal obesity leads to increased inflammation, oxidative stress, and altered placental function. Here, we review recent research exploring the involvement of inflammation and oxidative stress as mechanisms impacting the placenta and fetus during obese pregnancy. Understanding them is crucial for informing strategies that can mitigate the adverse health effects of maternal obesity on offspring development and disease risk.


Subject(s)
Inflammation , Obesity, Maternal , Oxidative Stress , Placenta , Humans , Pregnancy , Female , Oxidative Stress/physiology , Placenta/metabolism , Obesity, Maternal/metabolism , Inflammation/metabolism , Obesity/metabolism , Fetal Development/physiology , Animals , Pregnancy Complications/metabolism
20.
Biometals ; 37(4): 943-953, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38367126

ABSTRACT

Trace elements are important components in the body and have fundamental roles in maintaining a healthy and balanced pregnancy process. Either deficiency or excess of trace elements, including selenium, iron, zinc, copper, and magnesium can lead to pregnancy complications. As a rare disorder during pregnancy of unknown aetiology, intrahepatic cholestasis of pregnancy (ICP) poses a significant risk to the fetus of perinatal mortality. ICP is a multifactorial complication of which the pathogenesis is still an enigma. Epidemiological studies have demonstrated the association of ICP with some trace elements. Evidence from retrospective studies in humans further revealed the possible contributing roles of trace elements in the pathogenesis of ICP. The published literature on the association of trace elements with ICP was reviewed. Recent advances in molecular biological techniques from animal studies have helped to elucidate the possible mechanisms by how these trace elements function in regulating oxidative reactions, inflammatory reactions and immune balance in the maternal-fetal interface, as well as the influence on hepato-intestinal circulation of bile acid. The scenario regarding the role of trace elements in the pathogenesis of ICP is still developing. The administration or depletion of these trace elements may have promising effects in alleviating the symptoms and improving the pregnancy outcomes of ICP.


Subject(s)
Cholestasis, Intrahepatic , Pregnancy Complications , Trace Elements , Cholestasis, Intrahepatic/metabolism , Humans , Female , Pregnancy Complications/metabolism , Pregnancy , Trace Elements/metabolism , Animals , Zinc/metabolism , Zinc/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...