107 results on '"Plösch T"'
Search Results
2. Review identifies odorants used in neonatal clinical care but identifies need for further research.
- Author
-
Hinz C, Heep A, Plösch T, Huber J, and Lange M
- Subjects
- Humans, Infant, Newborn, Review Literature as Topic, Odorants
- Published
- 2024
- Full Text
- View/download PDF
3. Visualizing multimerization of plasticity-related gene 5 at the plasma membrane using FLIM-FRET.
- Author
-
Köper F, Vonk D, Dirksen MW, Gross I, Heep A, Plösch T, Hipp MS, and Bräuer AU
- Abstract
Plasticity-related gene (PRG) 5 is a vertebrate specific membrane protein, that belongs to the family of lipid-phosphate phosphatases (LPPs). It is prominently expressed in neurons and is involved in cellular processes such as growth-cone guidance and spine formation. At a functional level, PRG5 induces filopodia in non-neuronal cell lines, as well as the formation of plasma membrane protrusions in primary cortical and hippocampal neurons. Overexpression of PRG5 in immature neurons leads to the induction of spine-like structures, and regulates spine density and morphology in mature neurons. Understanding spine formation is pivotal, as spine abnormalities are associated with numerous neurological disorders. Although the importance of PRG5 in neuronal function is evident, the precise mechanisms as to how exactly it induces membrane protrusions and orchestrates cellular processes remain unresolved. Here we used in vitro biochemical assays to demonstrate that in HEK293T cells a large fraction of PRG5 can be found in homo dimers and lager multimers. By using Fluorescence Lifetime Imaging (FLIM) to quantify Förster Resonance Energy Transfer (FRET), we were able to visualize and quantify the specific localization of PRG5 multimers in living HEK293T cells and in fixed immature primary hippocampal neurons. Here, we provide the first evidence that PRG5 multimers are specifically localized in non-neuronal filopodia, as well as in neuronal spine-like structures. Our findings indicate a potential functional role for PRG5 multimerization, which might be required for interaction with extracellular matrix molecules or for maintaining the stability of membrane protrusions., Competing Interests: Author IG was employed by PicoQuant GmbH. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2024 Köper, Vonk, Dirksen, Gross, Heep, Plösch, Hipp and Bräuer.)
- Published
- 2024
- Full Text
- View/download PDF
4. The fetal programming effect of maternal immune activation (MIA) on the offspring's immune system.
- Author
-
Hofsink N, Groenink L, and Plösch T
- Subjects
- Humans, Pregnancy, Female, Animals, SARS-CoV-2 immunology, Epigenesis, Genetic, Cytokines metabolism, Neurodevelopmental Disorders immunology, Neurodevelopmental Disorders etiology, Maternal Exposure adverse effects, Infant, Newborn, Prenatal Exposure Delayed Effects immunology, Immune System immunology, Immune System metabolism, Fetal Development immunology, COVID-19 immunology
- Abstract
The first 1000 days of life is a critical period of development in which adverse circumstances can have long-term consequences for the child's health. Maternal immune activation is associated with increased risk of neurodevelopmental disorders in the child. Aberrant immune responses have been reported in individuals with neurodevelopmental disorders. Moreover, lasting effects of maternal immune activation on the offspring's immune system have been reported. Taken together, this indicates that the effect of maternal immune activation is not limited to the central nervous system. Here, we explore the impact of maternal immune activation on the immune system of the offspring. We first describe the development of the immune system and provide an overview of reported alterations in the cytokine profiles, immune cell profiles, immune cell function, and immune induction in pre-clinical models. Additionally, we highlight recent research on the impact of maternal COVID-19 exposure on the neonatal immune system and the potential health consequences for the child. Our review shows that maternal immune activation alters the offspring's immune system under certain conditions, but the reported effects are conflicting and inconsistent. In general, epigenetic modifications are considered the mechanism for fetal programming. The available data was insufficient to identify specific pathways that may contribute to immune programming. As a consequence of the COVID-19 pandemic, more research now focuses on the possible health effects of maternal immune activation on the offspring. Future research addressing the offspring's immune response to maternal immune activation can elucidate specific pathways that contribute to fetal immune programming and the long-term health effects for the offspring., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
5. Epigenome-wide association study on the plasma metabolome suggests self-regulation of the glycine and serine pathway through DNA methylation.
- Author
-
Wu J, Palasantzas V, Andreu-Sánchez S, Plösch T, Leonard S, Li S, Bonder MJ, Westra HJ, van Meurs J, Ghanbari M, Franke L, Zhernakova A, Fu J, Hoogerland JA, and Zhernakova DV
- Subjects
- Humans, Male, Female, Middle Aged, CpG Islands genetics, Epigenome genetics, Adult, Aged, Mendelian Randomization Analysis, Glycine blood, Serine blood, Serine genetics, DNA Methylation genetics, Genome-Wide Association Study methods, Metabolome genetics, Epigenesis, Genetic genetics
- Abstract
Background: The plasma metabolome reflects the physiological state of various biological processes and can serve as a proxy for disease risk. Plasma metabolite variation, influenced by genetic and epigenetic mechanisms, can also affect the cellular microenvironment and blood cell epigenetics. The interplay between the plasma metabolome and the blood cell epigenome remains elusive. In this study, we performed an epigenome-wide association study (EWAS) of 1183 plasma metabolites in 693 participants from the LifeLines-DEEP cohort and investigated the causal relationships in DNA methylation-metabolite associations using bidirectional Mendelian randomization and mediation analysis., Results: After rigorously adjusting for potential confounders, including genetics, we identified five robust associations between two plasma metabolites (L-serine and glycine) and three CpG sites located in two independent genomic regions (cg14476101 and cg16246545 in PHGDH and cg02711608 in SLC1A5) at a false discovery rate of less than 0.05. Further analysis revealed a complex bidirectional relationship between plasma glycine/serine levels and DNA methylation. Moreover, we observed a strong mediating role of DNA methylation in the effect of glycine/serine on the expression of their metabolism/transport genes, with the proportion of the mediated effect ranging from 11.8 to 54.3%. This result was also replicated in an independent population-based cohort, the Rotterdam Study. To validate our findings, we conducted in vitro cell studies which confirmed the mediating role of DNA methylation in the regulation of PHGDH gene expression., Conclusions: Our findings reveal a potential feedback mechanism in which glycine and serine regulate gene expression through DNA methylation., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
6. Fetal growth restriction induced by maternal gal-3 deficiency is associated with altered gut-placenta axis.
- Author
-
Xie Y, Zhao F, Wang Y, Borowski S, Freitag N, Tirado-Gonzalez I, Hofsink N, Matschl U, Plösch T, Garcia MG, and Blois SM
- Subjects
- Pregnancy, Female, Animals, Mice, Male, Gastrointestinal Microbiome, Mice, Inbred C57BL, Humans, Fetal Development, Insulin-Like Growth Factor II metabolism, Insulin-Like Growth Factor II genetics, Insulin-Like Growth Factor II deficiency, Trophoblasts metabolism, Fetal Growth Retardation metabolism, Fetal Growth Retardation genetics, Placenta metabolism, Galectin 3 metabolism, Galectin 3 deficiency, Galectin 3 genetics
- Abstract
Adverse intrauterine conditions may cause fetal growth restriction (FGR), a pregnancy complication frequently linked to perinatal morbidity and mortality. Although many studies have focused on FGR, the pathophysiological processes underlying this disorder are complex and incompletely understood. We have recently determined that galectin-3 (gal-3), a β-galactoside-binding protein, regulates pregnancy-associated processes, including uterine receptibility, maternal vascular adaptation and placentation. Because gal-3 is expressed at both sides of the maternal-fetal interface, we unraveled the contribution of maternal- and paternal-derived gal-3 on fetal-placental development in the prenatal window and its effects on the post-natal period. Deficiency of maternal gal-3 induced maternal gut microbiome dysbiosis, resulting in a sex-specific fetal growth restriction mainly observed in female fetuses and offspring. In addition, poor placental metabolic adaptions (characterized by decreased trophoblast glycogen content and insulin-like growth factor 2 (Igf2) gene hypomethylation) were only associated with a lack of maternal-derived gal-3. Paternal gal-3 deficiency caused compromised vascularization in the placental labyrinth without affecting fetal growth trajectory. Thus, maternal-derived gal-3 may play a key role in fetal-placental development through the gut-placenta axis., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
7. Placental glycosylation senses the anti-angiogenic milieu induced by human sFLT1 during pregnancy.
- Author
-
Kirkgöz K, Vogtmann R, Xie Y, Zhao F, Riedel A, Adam LM, Freitag N, Harms C, Garcia MG, Plösch T, Gellhaus A, and Blois SM
- Subjects
- Pregnancy, Female, Animals, Humans, Mice, Glycosylation, Galectins metabolism, Neovascularization, Pathologic metabolism, Disease Models, Animal, Vascular Endothelial Growth Factor Receptor-1 metabolism, Pre-Eclampsia metabolism, Placenta metabolism
- Abstract
Abnormal placental angiogenesis during gestation resulting from high levels of anti-angiogenic factors, soluble fms-like tyrosine kinase-1 (sFLT1) and soluble endoglin, has been implicated in the progression of preeclampsia (PE). This heterogeneous syndrome (defined by hypertension with or without proteinuria after 20 weeks of pregnancy) remains a major global health burden with long-term consequences for both mothers and child. Previously, we showed that in vivo systemic human (hsFLT1) overexpression led to reduced placental efficiency and PE-like syndrome in mice. Galectins (gal-1, -3 and -9) are critical determinants of vascular adaptation to pregnancy and dysregulation of the galectin-glycan circuits is associated with the development of this life-threatening disease. In this study, we assessed the galectin-glycan networks at the maternal-fetal interface associated with the hsFLT1-induced PE in mice. We observed an increase on the maternal gal-1 expression in the decidua and junctional zone layers of the placenta derived from hs FLT1
high pregnancies. In contrast, placental gal-3 and gal-9 expression were not sensitive to the hsFLT1 overexpression. In addition, O- and N-linked glycan expression, poly-LacNAc sequences and terminal sialylation were down-regulated in hsFLT1high placentas. Thus, the gal-1-glycan axis appear to play an important role counteracting the anti-angiogenic status caused by sFLT1, becoming critical for vascular adaptation at the maternal-fetal interface., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024. Published by Elsevier B.V.)- Published
- 2024
- Full Text
- View/download PDF
8. The role of mesenchymal stem cells in early programming of adipose tissue in the offspring of women with obesity.
- Author
-
Bellalta S, Plösch T, Faas M, and Casanello P
- Subjects
- Humans, Female, Pregnancy, Adipose Tissue, Pediatric Obesity, Adipogenesis physiology, Infant, Newborn, Adipocytes, Mesenchymal Stem Cells, Obesity, Maternal metabolism, Prenatal Exposure Delayed Effects
- Abstract
Maternal obesity is a well-known risk factor for developing premature obesity, metabolic syndrome, cardiovascular disease and type 2 diabetes in the progeny. The development of white adipose tissue is a dynamic process that starts during prenatal life: fat depots laid down in utero are associated with the proportion of fat in children later on. How early this programming takes place is still unknown. However, recent evidence shows that mesenchymal stem cells (MSC), the embryonic adipocyte precursor cells, show signatures of the early setting of an adipogenic committed phenotype when exposed to maternal obesity. This review aims to present current findings on the cellular adaptations of MSCs from the offspring of women with obesity and how the metabolic environment of MSCs could affect the early commitment towards adipocytes. In conclusion, maternal obesity can induce early programming of fetal adipose tissue by conditioning MSCs. These cells have higher expression of adipogenic markers, altered insulin signalling and mitochondrial performance, compared to MSCs of neonates from lean pregnancies. Fetal MSCs imprinting by maternal obesity could help explain the increased risk of childhood obesity and development of further noncommunicable diseases., (© 2024 The Authors. Pediatric Obesity published by John Wiley & Sons Ltd on behalf of World Obesity Federation.)
- Published
- 2024
- Full Text
- View/download PDF
9. Sex-Specific Transcriptomic Changes in the Villous Tissue of Placentas of Pregnant Women Using a Selective Serotonin Reuptake Inhibitor.
- Author
-
Staal L, Plösch T, Kunovac Kallak T, Sundström Poromaa I, Wertheim B, and Olivier JDA
- Subjects
- Infant, Female, Humans, Male, Pregnancy, Fluoxetine pharmacology, Fluoxetine therapeutic use, Pregnant Women, Transcriptome, Placenta metabolism, Gene Expression Profiling, Selective Serotonin Reuptake Inhibitors pharmacology, Selective Serotonin Reuptake Inhibitors therapeutic use, Prenatal Exposure Delayed Effects metabolism
- Abstract
About 5% of pregnant women are treated with selective serotonin reuptake inhibitor (SSRI) antidepressants to treat their depression. SSRIs influence serotonin levels, a key factor in neural embryonic development, and their use during pregnancy has been associated with adverse effects on the developing embryo. However, the role of the placenta in transmitting these negative effects is not well understood. In this study, we aim to elucidate how disturbances in the maternal serotonergic system affect the villous tissue of the placenta by assessing whole transcriptomes in the placentas of women with healthy pregnancies and women with depression and treated with the SSRI fluoxetine during pregnancy. Twelve placentas of the Biology, Affect, Stress, Imaging and Cognition in Pregnancy and the Puerperium (BASIC) project were selected for RNA sequencing to examine differentially expressed genes: six male infants and six female infants, equally distributed over women treated with SSRI and without SSRI treatment. Our results show that more genes in the placenta of male infants show changed expression associated with fluoxetine treatment than in placentas of female infants, stressing the importance of sex-specific analyses. In addition, we identified genes related to extracellular matrix organization to be significantly enriched in placentas of male infants born to women treated with fluoxetine. It remains to be established whether the differentially expressed genes that we found to be associated with SSRI treatment are the result of the SSRI treatment itself, the underlying depression, or a combination of the two.
- Published
- 2024
- Full Text
- View/download PDF
10. Neonatal stress exposure and DNA methylation of stress-related and neurodevelopmentally relevant genes: An exploratory study.
- Author
-
van Dokkum NH, Bao M, Verkaik-Schakel RN, Reijneveld SA, Bos AF, de Kroon MLA, and Plösch T
- Subjects
- Infant, Infant, Newborn, Humans, Prospective Studies, Infant, Extremely Premature, Gestational Age, Intensive Care Units, Neonatal, Serotonin Plasma Membrane Transport Proteins genetics, DNA Methylation, Brain-Derived Neurotrophic Factor genetics
- Abstract
Background: Stress exposure during Neonatal Intensive Care Unit (NICU) stay may have long-lasting effects on neurodevelopmental outcomes in extremely preterm infants. Altered DNA methylation of stress-related and neurodevelopmentally relevant genes may be an underlying mechanism., Aims: This exploratory study aimed to investigate the association between neonatal stress exposure and DNA methylation in these genes at two different time points: early during the NICU stay (7-14 days after birth) and later, at discharge from the NICU., Subjects: We included 45 extremely preterm infants in this prospective cohort study, gestational age 24-30 weeks., Outcome Measures: We collected fecal samples at days 7-14 (n = 44) and discharge (n = 28) and determined DNA methylation status in predefined regions of NR3C1, SLC6A4, HSD11B2, OPRM1, SLC7A5, SLC1A2, IGF2, NNAT, BDNF and GABRA6 using pyrosequencing. Because of low DNA concentrations in some fecal samples, we could do so in 25-50 % of collected samples. We prospectively quantified daily neonatal stress exposure using the Neonatal Infant Stressor Scale (NISS) and explored associations between cumulative NISS scores and average DNA methylation status., Results: Rates of methylation of most genes were not statistically different between day 7-14 and discharge, except for OPRM1. We found moderately high and mostly negative correlation coefficients upon discharge with the cumulative NISS for the NR3C1, SLC6A4, SLC1A2, IGF2, BDNF and OPRM1 genes, albeit not statistically significant., Conclusions: Findings suggest that expression of stress-related and neurodevelopmentally relevant genes may be differently regulated following higher neonatal stress exposure. Larger studies should challenge the findings of this study and ideally test the effects on gene expression., Competing Interests: Declaration of competing interest The authors have no conflict of interest to disclose., (Copyright © 2023 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
11. Gene expression in the intestine of newborn piglets after hypoxia-reoxygenation.
- Author
-
Dotinga BM, Bao M, Solberg R, Saugstad OD, Hulscher JBF, Bos AF, Plösch T, and Kooi EMW
- Subjects
- Animals, Humans, Infant, Newborn, Animals, Newborn, Gene Expression, Inflammation complications, Intestines, Oxygen, Swine, Random Allocation, Disease Models, Animal, Hypoxia, Infant, Premature
- Abstract
Background: In preterm infants, intestinal hypoxia may partly contribute to the pathophysiology of necrotizing enterocolitis through changes in gene expression. Splanchnic hypoxia can be detected with monitoring of regional splanchnic oxygen saturation (r
s SO2 ). Using a piglet model of asphyxia, we aimed to correlate changes in rs SO2 to gene expression., Methods: Forty-two newborn piglets were randomized to control or intervention groups. Intervention groups were subjected to hypoxia until they were acidotic and hypotensive. Next, they were reoxygenated for 30 min according to randomization, i.e., 21% O2 , 100% O2 , or 100% O2 for 3 min followed by 21% O2 , and observed for 9 h. We continuously measured rs SO2 and calculated mean rs SO2 and variability of rs SO2 (rs CoVar = SD/mean). Samples of terminal ileum were analyzed for mRNA expression of selected genes related to inflammation, erythropoiesis, fatty acid metabolism, and apoptosis., Results: The expression of selected genes was not significantly different between control and intervention groups. No associations between mean rs SO2 and gene expression were observed. However, lower rs CoVar was associated with the upregulation of apoptotic genes and the downregulation of inflammatory genes (P < 0.05)., Conclusion: Our study suggests that hypoxia and reoxygenation cause reduced vascular adaptability, which seems to be associated with the upregulation of apoptosis and downregulation of inflammation., Impact: Our results provide important insight into the (patho)physiological significance of changes in the variability of rs SO2 . Our findings may advance future research and clinical practice regarding resuscitation strategies of preterm infants., (© 2023. The Author(s), under exclusive licence to the International Pediatric Research Foundation, Inc.)- Published
- 2023
- Full Text
- View/download PDF
12. Prenatal Sildenafil and Fetal-placental Programming in Human Pregnancies Complicated by Fetal Growth Restriction: A Retrospective Gene Expression Analysis.
- Author
-
Terstappen F, Plösch T, Calis JJA, Ganzevoort W, Pels A, Paauw ND, Gordijn SJ, van Rijn BB, Mokry M, and Lely AT
- Abstract
Objective: Fetal growth restricted (FGR) offspring are more susceptible to develop cardiovascular and renal disease. The potential therapeutic value of sildenafil to improve fetal growth has recently been evaluated in several randomized clinical trials. Here we investigate whether administration of sildenafil during pregnancies complicated by FGR influences fetal-placental programming profiles, especially related to cardiorenal development and disease., Methods: We collected human umbilical vein endothelial cells (HUVECs) and placental tissue within the Dutch STRIDER trial, in which sildenafil versus placebo treatment were randomly assigned to pregnancies complicated by severe early-onset FGR. Differential expression of genes of these samples were studied by whole genome RNA-sequencing. In addition, we performed gene set enrichment analysis focused on cardiovascular and renal gene sets to examine differentially expressed gene sets related to cardiorenal development and health., Results: Our study showed similar gene expression profiles between treatment groups in HUVECs (n=12 sildenafil; n=8 placebo) and placentas (n=13 per group). Prenatal sildenafil exposure did not change cardiovascular or renal programming in pregnancies complicated by FGR. In placental tissue, prenatal sildenafil altered a few gene sets involved with the nitric oxide pathway potentially reflecting the mechanism of action of sildenafil. Prenatal sildenafil also upregulated gene sets related to immune pathways in placental tissue., Conclusions: Overall, our study showed that sildenafil has the potential to alter placental (but not fetal) expression of gene sets related to immune pathways and did not support (in)direct reprogramming of cardiovascular or renal health in human pregnancies complicated by FGR., Competing Interests: Conflict of interest None declared.
- Published
- 2023
- Full Text
- View/download PDF
13. The miRNA transcriptome of cerebrospinal fluid in preterm infants reveals the signaling pathways that promote reactive gliosis following cerebral hemorrhage.
- Author
-
Gialeli A, Spaull R, Plösch T, Uney J, Llana OC, and Heep A
- Abstract
Introduction: Germinal Matrix-Intraventricular Haemorrhage (GM-IVH) is one of the most common neurological complications in preterm infants, which can lead to accumulation of cerebrospinal fluid (CSF) and is a major cause of severe neurodevelopmental impairment in preterm infants. However, the pathophysiological mechanisms triggered by GM-IVH are poorly understood. Analyzing the CSF that accumulates following IVH may allow the molecular signaling and intracellular communication that contributes to pathogenesis to be elucidated. Growing evidence suggests that miRs, due to their key role in gene expression, have a significant utility as new therapeutics and biomarkers., Methods: The levels of 2,083 microRNAs (miRs) in 15 CSF samples from 10 infants with IVH were measured using miRNA whole transcriptome sequencing. Gene ontology (GO) and miR family analysis were used to uncover dysregulated signalling which were then validated in vitro in human foetal neural progenitor cells treated with IVH-CSF., Results: Five hundred eighty-seven miRs were differentially expressed in the CSF extracted at least 2 months after injury, compared to CSF extracted within the first month of injury. GO uncovered key pathways targeted by differentially expressed miRs including the MAPK cascade and the JAK/STAT pathway. Astrogliosis is known to occur in preterm infants, and we hypothesized that this could be due to abnormal CSF-miR signaling resulting in dysregulation of the JAK/STAT pathway - a key controller of astrocyte differentiation. We then confirmed that treatment with IVH-CSF promotes astrocyte differentiation from human fetal NPCs and that this effect could be prevented by JAK/STAT inhibition. Taken together, our results provide novel insights into the CSF/NPCs crosstalk following perinatal brain injury and reveal novel targets to improve neurodevelopmental outcomes in preterm infants., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Gialeli, Spaull, Plösch, Uney, Cordero Llana and Heep.)
- Published
- 2023
- Full Text
- View/download PDF
14. Growth-Restricted Fetuses and Offspring Reveal Adverse Sex-Specific Metabolic Responses in Preeclamptic Mice Expressing Human sFLT1.
- Author
-
Vogtmann R, Bao M, Dewan MV, Riedel A, Kimmig R, Felderhoff-Müser U, Bendix I, Plösch T, and Gellhaus A
- Subjects
- Humans, Pregnancy, Mice, Female, Male, Animals, Placenta metabolism, Vascular Endothelial Growth Factor Receptor-1 genetics, Fetus metabolism, Mice, Transgenic, Weight Gain, Fetal Growth Retardation genetics, Pre-Eclampsia metabolism
- Abstract
Fetal adaptations to harmful intrauterine environments due to pregnancy disorders such as preeclampsia (PE) can negatively program the offspring's metabolism, resulting in long-term metabolic changes. PE is characterized by increased circulating levels of sFLT1, placental dysfunction and fetal growth restriction (FGR). Here we examine the consequences of systemic human sFLT1 overexpression in transgenic PE/FGR mice on the offspring's metabolic phenotype. Histological and molecular analyses of fetal and offspring livers as well as examinations of offspring serum hormones were performed. At 18.5 dpc, sFLT1 overexpression resulted in growth-restricted fetuses with a reduced liver weight, combined with reduced hepatic glycogen storage and histological signs of hemorrhages and hepatocyte apoptosis. This was further associated with altered gene expression of the molecules involved in fatty acid and glucose/glycogen metabolism. In most analyzed features males were more affected than females. The postnatal follow-up revealed an increased weight gain of male PE offspring, and increased serum levels of Insulin and Leptin. This was associated with changes in hepatic gene expression regulating fatty acid and glucose metabolism in male PE offspring. To conclude, our results indicate that sFLT1-related PE/FGR in mice leads to altered fetal liver development, which might result in an adverse metabolic pre-programming of the offspring, specifically targeting males. This could be linked to the known sex differences seen in PE pregnancies in human.
- Published
- 2023
- Full Text
- View/download PDF
15. Hypermethylation of CTDSPL2 prior to necrotizing enterocolitis onset.
- Author
-
Klerk DH, Kooi EM, Bos AF, Hulscher JB, Verkaik-Schakel RN, and Plösch T
- Subjects
- Humans, Infant, Infant, Newborn, DNA Methylation, Feces, Enterocolitis, Necrotizing genetics, Infant, Premature
- Abstract
Background: Necrotizing enterocolitis (NEC) is the most common gastrointestinal emergency in preterm infants. Epigenetic changes in DNA methylation may be present prior to NEC onset. Methods: 24 preterm infants with NEC and 45 matched controls were included. Human DNA was isolated from stool samples and methylation of CTDSPL2 , HERC1 , NXPE3 and PTGDR was measured using pyrosequencing. Results: CTDSPL2 displayed a higher DNA methylation of 51% compared with 17% in controls, prior to NEC onset (p = 0.047). Discussion: Noninvasive measurement of methylation in stool allows for comparison with healthy preterm controls. This potentially allows future biomarker or risk predictor use. The effect of CTDSPL2 hypermethylation on gene expression remains unclear.
- Published
- 2023
- Full Text
- View/download PDF
16. Preeclampsia-induced alterations in brain and liver gene expression and DNA methylation patterns in fetal mice.
- Author
-
Hofsink N, Dijkstra DJ, Stojanovska V, Scherjon SA, and Plösch T
- Subjects
- Animals, Female, Humans, Male, Mice, Pregnancy, Brain metabolism, Gene Expression, Liver metabolism, Vascular Endothelial Growth Factor Receptor-1, DNA Methylation, Pre-Eclampsia genetics
- Abstract
Exposure to pregnancy complications, including preeclampsia (PE), has lifelong influences on offspring's health. We have previously reported that experimental PE, induced in mice by administration of adenoviral sFlt1 at gestational day 8.5 combined with LPS at day 10.5, results in symmetrical growth restriction in female and asymmetrical growth restriction in male offspring. Here, we characterize the molecular phenotype of the fetal brain and liver with respect to gene transcription and DNA methylation at the end of gestation.In fetal brain and liver, expression and DNA methylation of several key regulatory genes is altered by PE exposure, mostly independent of fetal sex. These alterations point toward a decreased gluconeogenesis in the liver and stimulated neurogenesis in the brain, potentially affecting long-term brain and liver function. The observed sex-specific growth restriction pattern is not reflected in the molecular data, showing that PE, rather than tissue growth, drives the molecular phenotype of PE-exposed offspring.
- Published
- 2023
- Full Text
- View/download PDF
17. Methylation of the serotonin reuptake transporter gene and non-motor symptoms in dystonia patients.
- Author
-
Timmers ER, Plösch T, Smit M, Hof IH, Verkaik-Schakel RN, Tijssen MAJ, de Koning TJ, and Niezen-Koning KE
- Subjects
- Humans, DNA Methylation, Serotonin, Serotonin Plasma Membrane Transport Proteins genetics, Dystonia genetics, Dystonia complications, Dystonic Disorders complications, Dystonic Disorders genetics
- Abstract
Background: Dystonia is a rare movement disorder, in which patients suffer from involuntary twisting movements or abnormal posturing. Next to these motor symptoms, patients have a high prevalence of psychiatric comorbidity, suggesting a role for serotonin in its pathophysiology. This study investigates the percentage of DNA methylation of the gene encoding for the serotonin reuptake transporter (SLC6A4) in dystonia patients and the associations between methylation levels and presence and severity of psychiatric symptoms., Methods: Patients with cervical dystonia (n = 49), myoclonus dystonia (n = 41) and dopa-responsive dystonia (DRD) (n = 27) and a group of healthy controls (n = 56) were included. Psychiatric comorbidity was evaluated with validated questionnaires. Methylation levels of 20 CpG sites situated 69 to 213 base pairs upstream of the start codon of SLC6A4 were investigated. Methylation in dystonia patients was compared to healthy controls, correcting for age, and correlated with psychiatric comorbidity., Results: Bootstrapped quantile regression analysis showed that being a dystonia patient compared to a healthy control significantly explains the methylation level at two CpG sites (CpG 24: pseudo-R
2 = 0.05, p = 0.04, CpG 32: pseudo-R2 = 0.14, p = 0.03). Subgroup analysis revealed that being a DRD patient significantly explained a part of the variance of methylation levels at two CpG sites (CpG 21: pseudo-R2 = 0.03, p = 0.00, CpG 24: pseudo-R2 = 0.06, p = 0.03). Regression analysis showed that methylation level at CpG 38 significantly explained a small proportion of the variance of severity score for anxiety (R2 = 0.07, p = 0.04) and having a diagnosis of depression (Nagelkerke R2 : 0.11, p = 0.00). Genotype of the 5-HTTLPR polymorphism had no additional effect on these associations., Conclusions: This study showed an association between percentage of methylation at several specific sites of the promoter region of SLCA64 and (dopa-responsive) dystonia patients compared to healthy controls. Furthermore, methylation levels were associated with severity of anxiety and presence of a depressive disorder in the dystonia group. This study suggests alterations in the serotonergic metabolism in dystonia patients, and its relation with the non-motor symptoms., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
18. Epigenetics in the primary and secondary prevention of cardiovascular disease: influence of exercise and nutrition.
- Author
-
Gevaert AB, Wood N, Boen JRA, Davos CH, Hansen D, Hanssen H, Krenning G, Moholdt T, Osto E, Paneni F, Pedretti RFE, Plösch T, Simonenko M, and Bowen TS
- Subjects
- Humans, Secondary Prevention, Exercise, Cardiovascular Diseases genetics, Cardiovascular Diseases prevention & control
- Abstract
Increasing evidence links changes in epigenetic systems, such as DNA methylation, histone modification, and non-coding RNA expression, to the occurrence of cardiovascular disease (CVD). These epigenetic modifications can change genetic function under influence of exogenous stimuli and can be transferred to next generations, providing a potential mechanism for inheritance of behavioural intervention effects. The benefits of exercise and nutritional interventions in the primary and secondary prevention of CVD are well established, but the mechanisms are not completely understood. In this review, we describe the acute and chronic epigenetic effects of physical activity and dietary changes. We propose exercise and nutrition as potential triggers of epigenetic signals, promoting the reshaping of transcriptional programmes with effects on CVD phenotypes. Finally, we highlight recent developments in epigenetic therapeutics with implications for primary and secondary CVD prevention., Competing Interests: Conflict of interest: A.B.G. reported receiving speaker fees from Abbott, AstraZeneca, and Boehringer Ingelheim (lectures) outside of the submitted work. No potential competing interest was reported by the other authors., (© The Author(s) 2022. Published by Oxford University Press on behalf of the European Society of Cardiology.)
- Published
- 2022
- Full Text
- View/download PDF
19. 14-weeks combined exercise epigenetically modulated 118 genes of menopausal women with prediabetes.
- Author
-
Yumi Noronha N, da Silva Rodrigues G, Harumi Yonehara Noma I, Fernanda Cunha Brandao C, Pereira Rodrigues K, Colello Bruno A, Sae-Lee C, Moriguchi Watanabe L, Augusta de Souza Pinhel M, Mello Schineider I, Luciano de Almeida M, Barbosa Júnior F, Araújo Morais D, Tavares de Sousa Júnior W, Plösch T, Roberto Bueno Junior C, and Barbosa Nonino C
- Subjects
- Exercise, Female, Humans, Menopause genetics, Diabetes Mellitus, Hyperglycemia, Prediabetic State genetics, Prediabetic State therapy
- Abstract
Background: Pre-diabetes precedes Diabetes Mellitus (DM) disease and is a critical period for hyperglycemia treatment, especially for menopausal women, considering all metabolic alterations due to hormonal changes. Recently, the literature has demonstrated the role of physical exercise in epigenetic reprogramming to modulate the gene expression patterns of metabolic conditions, such as hyperglycemia, and prevent DM development. In the present study, we hypothesized that physical exercise training could modify the epigenetic patterns of women with poor glycemic control., Methods: 48 post-menopause women aged 60.3 ± 4.5 years were divided according to their fasting blood glucose levels into two groups: Prediabetes Group, PG (n=24), and Normal Glucose Group, NGG (n=24). All participants performed 14 weeks of physical exercise three times a week. The Infinium Methylation EPIC BeadChip measured the participants' Different Methylated Regions (DMRs)., Results: Before the intervention, the PG group had 12 DMRs compared to NGG. After the intervention, five DMRs remained different. Interestingly, when comparing the PG group before and after training, 118 DMRs were found. The enrichment analysis revealed that the genes were related to different biological functions such as energy metabolism, cell differentiation, and tumor suppression., Conclusion: Physical exercise is a relevant alternative in treating hyperglycemia and preventing DM in post-menopause women with poor glycemic control., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Yumi Noronha, da Silva Rodrigues, Harumi Yonehara Noma, Fernanda Cunha Brandao, Pereira Rodrigues, Colello Bruno, Sae-Lee, Moriguchi Watanabe, Augusta de Souza Pinhel, Mello Schineider, Luciano de Almeida, Barbosa Júnior, Araújo Morais, Tavares de Sousa Júnior, Plösch, Roberto Bueno Junior and Barbosa Nonino.)
- Published
- 2022
- Full Text
- View/download PDF
20. Perinatal exposure to fluoxetine and maternal adversity affect myelin-related gene expression and epigenetic regulation in the corticolimbic circuit of juvenile rats.
- Author
-
Ramsteijn AS, Verkaik-Schakel RN, Houwing DJ, Plösch T, and Olivier JDA
- Subjects
- Animals, Epigenesis, Genetic, Female, Gene Expression, Hippocampus, Humans, Male, Myelin Sheath metabolism, Pregnancy, Rats, Rats, Sprague-Dawley, Selective Serotonin Reuptake Inhibitors, Fluoxetine pharmacology, Prenatal Exposure Delayed Effects metabolism
- Abstract
Many pregnant women experience symptoms of depression, and are often treated with selective serotonin reuptake inhibitor (SSRI) antidepressants, such as fluoxetine. In utero exposure to SSRIs and maternal depressive symptoms is associated with sex-specific effects on the brain and behavior. However, knowledge about the neurobiological mechanisms underlying these sex differences is limited. In addition, most animal research into developmental SSRI exposure neglects the influence of maternal adversity. Therefore, we used a rat model relevant to depression to investigate the molecular effects of perinatal fluoxetine exposure in male and female juvenile offspring. We performed RNA sequencing and targeted DNA methylation analyses on the prefrontal cortex and basolateral amygdala; key regions of the corticolimbic circuit. Perinatal fluoxetine enhanced myelin-related gene expression in the prefrontal cortex, while inhibiting it in the basolateral amygdala. SSRI exposure and maternal adversity interacted to affect expression of genes such as myelin-associated glycoprotein (Mag) and myelin basic protein (Mbp). We speculate that altered myelination reflects altered brain maturation. In addition, these effects are stronger in males than in females, resembling known behavioral outcomes. Finally, Mag and Mbp expression correlated with DNA methylation, highlighting epigenetic regulation as a potential mechanism for developmental fluoxetine-induced changes in myelination., (© 2022. The Author(s), under exclusive licence to American College of Neuropsychopharmacology.)
- Published
- 2022
- Full Text
- View/download PDF
21. Differential Placental DNA Methylation of NR3C1 in Extremely Preterm Infants With Poorer Neurological Functioning.
- Author
-
van Dokkum NH, Bachini S, Verkaik-Schakel RN, Baptist DH, Salavati S, Kraft KE, Scherjon SA, Bos AF, and Plösch T
- Abstract
Background: Understanding underlying mechanisms of neurodevelopmental impairment following preterm birth may enhance opportunities for targeted interventions. We aimed to assess whether placental DNA methylation of selected genes affected early neurological functioning in preterm infants., Methods: We included 43 infants, with gestational age <30 weeks and/or birth weight <1,000 g and placental samples at birth. We selected genes based on their associations with several prenatal conditions that may be related to poor neurodevelopmental outcomes. We determined DNA methylation using pyrosequencing, and neurological functioning at 3 months post-term using Prechtl's General Movement Assessment, including the Motor Optimality Score-Revised (MOS-R)., Results: Twenty-four infants had atypical MOS-R, 19 infants had near-optimal MOS-R. We identified differences in average methylation of NR3C1 (encoding for the glucocorticoid receptor) [3.3% (95%-CI: 2.4%-3.9%) for near-optimal vs. 2.3% (95%-CI: 1.7%-3.0%), p = 0.008 for atypical], and at three of the five individual CpG-sites. For EPO, SLC6A3, TLR4, VEGFA, LEP and HSD11B2 we found no differences between the groups., Conclusion: Hypomethylation of NR3C1 in placental tissue is associated with poorer neurological functioning at 3 months post-term in extremely preterm infants. Alleviating stress during pregnancy and its impact on preterm infants and their neurodevelopmental outcomes should be further investigated., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 van Dokkum, Bachini, Verkaik-Schakel, Baptist, Salavati, Kraft, Scherjon, Bos and Plösch.)
- Published
- 2022
- Full Text
- View/download PDF
22. Altered neurodevelopmental DNA methylation status after fetal growth restriction with brain-sparing.
- Author
-
Richter AE, Bekkering-Bauer I, Verkaik-Schakel RN, Leeuwerke M, Tanis JC, Bilardo CM, Kooi EMW, Scherjon SA, Bos AF, and Plösch T
- Subjects
- Brain diagnostic imaging, Child Behavior, Child, Preschool, DNA Methylation, Female, Follow-Up Studies, Humans, Hypoxia, Pregnancy, Vascular Endothelial Growth Factor A, Brain-Derived Neurotrophic Factor genetics, Fetal Growth Retardation genetics
- Abstract
It is under debate how preferential perfusion of the brain (brain-sparing) in fetal growth restriction (FGR) relates to long-term neurodevelopmental outcome. Epigenetic modification of neurotrophic genes by altered fetal oxygenation may be involved. To explore this theory, we performed a follow-up study of 21 FGR children, in whom we prospectively measured the prenatal cerebroplacental ratio (CPR) with Doppler sonography. At 4 years of age, we tested their neurodevelopmental outcome using the Wechsler Preschool and Primary Scale of Intelligence, the Child Behavior Checklist, and the Behavior Rating Inventory of Executive Function. In addition, we collected their buccal DNA to determine the methylation status at predefined genetic regions within the genes hypoxia-inducible factor-1 alpha (HIF1A), vascular endothelial growth factor A (VEGFA), erythropoietin (EPO), EPO-receptor (EPOR), brain-derived neurotrophic factor (BDNF), and neurotrophic tyrosine kinase, receptor, type 2 (NTRK2) by pyrosequencing. We found that FGR children with fetal brain-sparing (CPR <1, n = 8) demonstrated a trend (0.05 < p < 0.1) toward hypermethylation of HIF1A and VEGFA at their hypoxia-response element (HRE) compared with FGR children without fetal brain-sparing. Moreover, in cases with fetal brain-sparing, we observed statistically significant hypermethylation at a binding site for cyclic adenosine monophophate response element binding protein (CREB) of BDNF promoter exon 4 and hypomethylation at an HRE located within the NTRK2 promoter (both p <0.05). Hypermethylation of VEGFA was associated with a poorer Performance Intelligence Quotient, while hypermethylation of BDNF was associated with better inhibitory self-control (both p <0.05). These results led us to formulate the hypothesis that early oxygen-dependent epigenetic alterations due to hemodynamic alterations in FGR may be associated with altered neurodevelopmental outcome in later life. We recommend further studies to test this hypothesis.
- Published
- 2022
- Full Text
- View/download PDF
23. Corrigendum: Novel Zinc-Related Differentially Methylated Regions in Leukocytes of Women With and Without Obesity.
- Author
-
Noronha NY, Barato M, Sae-Lee C, Pinhel MAS, Watanabe LM, Pereira VAB, Rodrigues GDS, Morais DA, de Sousa WT Jr, Souza VCO, Plaça JR, Salgado W Jr, Barbosa F Jr, Plösch T, and Nonino CB
- Abstract
[This corrects the article DOI: 10.3389/fnut.2022.785281.]., (Copyright © 2022 Noronha, Barato, Sae-Lee, Pinhel, Watanabe, Pereira, Rodrigues, Morais, de Sousa, Souza, Plaça, Salgado, Barbosa, Plösch and Nonino.)
- Published
- 2022
- Full Text
- View/download PDF
24. Plasticity-Related Gene 5 Is Expressed in a Late Phase of Neurodifferentiation After Neuronal Cell-Fate Determination.
- Author
-
Gross I, Brandt N, Vonk D, Köper F, Wöhlbrand L, Rabus R, Witt M, Heep A, Plösch T, Hipp MS, and Bräuer AU
- Abstract
During adult neurogenesis, neuronal stem cells differentiate into mature neurons that are functionally integrated into the existing network. One hallmark during the late phase of this neurodifferentiation process is the formation of dendritic spines. These morphological specialized structures form the basis of most excitatory synapses in the brain, and are essential for neuronal communication. Additionally, dendritic spines are affected in neurological disorders, such as Alzheimer's disease or schizophrenia. However, the mechanisms underlying spinogenesis, as well as spine pathologies, are poorly understood. Plasticity-related Gene 5 (PRG5), a neuronal transmembrane protein, has previously been linked to spinogenesis in vitro . Here, we analyze endogenous expression of the PRG5 protein in different mouse brain areas, as well as on a subcellular level. We found that native PRG5 is expressed dendritically, and in high abundance in areas characterized by their regenerative capacity, such as the hippocampus and the olfactory bulb. During adult neurogenesis, PRG5 is specifically expressed in a late phase after neuronal cell-fate determination associated with dendritic spine formation. On a subcellular level, we found PRG5 not to be localized at the postsynaptic density, but at the base of the synapse. In addition, we showed that PRG5-induced formation of membrane protrusions is independent from neuronal activity, supporting a possible role in the morphology and stabilization of spines., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Gross, Brandt, Vonk, Köper, Wöhlbrand, Rabus, Witt, Heep, Plösch, Hipp and Bräuer.)
- Published
- 2022
- Full Text
- View/download PDF
25. Novel Zinc-Related Differentially Methylated Regions in Leukocytes of Women With and Without Obesity.
- Author
-
Noronha NY, Barato M, Sae-Lee C, Pinhel MAS, Watanabe LM, Pereira VAB, Rodrigues GDS, Morais DA, de Sousa WT Jr, Souza VCO, Plaça JR, Salgado W Jr, Barbosa F Jr, Plösch T, and Nonino CB
- Abstract
Introduction: Nutriepigenetic markers are predictive responses associated with changes in "surrounding" environmental conditions of humans, which may influence metabolic diseases. Although rich in calories, Western diets could be linked with the deficiency of micronutrients, resulting in the downstream of epigenetic and metabolic effects and consequently in obesity. Zinc (Zn) is an essential nutrient associated with distinct biological roles in human health. Despite the importance of Zn in metabolic processes, little is known about the relationship between Zn and epigenetic. Thus, the present study aimed to identify the epigenetic variables associated with Zn daily ingestion (ZnDI) and serum Zinc (ZnS) levels in women with and without obesity., Materials and Methods: This is a case-control, non-randomized, single-center study conducted with 21 women allocated into two groups: control group (CG), composed of 11 women without obesity, and study group (SG), composed of 10 women with obesity. Anthropometric measurements, ZnDI, and ZnS levels were evaluated. Also, leukocyte DNA was extracted for DNA methylation analysis using 450 k Illumina BeadChips. The epigenetic clock was calculated by Horvath method. The chip analysis methylation pipeline (ChAMP) package selected the differentially methylated regions (DMRs)., Results: The SG had lower ZnS levels than the CG. Moreover, in SG, the ZnS levels were negatively associated with the epigenetic age acceleration. The DMR analysis revealed 37 DMRs associated with ZnDI and ZnS levels. The DMR of PM20D1 gene was commonly associated with ZnDI and ZnS levels and was hypomethylated in the SG., Conclusion: Our findings provide new information on Zn's modulation of DNA methylation patterns and bring new perspectives for understanding the nutriepigenetic mechanisms in obesity., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Noronha, Barato, Sae-Lee, Pinhel, Watanabe, Pereira, Rodrigues, Morais, de Sousa, Souza, Plaça, Salgado, Barbosa, Plösch and Nonino.)
- Published
- 2022
- Full Text
- View/download PDF
26. LPS versus Poly I:C model: comparison of long-term effects of bacterial and viral maternal immune activation on the offspring.
- Author
-
Bao M, Hofsink N, and Plösch T
- Subjects
- Bacterial Infections immunology, Female, Humans, Pregnancy, COVID-19 complications, COVID-19 immunology, Lipopolysaccharides toxicity, Poly I-C toxicity, Prenatal Exposure Delayed Effects immunology, SARS-CoV-2
- Abstract
A prominent health issue nowadays is the COVID-19 pandemic, which poses acute risks to human health. However, the long-term health consequences are largely unknown and cannot be neglected. An especially vulnerable period for infection is pregnancy, when infections could have long-term health effect on the child. Evidence suggests that maternal immune activation (MIA) induced by either bacteria or viruses presents various effects on the offspring, leading to adverse phenotypes in many organ systems. This review compares the mechanisms of bacterial and viral MIA and the possible long-term outcomes for the offspring by summarizing the outcome in animal LPS and Poly I:C models. Both models are activated immune responses mediated by Toll-like receptors. The outcomes for MIA offspring include neurodevelopment, immune response, circulation, metabolism, and reproduction. Some of these changes continue to exist until later life. Besides different doses and batches of LPS and Poly I:C, the injection day, administration route, and also different animal species influence the outcomes. Here, we specifically aim to support colleagues when choosing their animal models for future studies.
- Published
- 2022
- Full Text
- View/download PDF
27. Disruption of paternal circadian rhythm affects metabolic health in male offspring via nongerm cell factors.
- Author
-
Lassi M, Tomar A, Comas-Armangué G, Vogtmann R, Dijkstra DJ, Corujo D, Gerlini R, Darr J, Scheid F, Rozman J, Aguilar-Pimentel A, Koren O, Buschbeck M, Fuchs H, Marschall S, Gailus-Durner V, Hrabe de Angelis M, Plösch T, Gellhaus A, and Teperino R
- Subjects
- Animals, Male, Mice, Phenotype, Circadian Rhythm genetics
- Abstract
Circadian rhythm synchronizes each body function with the environment and regulates physiology. Disruption of normal circadian rhythm alters organismal physiology and increases disease risk. Recent epidemiological data and studies in model organisms have shown that maternal circadian disruption is important for offspring health and adult phenotypes. Less is known about the role of paternal circadian rhythm for offspring health. Here, we disrupted circadian rhythm in male mice by night-restricted feeding and showed that paternal circadian disruption at conception is important for offspring feeding behavior, metabolic health, and oscillatory transcription. Mechanistically, our data suggest that the effect of paternal circadian disruption is not transferred to the offspring via the germ cells but initiated by corticosterone-based parental communication at conception and programmed during in utero development through a state of fetal growth restriction. These findings indicate paternal circadian health at conception as a newly identified determinant of offspring phenotypes., (Copyright © 2021 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works. Distributed under a Creative Commons Attribution License 4.0 (CC BY).)
- Published
- 2021
- Full Text
- View/download PDF
28. Antioxidative Molecules in Human Milk and Environmental Contaminants.
- Author
-
Lorenzetti S, Plösch T, and Teller IC
- Abstract
Breastfeeding provides overall beneficial health to the mother-child dyad and is universally recognized as the preferred feeding mode for infants up to 6-months and beyond. Human milk provides immuno-protection and supplies nutrients and bioactive compounds whose concentrations vary with lactation stage. Environmental and dietary factors potentially lead to excessive chemical exposure in critical windows of development such as neonatal life, including lactation. This review discusses current knowledge on these environmental and dietary contaminants and summarizes the known effects of these chemicals in human milk, taking into account the protective presence of antioxidative molecules. Particular attention is given to short- and long-term effects of these contaminants, considering their role as endocrine disruptors and potential epigenetic modulators. Finally, we identify knowledge gaps and indicate potential future research directions.
- Published
- 2021
- Full Text
- View/download PDF
29. DNA Methylation of TLR4, VEGFA , and DEFA5 Is Associated With Necrotizing Enterocolitis in Preterm Infants.
- Author
-
Klerk DH, Plösch T, Verkaik-Schakel RN, Hulscher JBF, Kooi EMW, and Bos AF
- Abstract
Background: Epigenetic changes, such as DNA methylation, may contribute to an increased susceptibility for developing necrotizing enterocolitis (NEC) in preterm infants. We assessed DNA methylation in five NEC-associated genes, selected from literature: EPO, VEGFA, ENOS, DEFA5 , and TLR4 in infants with NEC and controls. Methods: Observational cohort study including 24 preterm infants who developed NEC (≥Bell Stage IIA) and 45 matched controls. DNA was isolated from stool samples and methylation measured using pyrosequencing. We investigated differences in methylation prior to NEC compared with controls. Next, in NEC infants, we investigated methylation patterns long before, a short time before NEC onset, and after NEC. Results: Prior to NEC, only TLR4 CpG 2 methylation was increased in NEC infants (median = 75.4%, IQR = 71.3-83.8%) versus controls (median = 69.0%, IQR = 64.5-77.4%, p = 0.025). In NEC infants, VEGFA CpG 3 methylation was 0.8% long before NEC, increasing to 1.8% a short time before NEC and 2.0% after NEC ( p = 0.011; p = 0.021, respectively). A similar pattern was found in DEFA5 CpG 1, which increased from 75.4 to 81.4% and remained 85.3% ( p = 0.027; p = 0.019, respectively). These changes were not present for EPO, ENOS , and TLR4 . Conclusion: Epigenetic changes of TLR4, VEGFA , and DEFA5 are present in NEC infants and can differ in relation to the time of NEC onset. Differences in DNA methylation of TLR4, VEGFA , and DEFA5 may influence gene expression and increase the risk for developing NEC. This study also demonstrates the use of human DNA extraction from stool samples as a novel non-invasive method for exploring the bowel of preterm infants and which can also be used for necrotizing enterocolitis patients., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Klerk, Plösch, Verkaik-Schakel, Hulscher, Kooi and Bos.)
- Published
- 2021
- Full Text
- View/download PDF
30. Antidepressant treatment is associated with epigenetic alterations of Homer1 promoter in a mouse model of chronic depression.
- Author
-
Sun L, Verkaik-Schakel RN, Biber K, Plösch T, and Serchov T
- Subjects
- Animals, Disease Models, Animal, Epigenesis, Genetic, Homer Scaffolding Proteins metabolism, Imipramine, Mice, Promoter Regions, Genetic genetics, Antidepressive Agents pharmacology, Depression drug therapy, Depression genetics
- Abstract
Background: Understanding the neurobiology of depression and the mechanism of action of therapeutic measures is currently a research priority. We have shown that the expression of the synaptic protein Homer1a correlates with depression-like behavior and its induction is a common mechanism of action of different antidepressant treatments. However, the mechanism of Homer1a regulation is still unknown., Methods: We combined the chronic despair mouse model (CDM) of chronic depression with different antidepressant treatments. Depression-like behavior was characterized by forced swim and tail suspension tests, and via automatic measurement of sucrose preference in IntelliCage. The Homer1 mRNA expression and promoter DNA methylation were analyzed in cortex and peripheral blood by qRT-PCR and pyrosequencing., Results: CDM mice show decreased Homer1a and Homer1b/c mRNA expression in cortex and blood samples, while chronic treatment with imipramine and fluoxetine or acute ketamine application increases their level only in the cortex. The quantitative analyses of the methylation of 7 CpG sites, located on the Homer1 promoter region containing several CRE binding sites, show a significant increase in DNA methylation in the cortex of CDM mice. In contrast, antidepressant treatments reduce the methylation level., Limitations: Homer1 expression and promotor methylation were not analyzed in different blood cell types. Other CpG sites of Homer1 promoter should be investigated in future studies. Our experimental approach does not distinguish between methylation and hydroxymethylation., Conclusions: We demonstrate that stress-induced depression-like behavior and antidepressant treatments are associated with epigenetic alterations of Homer1 promoter, providing new insights into the mechanism of antidepressant treatment., (Copyright © 2020. Published by Elsevier B.V.)
- Published
- 2021
- Full Text
- View/download PDF
31. Sex and Exposure to Postnatal Chlorpyrifos Influence the Epigenetics of Feeding-Related Genes in a Transgenic APOE Mouse Model: Long-Term Implications on Body Weight after a High-Fat Diet.
- Author
-
Guardia-Escote L, Blanco J, Basaure P, Biosca-Brull J, Verkaik-Schakel RN, Cabré M, Peris-Sampedro F, Pérez-Fernández C, Sánchez-Santed F, Plösch T, Domingo JL, and Colomina MT
- Subjects
- Animals, Diet, High-Fat adverse effects, Female, Genotype, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, ApoE, Body Weight, Chlorpyrifos toxicity, Epigenesis, Genetic, Insecticides toxicity, Sex Factors
- Abstract
Developmental exposure to toxicants and diet can interact with an individual's genetics and produce long-lasting metabolic adaptations. The different isoforms of the apolipoprotein E ( APOE ) are an important source of variability in metabolic disorders and influence the response to the pesticide chlorpyrifos (CPF). We aimed to study the epigenetic regulation on feeding control genes and the influence of postnatal CPF exposure, APOE genotype, and sex, and how these modifications impact on the metabolic response to a high-fat diet (HFD). Both male and female apoE3- and apoE4-TR mice were exposed to CPF on postnatal days 10-15. The DNA methylation pattern of proopiomelanocortin, neuropeptide Y, leptin receptor, and insulin-like growth factor 2 was studied in the hypothalamus. At adulthood, the mice were given a HFD for eight weeks. The results highlight the importance of sex in the epigenetic regulation and the implication of CPF treatment and APOE genotype. The body weight progression exhibited sex-dimorphic differences, apoE4-TR males being the most susceptible to the effects induced by CPF and HFD. Overall, these results underscore the pivotal role of sex, APOE genotype, and developmental exposure to CPF on subsequent metabolic disturbances later in life and show that sex is a key variable in epigenetic regulation.
- Published
- 2020
- Full Text
- View/download PDF
32. Postnatal Smoke Exposure Further Increases the Hepatic Nicotine Metabolism in Prenatally Smoke Exposed Male Offspring and Is Linked with Aberrant Cyp2a5 Methylation.
- Author
-
Lkhagvadorj K, Zeng Z, Meyer KF, Verweij LP, Kooistra W, Reinders-Luinge M, Dijkhuizen HW, de Graaf IAM, Plösch T, and Hylkema MN
- Subjects
- Animals, Animals, Newborn, Aryl Hydrocarbon Hydroxylases chemistry, Aryl Hydrocarbon Hydroxylases genetics, Cytochrome P450 Family 2 chemistry, Cytochrome P450 Family 2 genetics, Female, Male, Mice, Mice, Inbred C57BL, Pregnancy, Prenatal Exposure Delayed Effects etiology, Prenatal Exposure Delayed Effects metabolism, Promoter Regions, Genetic, Aryl Hydrocarbon Hydroxylases metabolism, Cytochrome P450 Family 2 metabolism, DNA Methylation, Gene Expression Regulation drug effects, Inactivation, Metabolic, Nicotine metabolism, Prenatal Exposure Delayed Effects pathology, Smoke adverse effects
- Abstract
Prenatal smoke exposure (PreSE) is a risk factor for nicotine dependence, which is further enhanced by postnatal smoke exposure (PostSE). One susceptibility gene to nicotine dependence is Cytochrome P450 (CYP) 2A6, an enzyme responsible for the conversion of nicotine to cotinine in the liver. Higher CYP2A6 activity is associated with nicotine dependence and could be regulated through DNA methylation. In this study we investigated whether PostSE further impaired PreSE-induced effects on nicotine metabolism, along with Cyp2a5 , orthologue of CYP2A6 , mRNA expression and DNA methylation. Using a mouse model where prenatally smoke-exposed adult offspring were exposed to cigarette smoke for 3 months, enzyme activity, mRNA levels, and promoter methylation of hepatic Cyp2a5 were evaluated. We found that in male offspring, PostSE increased PreSE-induced cotinine levels and Cyp2a5 mRNA expression. In addition, both PostSE and PreSE changed Cyp2a5 DNA methylation in male groups. PreSE however decreased cotinine levels whereas it had no effect on Cyp2a5 mRNA expression or methylation. These adverse outcomes of PreSE and PostSE were most prominent in males. When considered in the context of the human health aspects, the combined effect of prenatal and adolescent smoke exposure could lead to an accelerated risk for nicotine dependence later in life.
- Published
- 2020
- Full Text
- View/download PDF
33. Prenatal smoke exposure induces persistent Cyp2a5 methylation and increases nicotine metabolism in the liver of neonatal and adult male offspring.
- Author
-
Lkhagvadorj K, Meyer KF, Verweij LP, Kooistra W, Reinders-Luinge M, Dijkhuizen HW, de Graaf IAM, Plösch T, and Hylkema MN
- Subjects
- Animals, CpG Islands, Female, Liver growth & development, Male, Mice, Mice, Inbred C57BL, Pregnancy, Prenatal Exposure Delayed Effects metabolism, Promoter Regions, Genetic, RNA, Messenger genetics, RNA, Messenger metabolism, Aryl Hydrocarbon Hydroxylases genetics, Cytochrome P450 Family 2 genetics, DNA Methylation, Liver metabolism, Nicotine metabolism, Prenatal Exposure Delayed Effects genetics, Tobacco Smoke Pollution adverse effects
- Abstract
Prenatal smoke exposure (PSE) is a risk factor for nicotine dependence. One susceptibility gene for nicotine dependence is Cytochrome P450 (CYP) 2A6, an enzyme responsible for the conversion of nicotine to cotinine and nicotine clearance in the liver. Higher activity of the CYP2A6 enzyme is associated with nicotine dependence, but no research has addressed the PSE effects on the CYP2A6 gene or its mouse homologue Cyp2a5 . We hypothesized that PSE affects Cyp2a5 promoter methylation, Cyp2a5 mRNA levels, and nicotine metabolism in offspring. We used a smoke-exposed pregnant mouse model. RNA, DNA, and microsomal protein were isolated from liver tissue of foetal, neonatal, and adult offspring. Enzyme activity, Cyp2a5 mRNA levels, and Cyp2a5 methylation status of six CpG sites within the promoter region were analysed via HPLC, RT-PCR, and bisulphite pyrosequencing. Our data show that PSE induced higher cotinine levels in livers of male neonatal and adult offspring compared to controls. PSE-induced cotinine levels in neonates correlated with Cyp2a5 mRNA expression and promoter methylation at CpG-7 and CpG+45. PSE increased methylation in almost all CpG sites in foetal offspring, and this effect persisted at CpG-74 in male neonatal and adult offspring. Our results indicate that male offspring of mothers which were exposed to cigarette smoke during pregnancy have a higher hepatic nicotine metabolism, which could be regulated by DNA methylation. Given the detected persistence into adulthood, extrapolation to the human situation suggests that sons born from smoking mothers could be more susceptible to nicotine dependence later in life.
- Published
- 2020
- Full Text
- View/download PDF
34. Developmental programming in human umbilical cord vein endothelial cells following fetal growth restriction.
- Author
-
Terstappen F, Calis JJA, Paauw ND, Joles JA, van Rijn BB, Mokry M, Plösch T, and Lely AT
- Subjects
- Adult, Cardiovascular Diseases embryology, Case-Control Studies, CpG Islands, DNA Methylation, Epigenomics, Female, Fetal Growth Retardation diagnosis, Fetal Growth Retardation etiology, Fetus chemistry, Fetus metabolism, Fetus physiopathology, Galectin 1, Gene Expression Regulation genetics, Humans, Kidney Diseases embryology, Male, Membrane Proteins, Nuclear Proteins, Placental Insufficiency metabolism, Placental Insufficiency physiopathology, Pregnancy, Pregnancy Complications metabolism, Prospective Studies, RNA, Long Noncoding genetics, RNA-Seq methods, Receptors, Formyl Peptide, Cardiovascular Diseases genetics, Fetal Growth Retardation genetics, Fetus blood supply, Gene Expression Profiling methods, Human Umbilical Vein Endothelial Cells metabolism, Kidney Diseases genetics
- Abstract
Background: Fetal growth restriction (FGR) is associated with an increased susceptibility for various noncommunicable diseases in adulthood, including cardiovascular and renal disease. During FGR, reduced uteroplacental blood flow, oxygen and nutrient supply to the fetus are hypothesized to detrimentally influence cardiovascular and renal programming. This study examined whether developmental programming profiles, especially related to the cardiovascular and renal system, differ in human umbilical vein endothelial cells (HUVECs) collected from pregnancies complicated by placental insufficiency-induced FGR compared to normal growth pregnancies. Our approach, involving transcriptomic profiling by RNA-sequencing and gene set enrichment analysis focused on cardiovascular and renal gene sets and targeted DNA methylation assays, contributes to the identification of targets underlying long-term cardiovascular and renal diseases., Results: Gene set enrichment analysis showed several downregulated gene sets, most of them involved in immune or inflammatory pathways or cell cycle pathways. seven of the 22 significantly upregulated gene sets related to kidney development and four gene sets involved with cardiovascular health and function were downregulated in FGR (n = 11) versus control (n = 8). Transcriptomic profiling by RNA-sequencing revealed downregulated expression of LGALS1, FPR3 and NRM and upregulation of lincRNA RP5-855F14.1 in FGR compared to controls. DNA methylation was similar for LGALS1 between study groups, but relative hypomethylation of FPR3 and hypermethylation of NRM were present in FGR, especially in male offspring. Absolute differences in methylation were, however, small., Conclusion: This study showed upregulation of gene sets related to renal development in HUVECs collected from pregnancies complicated by FGR compared to control donors. The differentially expressed gene sets related to cardiovascular function and health might be in line with the downregulated expression of NRM and upregulated expression of lincRNA RP5-855F14.1 in FGR samples; NRM is involved in cardiac remodeling, and lincRNAs are correlated with cardiovascular diseases. Future studies should elucidate whether the downregulated LGALS1 and FPR3 expressions in FGR are angiogenesis-modulating regulators leading to placental insufficiency-induced FGR or whether the expression of these genes can be used as a biomarker for increased cardiovascular risk. Altered DNA methylation might partly underlie FPR3 and NRM differential gene expression differences in a sex-dependent manner.
- Published
- 2020
- Full Text
- View/download PDF
35. Mid-gestation low-dose LPS administration results in female-specific excessive weight gain upon a western style diet in mouse offspring.
- Author
-
Dijkstra DJ, Verkaik-Schakel RN, Eskandar S, Limonciel A, Stojanovska V, Scherjon SA, and Plösch T
- Subjects
- Adipose Tissue metabolism, Animals, Appetite Regulation genetics, Female, Hypothalamus physiopathology, Insulin metabolism, Leptin metabolism, Liver metabolism, Mice, Inbred C57BL, Oxidative Stress drug effects, Pregnancy, Diet, High-Fat adverse effects, Hyperphagia etiology, Lipopolysaccharides administration & dosage, Lipopolysaccharides adverse effects, Maternal-Fetal Exchange physiology, Sex Characteristics, Weight Gain
- Abstract
Gestational complications, including preeclampsia and gestational diabetes, have long-term adverse consequences for offspring's metabolic and cardiovascular health. A low-grade systemic inflammatory response is likely mediating this. Here, we examine the consequences of LPS-induced gestational inflammation on offspring's health in adulthood. LPS was administered to pregnant C57Bl/6J mice on gestational day 10.5. Maternal plasma metabolomics showed oxidative stress, remaining for at least 5 days after LPS administration, likely mediating the consequences for the offspring. From weaning on, all offspring was fed a control diet; from 12 to 24 weeks of age, half of the offspring received a western-style diet (WSD). The combination of LPS-exposure and WSD resulted in hyperphagia and increased body weight and body fat mass in the female offspring. This was accompanied by changes in glucose tolerance, leptin and insulin levels and gene expression in liver and adipose tissue. In the hypothalamus, expression of genes involved in food intake regulation was slightly changed. We speculate that altered food intake behaviour is a result of dysregulation of hypothalamic signalling. Our results add to understanding of how maternal inflammation can mediate long-term health consequences for the offspring. This is relevant to many gestational complications with a pro-inflammatory reaction in place.
- Published
- 2020
- Full Text
- View/download PDF
36. Early-onset preeclampsia, plasma microRNAs, and endothelial cell function.
- Author
-
Lip SV, Boekschoten MV, Hooiveld GJ, van Pampus MG, Scherjon SA, Plösch T, and Faas MM
- Subjects
- Adult, Cell Movement, Female, Gene Expression Profiling, Gestational Age, Humans, Pregnancy, Young Adult, Human Umbilical Vein Endothelial Cells metabolism, MicroRNAs blood, Pre-Eclampsia blood
- Abstract
Background: Preeclampsia is a hypertensive pregnancy disorder in which generalized systemic inflammation and maternal endothelial dysfunction are involved in the pathophysiology. MiRNAs are small noncoding RNAs responsible for post-transcriptional regulation of gene expression and involved in many physiological processes. They mainly downregulate translation of their target genes., Objective: We aimed to compare the plasma miRNA concentrations in preeclampsia, healthy pregnant women, and nonpregnant women. Furthermore, we aimed to evaluate the effect of 3 highly increased plasma miRNAs in preeclampsia on endothelial cell function in vitro., Study Design: We compared 3391 (precursor) miRNA concentrations in plasma samples from early-onset preeclamptic women, gestational age-matched healthy pregnant women, and nonpregnant women using miRNA 3.1. arrays (Affymetrix) and validated our findings by real-time quantitative polymerase chain reaction. Subsequently, endothelial cells (human umbilical vein endothelial cells) were transfected with microRNA mimics (we choose the 3 miRNAs with the greatest fold change and lowest false-discovery rate in preeclampsia vs healthy pregnancy). After transfection, functional assays were performed to evaluate whether overexpression of the microRNAs in endothelial cells affected endothelial cell function in vitro. Functional assays were the wound-healing assay (which measures cell migration and proliferation), the proliferation assay, and the tube-formation assay (which assesses formation of endothelial cell tubes during the angiogenic process). To determine whether the miRNAs are able to decrease gene expression of certain genes, RNA was isolated from transfected endothelial cells and gene expression (by measuring RNA expression) was evaluated by gene expression microarray (Genechip Human Gene 2.1 ST arrays; Life Technologies). For the microarray, we used pooled samples, but the differently expressed genes in the microarray were validated by real-time quantitative polymerase chain reaction in individual samples., Results: No significant differences (fold change <-1.2 or >1.2 with a false-discovery rate <0.05) were found in miRNA plasma concentrations between healthy pregnant and nonpregnant women. The plasma concentrations of 26 (precursor) miRNAs were different between preeclampsia and healthy pregnancy. The 3 miRNAs that were increased with the greatest fold change and lowest false-discovery rate in preeclampsia vs healthy pregnancy were miR-574-5p, miR-1972, and miR-4793-3p. Transfection of endothelial cells with these miRNAs in showed that miR-574-5p decreased (P<.05) the wound-healing capacity (ie, decreased endothelial cell migration and/or proliferation) and tended (P<.1) to decrease proliferation, miR-1972 decreased tube formation (P<.05), and also tended (P<.1) to decrease proliferation, and miR-4793-3p tended (P<.1) to decrease both the wound-healing capacity and tube formation in vitro. Gene expression analysis of transfected endothelial cells revealed that miR-574-5p tended (P<.1) to decrease the expression of the proliferation marker MKI67., Conclusion: We conclude that in the early-onset preeclampsia group in our study different concentrations of plasma miRNAs are present as compared with healthy pregnancy. Our results suggest that miR-574-5p and miR-1972 decrease the proliferation (probably via decreasing MKI67) and/or migration as well as the tube-formation capacity of endothelial cells. Therefore, these miRNAs may be antiangiogenic factors affecting endothelial cells in preeclampsia., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
37. Prenatal smoke effect on mouse offspring Igf1 promoter methylation from fetal stage to adulthood is organ and sex specific.
- Author
-
Zeng Z, Meyer KF, Lkhagvadorj K, Kooistra W, Reinders-Luinge M, Xu X, Huo X, Song J, Plösch T, and Hylkema MN
- Subjects
- Animals, Animals, Newborn, Epigenesis, Genetic, Female, Fetal Growth Retardation etiology, Fetal Growth Retardation metabolism, Male, Mice, Organ Specificity, Pregnancy, Prenatal Exposure Delayed Effects etiology, Prenatal Exposure Delayed Effects metabolism, Sex Factors, DNA Methylation, Fetal Growth Retardation pathology, Gene Expression Regulation, Developmental, Insulin-Like Growth Factor I genetics, Prenatal Exposure Delayed Effects pathology, Promoter Regions, Genetic, Smoke adverse effects
- Abstract
Prenatal smoke exposure (PSE) is associated with reduced birth weight, impaired fetal development, and increased risk for diseases later in life. Changes in DNA methylation may be involved, as multiple large-scale epigenome-wide association studies showed that PSE is robustly associated with DNA methylation changes in blood among offspring in early life. Insulin-like growth factor-1 (IGF1) is important in growth, differentiation, and repair processes after injury. However, no studies investigated the organ-specific persistence of PSE-induced methylation change of Igf1 into adulthood. Based on our previous studies on the PSE effect on Igf1 promoter methylation in fetal and neonatal mouse offspring, we now have extended our studies to adulthood. Our data show that basal Igf1 promoter methylation generally increased in the lung but decreased in the liver (except for 2 persistent CpG sites in both organs) across three different developmental stages. PSE changed Igf1 promoter methylation in all three developmental stages, which was organ and sex specific. The PSE effect was less pronounced in adult offspring compared with the fetal and neonatal stages. In addition, the PSE effect in the adult stage was more pronounced in the lung compared with the liver. For most CpG sites, an inverse correlation was found for promoter methylation and mRNA expression when the data of all three stages were combined. This was more prominent in the liver. Our findings provide additional evidence for sex- and organ-dependent prenatal programming, which supports the developmental origins of health and disease (DOHaD) hypothesis.
- Published
- 2020
- Full Text
- View/download PDF
38. Adenosine kinase and cardiovascular fetal programming in gestational diabetes mellitus.
- Author
-
Silva L, Plösch T, Toledo F, Faas MM, and Sobrevia L
- Subjects
- Adenosine metabolism, Adenosine Kinase genetics, Animals, DNA Methylation, Diabetes Mellitus metabolism, Diabetes, Gestational genetics, Endothelium metabolism, Epigenomics, Female, Fetal Development genetics, Gene Expression Regulation, Humans, Inflammation, Mice, Pregnancy, Adenosine Kinase metabolism, Cardiovascular System metabolism, Diabetes, Gestational metabolism, Fetal Development physiology, Placenta metabolism
- Abstract
Gestational diabetes mellitus (GDM) is a detrimental condition for human pregnancy associated with endothelial dysfunction and endothelial inflammation in the fetoplacental vasculature and leads to increased cardio-metabolic risk in the offspring. In the fetoplacental vasculature, GDM is associated with altered adenosine metabolism. Adenosine is an important vasoactive molecule and is an intermediary and final product of transmethylation reactions in the cell. Adenosine kinase is the major regulator of adenosine levels. Disruption of this enzyme is associated with alterations in methylation-dependent gene expression regulation mechanisms, which are associated with the fetal programming phenomenon. Here we propose that cellular and molecular alterations associated with GDM can dysregulate adenosine kinase leading to fetal programming in the fetoplacental vasculature. This can contribute to the cardio-metabolic long-term consequences observed in offspring after exposure to GDM., (Copyright © 2019 Elsevier B.V. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
39. Gestational oxidative stress protects against adult obesity and insulin resistance.
- Author
-
Dimova LG, Battista S, Plösch T, Kampen RA, Liu F, Verkaik-Schakel RN, Pratico D, Verkade HJ, and Tietge UJF
- Subjects
- Adipose Tissue metabolism, Adiposity, Animals, Body Composition, Disease Models, Animal, Female, Fetal Growth Retardation etiology, Fetal Growth Retardation metabolism, Glucose metabolism, Lipid Metabolism, Male, Maternal Exposure, Mice, Mice, Transgenic, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha genetics, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha metabolism, Pregnancy, Prenatal Exposure Delayed Effects, Superoxide Dismutase genetics, Superoxide Dismutase metabolism, Insulin Resistance, Obesity metabolism, Oxidative Stress
- Abstract
Pregnancy complications such as preeclampsia cause increased fetal oxidative stress and fetal growth restriction, and associate with a higher incidence of adult metabolic syndrome. However, the pathophysiological contribution of oxidative stress per se is experimentally difficult to discern and has not been investigated. This study determined, if increased intrauterine oxidative stress (IUOx) affects adiposity, glucose and cholesterol metabolism in adult Ldlr-/-xSod2+/+ offspring from crossing male Ldlr-/-xSod2+/+ mice with Ldlr-/-xSod2 +/- dams (IUOx) or Ldlr-/-xSod2 +/- males with Ldlr-/-xSod2+/+ dams (control). At 12 weeks of age mice received Western diet for an additional 12 weeks. Adult male IUOx offspring displayed lower body weight and reduced adiposity associated with improved glucose tolerance compared to controls. Reduced weight gain in IUOx was conceivably due to increased energy dissipation in white adipose tissue conveyed by higher expression of Ucp1 and an accompanying decrease in DNA methylation in the Ucp1 enhancer region. Female offspring did not show comparable phenotypes. These results demonstrate that fetal oxidative stress protects against the obesogenic effects of Western diet in adulthood by programming energy dissipation in white adipose tissue at the level of Ucp1., (Copyright © 2019 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
40. Differential placental DNA methylation of VEGFA and LEP in small-for-gestational age fetuses with an abnormal cerebroplacental ratio.
- Author
-
Bekkering I, Leeuwerke M, Tanis JC, Schoots MH, Verkaik-Schakel RN, Plösch T, Bilardo CM, Eijsink JJH, Bos AF, and Scherjon SA
- Subjects
- Adult, Alleles, CpG Islands, Epigenesis, Genetic, Female, Gene Expression Regulation, Gestational Age, Humans, Infant, Small for Gestational Age, Pregnancy, Ultrasonography, Prenatal, Young Adult, DNA Methylation, Leptin genetics, Placenta metabolism, Vascular Endothelial Growth Factor A genetics
- Abstract
Background: In Fetal Growth Restriction 'fetal programming' may take place via DNA methylation, which has implications for short-term and long-term health outcomes. Small-for-gestational age fetuses are considered fetal growth restricted, characterized by brain-sparing when fetal Doppler hemodynamics are abnormal, expressed as a cerebroplacental ratio (CPR) <1. We aimed to determine whether brain-sparing is associated with altered DNA methylation of selected genes., Methods: We compared DNA methylation of six genes in 41 small-for-gestational age placentas with a normal or abnormal CPR. We selected EPO, HIF1A, VEGFA, LEP, PHLDA2, and DHCR24 for their role in angiogenesis, immunomodulation, and placental and fetal growth. DNA methylation was analyzed by pyrosequencing., Results: Growth restricted fetuses with an abnormal CPR showed hypermethylation of the VEGFA gene at one CpG (VEGFA-309, p = .001) and an overall hypomethylation of the LEP gene, being significant at two CpGs (LEP-123, p = .049; LEP-51, p = .020). No differences in methylation were observed for the other genes., Conclusions: VEGFA and LEP genes are differentially methylated in placentas of small-for-gestational age fetuses with brain-sparing. Hypermethylation of VEGFA-309 in abnormal CPR-placentas could indicate successful compensatory mechanisms. Methylation of LEP-51 is known to suppress LEP expression. Hypomethylation in small-for-gestational age placentas with abnormal CPR may result in hyperleptinemia and predispose to leptin-resistance later in life., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2019
- Full Text
- View/download PDF
41. In utero sFlt-1 exposure differentially affects gene expression patterns in fetal liver.
- Author
-
Stojanovska V, Holwerda KM, van der Graaf AM, Verkaik-Schakel RN, Boekschoten MV, Faas MM, Scherjon SA, and Plösch T
- Subjects
- Animals, Female, Fetal Growth Retardation etiology, Fetal Growth Retardation pathology, Fetus pathology, Gene Expression Profiling, Liver pathology, Male, Pregnancy, Prenatal Exposure Delayed Effects etiology, Prenatal Exposure Delayed Effects pathology, Rats, Rats, Sprague-Dawley, Fetal Growth Retardation metabolism, Fetus metabolism, Gene Expression Regulation, Liver metabolism, Prenatal Exposure Delayed Effects metabolism, Vascular Endothelial Growth Factor Receptor-1 adverse effects
- Abstract
The soluble fms-like tyrosine kinase factor 1 (sFlt-1) is a major contributor to antiangiogenesis during preeclampsia. However, little is known about the effects of sFlt-1 on fetal health. In this study we aim to evaluate the effects of the sFlt-1 concentration during pregnancy on fetal liver physiology. We used adenoviral gene delivery in Sprague-Dawley dams (seven females, 10 weeks old) during mid-gestation (gestational day 8) with adenovirus overexpressing sFlt-1, and age-matched controls (six females, 10 weeks old) with empty adenoviral virus in order to quantify the sFlt-1 concentrations in pregnant dams. Dams exposed to adenoviral sFlt-1 delivery were subdivided into a low (n=4) and high sFlt-1 (n=3) group based on host response to the virus. One-way analysis of variance showed that fetuses (five per dam) exposed to high sFlt-1 concentrations in utero show fetal growth restriction (1.84±0.043 g high sFlt-1 v. 2.32±0.036 g control; mean (M)±s.e.m.; P<0.001), without hypertension or proteinuria in the dams. In continuation, the microarray analysis of the fetal liver of the high sFlt-1 group showed significant enrichment of key genes for fatty acid metabolism and Ppara targets. In addition, using pyrosequencing, we found that the Ppara enrichment in the high sFlt-1 group is accompanied by decreased methylation of its promoter (1.89±0.097 mean % methylation in high sFlt-1 v. 2.26±0.095 mean % methylation in control, M±s.e.m., P<0.02). Our data show that high sFlt-1 concentrations during pregnancy have detrimental effects on the fatty acid metabolism genes and the Ppara targets in the fetal liver.
- Published
- 2019
- Full Text
- View/download PDF
42. More Maternal Vascular Malperfusion and Chorioamnionitis in Placentas After Expectant Management vs. Immediate Delivery in Fetal Growth Restriction at (Near) Term: A Further Analysis of the DIGITAT Trial.
- Author
-
Feenstra ME, Schoots MH, Plösch T, Prins JR, Scherjon SA, Timmer A, van Goor H, and Gordijn SJ
- Abstract
Objective: Management of late fetal growth restriction (FGR) is limited to adequate fetal monitoring and optimal timing of delivery. The Disproportionate Intrauterine Growth Intervention Trial At Term (DIGITAT) trial compared induction of labor with expectant management in pregnancies at (near) term complicated by suspected FGR. Findings of the DIGITAT trial were that expectant monitoring prolonged pregnancy for 10 days and increased birth weight with only 130 grams. This resulted in more infants born below the 2.3rd percentile compared to induction of labor, respectively, 12.5% in induction of labor and 30.6% in expectant monitoring group. The main placental lesions associated with FGR are maternal vascular malperfusion, fetal vascular malperfusion, and villitis of unknown etiology. We investigated whether placentas of pregnancies complicated with FGR in the expectant monitoring group reveal more and more severe pathology due to pregnancy prolongation. Material and methods: The DIGITAT trial was a multicenter, randomized controlled trial with suspected FGR beyond 36 + 0 weeks. We now analyzed all available cases ( n = 191) for placental pathology. The macroscopic details were collected and histological slides were recorded and classified by a single perinatal pathologist, blinded for pregnancy details and outcome. The different placental lesions were scored based on the latest international criteria for placental lesions as defined in the Amsterdam Placental Workshop Group Consensus Statement. Results: The presence of maternal vascular malperfusion and chorioamnionitis were higher in the expectant management group ( p < 0.05 and p < 0.01, respectively). No differences in placental weight and maturation of the placenta between the induction of labor and the expectant management group were seen. Fetal vascular malperfusion, villitis of unknown etiology and nucleated red blood cell count did not differ between the groups. Conclusion: Expectant management of late FGR is associated with increased maternal vascular malperfusion and chorioamnionitis. This may have implications for fetal and neonatal outcome, such as programming in the developing child influencing health outcomes later in life.
- Published
- 2019
- Full Text
- View/download PDF
43. Human sFLT1 Leads to Severe Changes in Placental Differentiation and Vascularization in a Transgenic hsFLT1/rtTA FGR Mouse Model.
- Author
-
Vogtmann R, Kühnel E, Dicke N, Verkaik-Schakel RN, Plösch T, Schorle H, Stojanovska V, Herse F, Köninger A, Kimmig R, Winterhager E, and Gellhaus A
- Abstract
The anti-angiogenic soluble fms-like tyrosine kinase 1 (sFLT1) is one of the candidates in the progression of preeclampsia, often associated with fetal growth restriction (FGR). Therapeutic agents against preeclampsia with/without FGR, as well as adequate transgenic sFLT1 mouse models for testing such agents, are still missing. Much is known about sFLT1-mediated endothelial dysfunction in several tissues; however, the influence of sFLT1 on placental and fetal development is currently unknown. We hypothesize that sFLT1 is involved in the progression of FGR by influencing placental differentiation and vascularization and is a prime candidate for interventional strategies. Therefore, we generated transgenic inducible human sFLT1/reverse tetracycline-controlled transactivator (hsFLT1/rtTA) mice, in which hsFLT1 is ubiquitously overexpressed during pregnancy in dams and according to the genetics in hsFLT1/rtTA homozygous and heterozygous fetuses. Induction of hsFLT1 led to elevated hsFLT1 levels in the serum of dams and on mRNA level in all placentas and hetero-/homozygous fetuses, resulting in FGR in all fetuses at term. The strongest effects in respect to FGR were observed in the hsFLT1/rtTA homozygous fetuses, which exhibited the highest hsFLT1 levels. Only fetal hsFLT1 expression led to impaired placental morphology characterized by reduced placental efficiency, enlarged maternal sinusoids, reduced fetal capillaries, and impaired labyrinthine differentiation, associated with increased apoptosis. Besides impaired placental vascularization, the expression of several transporter systems, such as glucose transporter 1 and 3 ( Glut-1 ; Glut-3 ); amino acid transporters, solute carrier family 38, member one and two ( Slc38a1 ; Slc38a2 ); and most severely the fatty acid translocase Cd36 and fatty acid binding protein 3 ( Fabp3 ) was reduced upon hsFLT1 expression, associated with an accumulation of phospholipids in the maternal serum. Moreover, the Vegf pathway showed alterations, resulting in reduced Vegf, Vegfb, and Plgf protein levels and increased Bad and Caspase 9 mRNA levels. We suggest that hsFLT1 exerts an inhibitory influence on placental vascularization by reducing Vegf signaling, which leads to apoptosis in fetal vessels, impairing placental differentiation, and the nutrient exchange function of the labyrinth. These effects were more pronounced when both the dam and the fetus expressed hsFLT1 and ultimately result in FGR and resemble the preeclamptic phenotype in humans.
- Published
- 2019
- Full Text
- View/download PDF
44. A double-hit pre-eclampsia model results in sex-specific growth restriction patterns.
- Author
-
Stojanovska V, Dijkstra DJ, Vogtmann R, Gellhaus A, Scherjon SA, and Plösch T
- Subjects
- Animals, Disease Models, Animal, Female, Fetal Growth Retardation pathology, Gene Expression Regulation, Developmental, Lipopolysaccharides, Male, Membrane Transport Proteins genetics, Membrane Transport Proteins metabolism, Metabolome, Metabolomics, Mice, Inbred C57BL, Phosphatidylcholines blood, Placenta metabolism, Placenta pathology, Pre-Eclampsia blood, Pre-Eclampsia metabolism, Pregnancy, Vascular Endothelial Growth Factor Receptor-1 metabolism, Pre-Eclampsia pathology
- Abstract
Pre-eclampsia is a multifactorial pregnancy-associated disorder characterized by angiogenic dysbalance and systemic inflammation; however, animal models that combine these two pathophysiological conditions are missing. Here, we introduce a novel double-hit pre-eclampsia mouse model that mimics the complex multifactorial conditions present during pre-eclampsia and allows for the investigation of early consequences for the fetus. Adenoviral overexpression of soluble fms-like tyrosine kinase (sFlt-1) and lipopolysaccharide (LPS) administration at mid-gestation in pregnant mice resulted in hypertension and albuminuria comparable to that of the manifestation in humans. A metabolomics analysis revealed that pre-eclamptic dams have increased plasma concentrations of phosphadytilcholines. The fetuses of both sexes were growth restricted; however, in males a brain-sparing effect was seen as compensation for this growth restriction. According to the plasma metabolomics, male fetuses showed changes in amino acid metabolism, while female fetuses showed pronounced alterations in lipid metabolism. Our results show that combined exposure to sFlt-1 and LPS mimics the clinical symptoms of pre-eclampsia and affects fetal growth in a sex-specific manner, with accompanying metabolome changes., Competing Interests: Competing interestsThe authors declare no competing or financial interests., (© 2019. Published by The Company of Biologists Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
45. The influence of maternal obesity on macrophage subsets in the human decidua.
- Author
-
Laskewitz A, van Benthem KL, Kieffer TEC, Faas MM, Verkaik-Schakel RN, Plösch T, Scherjon SA, and Prins JR
- Subjects
- Adult, Antigens, CD analysis, Antigens, Differentiation, Myelomonocytic analysis, Female, HLA-DR Antigens analysis, Humans, Pregnancy, Receptors, Cell Surface analysis, Decidua immunology, Macrophages classification, Obesity, Maternal immunology
- Abstract
Obesity is seen as a low grade inflammatory state, and is associated with adverse pregnancy outcomes. Disturbed macrophage characteristics might be essential in obesity associated pregnancy pathology via effects on the regulation of angiogenesis and placental development. This study aims to address the effects of maternal obesity on macrophage subsets in the decidua of women with term uncomplicated pregnancies. Macrophages were isolated from the decidua basalis and decidua parietalis of women with pre-gravid BMI < 25 (control) and BMI > 30 (obese). Macrophages were characterized and quantified using multi-color flow cytometry. Placentas of 10 obese and 10 control women after an uncomplicated term pregnancy were included. The decidua parietalis, but not decidua basalis, showed significantly lower levels of M1-type (HLA-DR
+ , CD163- ) macrophages (p < 0.05) in obese women (4,3% of total macrophages) compared to control women (5,3% of total macrophages). The lower levels of M1 macrophages, considered to be pro-inflammatory, might indicate a mechanism to compensate for the pro-inflammatory environment in obese women to ensure healthy pregnancy outcomes., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
46. The Possible Role of Placental Morphometry in the Detection of Fetal Growth Restriction.
- Author
-
Salavati N, Smies M, Ganzevoort W, Charles AK, Erwich JJ, Plösch T, and Gordijn SJ
- Abstract
Fetal growth restriction (FGR) is often the result of placental insufficiency and is characterized by insufficient transplacental transport of nutrients and oxygen. The main underlying entities of placental insufficiency, the pathophysiologic mechanism, can broadly be divided into impairments in blood flow and exchange capacity over the syncytiovascular membranes of the fetal placenta villi. Fetal growth restriction is not synonymous with small for gestational age and techniques to distinguish between both are needed. Placental insufficiency has significant associations with adverse pregnancy outcomes (perinatal mortality and morbidity). Even in apparently healthy survivors, altered fetal programming may lead to long-term neurodevelopmental and metabolic effects. Although the concept of fetal growth restriction is well appreciated in contemporary obstetrics, the appropriate detection of FGR remains an issue in clinical practice. Several approaches have aimed to improve detection, e.g., uniform definition of FGR, use of Doppler ultrasound profiles and use of growth trajectories by ultrasound fetal biometry. However, the role of placental morphometry (placental dimensions/shape and weight) deserves further exploration. This review article covers the clinical relevance of placental morphometry during pregnancy and at birth to help recognize fetuses who are growth restricted. The assessment has wide intra- and interindividual variability with various consequences. Previous studies have shown that a small placental surface area and low placental weight are associated with a slower growth of the fetus. Parameters such as placental surface area, placental volume and placental weight in relation to birth weight can help to identify FGR. In the future, a model including sophisticated antenatal placental morphometry may prove to be a clinically useful method for screening or diagnosing growth restricted fetuses, in order to provide optimal monitoring.
- Published
- 2019
- Full Text
- View/download PDF
47. Placental insufficiency contributes to fatty acid metabolism alterations in aged female mouse offspring.
- Author
-
Stojanovska V, Sharma N, Dijkstra DJ, Scherjon SA, Jäger A, Schorle H, and Plösch T
- Subjects
- Animals, Cell Differentiation genetics, Diabetes Mellitus, Type 2 metabolism, Female, Fetal Growth Retardation metabolism, Lipid Metabolism physiology, Mice, Transgenic, Obesity metabolism, Placental Insufficiency metabolism, Pregnancy, Pregnancy Proteins genetics, Aging, Fatty Acids metabolism, Lipid Metabolism genetics, Placenta metabolism, Pregnancy Proteins metabolism
- Abstract
Intrauterine growth restriction (IUGR) is an accepted risk factor for metabolic disorders in later life, including obesity and type 2 diabetes. The level of metabolic dysregulation can vary between subjects and is dependent on the severity and the type of IUGR insult. Classical IUGR animal models involve nutritional deprivation of the mother or uterine artery ligation. The latter aims to mimic a placental insufficiency, which is the most frequent cause of IUGR. In this study, we investigated whether IUGR attributable to placental insufficiency impacts the glucose and lipid homeostasis at advanced age. Placental insufficiency was achieved by deletion of the transcription factor AP-2y ( Tfap2c), which serves as one of the major trophoblast differentiation regulators. TdelT-IUGR mice were obtained by crossing mice with a floxed Tfap2c allele and mice with Cre recombinase under the control of the Tpbpa promoter. In advanced adulthood (9-12 mo), female and male IUGR mice are respectively 20% and 12% leaner compared with controls. At this age, IUGR mice have unaffected glucose clearance and lipid parameters (cholesterol, triglycerides, and phospholipids) in the liver. However, female IUGR mice have increased plasma free fatty acids (+87%) compared with controls. This is accompanied by increased mRNA levels of fatty acid synthase and endoplasmic reticulum stress markers in white adipose tissue. Taken together, our results suggest that IUGR by placental insufficiency may lead to higher lipogenesis in female mice in advanced adulthood, at least indicated by greater Fasn expression. This effect was sex specific for the aged IUGR females.
- Published
- 2018
- Full Text
- View/download PDF
48. Diabetes Mellitus in Pregnancy Leads to Growth Restriction and Epigenetic Modification of the Srebf2 Gene in Rat Fetuses.
- Author
-
Golic M, Stojanovska V, Bendix I, Wehner A, Herse F, Haase N, Kräker K, Fischer C, Alenina N, Bader M, Schütte T, Schuchardt M, van der Giet M, Henrich W, Muller DN, Felderhoff-Müser U, Scherjon S, Plösch T, and Dechend R
- Subjects
- Animals, DNA Methylation genetics, Diabetes Mellitus genetics, Diabetes Mellitus physiopathology, Diabetes, Gestational physiopathology, Female, Hyperglycemia genetics, Hyperglycemia physiopathology, Hyperinsulinism genetics, Hyperinsulinism physiopathology, Immunohistochemistry, Pregnancy, Rats, Real-Time Polymerase Chain Reaction methods, Statistics, Nonparametric, Diabetes, Gestational genetics, Epigenesis, Genetic genetics, Fetal Growth Retardation genetics, Insulin Resistance genetics, Pregnancy, Animal, Sterol Regulatory Element Binding Proteins genetics
- Abstract
Diabetic pregnancy is correlated with increased risk of metabolic and neurological disorders in the offspring putatively mediated epigenetically. Little is known about epigenetic changes already present in fetuses of diabetic pregnancies. We aimed at characterizing the perinatal environment after preexisting maternal diabetes mellitus and at identifying relevant epigenetic changes in the fetus. We focused on the transcription factor Srebf2 (sterol regulatory element binding transcription factor 2), a master gene in regulation of cholesterol metabolism. We tested whether diabetic pregnancy induces epigenetic changes in the Srebf2 promoter and if they become manifest in altered Srebf2 gene expression. We worked with a transgenic rat model of type 2 diabetes mellitus (Tet29) in which the insulin receptor is knocked down by doxycycline-induced RNA interference. Doxycycline was administered preconceptionally to Tet29 and wild-type control rats. Only Tet29 doxycycline dams were hyperglycemic, hyperinsulinemic, and hyperlipidemic. Gene expression was analyzed with quantitative real-time reverse transcriptase polymerase chain reaction and CpG promoter methylation with pyrosequencing. Immunohistochemistry was performed on fetal brains. Fetuses from diabetic Tet29 dams were hyperglycemic and growth restricted at the end of pregnancy. They further displayed decreased liver and brain weight with concomitant decreased microglial activation in the hippocampus in comparison to fetuses of normoglycemic mothers. Importantly, diabetic pregnancy induced CpG hypermethylation of the Srebf2 promoter in the fetal liver and brain, which was associated with decreased Srebf2 gene expression. In conclusion, diabetic and hyperlipidemic pregnancy induces neurological, metabolic, and epigenetic alterations in the rat fetus. Srebf2 is a potential candidate mediating intrauterine environment-driven epigenetic changes and later diabetic offspring health., (© 2018 American Heart Association, Inc.)
- Published
- 2018
- Full Text
- View/download PDF
49. Immune-modulatory effects of syncytiotrophoblast extracellular vesicles in pregnancy and preeclampsia.
- Author
-
Göhner C, Plösch T, and Faas MM
- Subjects
- Adaptive Immunity, Animals, Awards and Prizes, Exosomes immunology, Exosomes pathology, Exosomes physiology, Extracellular Vesicles pathology, Extracellular Vesicles physiology, Female, Humans, Immunity, Innate, Placenta immunology, Placenta pathology, Placenta physiology, Placenta physiopathology, Pre-Eclampsia pathology, Pregnancy, Trophoblasts cytology, Trophoblasts pathology, Trophoblasts physiology, Extracellular Vesicles immunology, Immunomodulation, Models, Immunological, Pre-Eclampsia immunology, Pre-Eclampsia therapy, Trophoblasts immunology
- Abstract
Unique immunologic adaptations exist to successfully establish and maintain pregnancy and to avoid an immune attack against the semi allogenic fetus. These adaptations occur both locally at the maternofetal interface and in the peripheral circulation and affect the innate as well as the adaptive immune system. Pregnancy is characterized by a general inflammatory state with activation of monocytes and granulocytes, but also with suppressive lymphocytes (regulatory T cells), and skewing towards T helper 2 immunity. The pregnancy complication preeclampsia is associated with an exaggerated inflammatory state and predominance of T helper 1 and 17 immunity. The syncytiotrophoblast has been found to secrete extracellular vesicles as communication factors into the maternal circulation. Syncytiotrophoblast extracellular vesicles from normal pregnancy have been shown to interact with monocytes, granulocytes, T cells and natural killer cells and influence the function of these cells. In doing so, they may support the inflammatory state of normal pregnancy as well as the suppressive lymphocyte phenotype. During preeclampsia, syncytiotrophoblast extracellular vesicles are not only increased in numbers but also showed an altered molecular load. Based on data from in vitro studies, it can be suggested that syncytiotrophoblast extracellular vesicles from preeclamptic pregnancies may support the exaggerated inflammatory state during preeclampsia. In this review, we discuss the immunological functions of syncytiotrophoblast extracellular vesicles and their involvement in adapting the maternal peripheral immunological adaptations to pregnancy., (Copyright © 2017 IFPA, Elsevier Ltd. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
50. Experimental preeclampsia in rats affects vascular gene expression patterns.
- Author
-
Lip SV, van der Graaf AM, Wiegman MJ, Scherjon SA, Boekschoten MV, Plösch T, and Faas MM
- Subjects
- Animals, Aorta pathology, Aorta physiopathology, Female, Pre-Eclampsia genetics, Pre-Eclampsia pathology, Pre-Eclampsia physiopathology, Pregnancy, Rats, Rats, Wistar, Aorta metabolism, Gene Expression Profiling, Gene Expression Regulation, Oligonucleotide Array Sequence Analysis, Pre-Eclampsia metabolism
- Abstract
Normal pregnancy requires adaptations of the maternal vasculature. During preeclampsia these adaptations are not well established, which may be related to maternal hypertension and proteinuria. The effects of preeclampsia on the maternal vasculature are not yet fully understood. We aimed to evaluate gene expression in aortas of pregnant rats with experimental preeclampsia using a genome wide microarray. Aortas were isolated from pregnant Wistar outbred rats with low-dose LPS-induced preeclampsia (ExpPE), healthy pregnant (Pr), non-pregnant and low-dose LPS-infused non-pregnant rats. Gene expression was measured by microarray and validated by real-time quantitative PCR. Gene Set Enrichment Analysis was performed to compare the groups. Functional analysis of the aorta was done by isotonic contraction measurements while stimulating aortic rings with potassium chloride. 526 genes were differentially expressed, and positive enrichment of "potassium channels", "striated muscle contraction", and "neuronal system" gene sets were found in ExpPE vs. Pr. The potassium chloride-induced contractile response of ExpPE aortic rings was significantly decreased compared to this response in Pr animals. Our data suggest that potassium channels, neuronal system and (striated) muscle contraction in the aorta may play a role in the pathophysiology of experimental preeclampsia. Whether these changes are also present in preeclamptic women needs further investigation.
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.