Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
1.
Cell Death Dis ; 15(8): 575, 2024 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-39117607

RESUMO

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 ß-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.


Assuntos
Retardo do Crescimento Fetal , Galectina 3 , Placenta , Retardo do Crescimento Fetal/metabolismo , Retardo do Crescimento Fetal/genética , Gravidez , Feminino , Animais , Placenta/metabolismo , Camundongos , Galectina 3/metabolismo , Galectina 3/deficiência , Galectina 3/genética , Masculino , Microbioma Gastrointestinal , Camundongos Endogâmicos C57BL , Humanos , Desenvolvimento Fetal , Fator de Crescimento Insulin-Like II/metabolismo , Fator de Crescimento Insulin-Like II/genética , Fator de Crescimento Insulin-Like II/deficiência , Trofoblastos/metabolismo
2.
Clin Epigenetics ; 16(1): 104, 2024 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-39138531

RESUMO

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.


Assuntos
Metilação de DNA , Epigênese Genética , Estudo de Associação Genômica Ampla , Glicina , Metaboloma , Serina , Humanos , Glicina/sangue , Serina/sangue , Serina/genética , Metilação de DNA/genética , Masculino , Feminino , Estudo de Associação Genômica Ampla/métodos , Metaboloma/genética , Epigênese Genética/genética , Pessoa de Meia-Idade , Ilhas de CpG/genética , Epigenoma/genética , Adulto , Idoso , Análise da Randomização Mendeliana
3.
J Reprod Immunol ; 164: 104284, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38908337

RESUMO

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 FLT1high 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 hsFLT1 high 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.


Assuntos
Placenta , Pré-Eclâmpsia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular , Gravidez , Feminino , Animais , Humanos , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Camundongos , Pré-Eclâmpsia/metabolismo , Placenta/metabolismo , Glicosilação , Galectinas/metabolismo , Neovascularização Patológica/metabolismo , Modelos Animais de Doenças
5.
Pediatr Obes ; 19(6): e13120, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38590200

RESUMO

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.


Assuntos
Células-Tronco Mesenquimais , Obesidade Materna , Efeitos Tardios da Exposição Pré-Natal , Humanos , Feminino , Gravidez , Obesidade Materna/metabolismo , Tecido Adiposo , Obesidade Infantil , Adipogenia/fisiologia , Recém-Nascido , Adipócitos
6.
ACS Chem Neurosci ; 15(6): 1074-1083, 2024 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-38421943

RESUMO

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.


Assuntos
Efeitos Tardios da Exposição Pré-Natal , Inibidores Seletivos de Recaptação de Serotonina , Lactente , Feminino , Humanos , Masculino , Gravidez , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Inibidores Seletivos de Recaptação de Serotonina/uso terapêutico , Fluoxetina/farmacologia , Fluoxetina/uso terapêutico , Gestantes , Transcriptoma , Placenta/metabolismo , Perfilação da Expressão Gênica , Efeitos Tardios da Exposição Pré-Natal/metabolismo
7.
ACS Cent Sci ; 9(9): 1784-1798, 2023 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-37780363

RESUMO

Cumulus granulosa cells (cGCs) and mural granulosa cells (mGCs), although derived from the same precursors, are anatomically and functionally heterogeneous. They are critical for female fertility by supporting oocyte competence and follicular development. There are various techniques used to investigate the role of free radicals in mGCs and cCGs. Yet, temporospatial resolution remains a challenge. We used a quantum sensing approach to study free radical generation at nanoscale in cGCs and mGCs isolated from women undergoing oocyte retrieval during in vitro fertilization (IVF). Cells were incubated with bare fluorescent nanodiamonds (FNDs) or mitochondria targeted FNDs to detect free radicals in the cytoplasm and mitochondria. After inducing oxidative stress with menadione, we continued to detect free radical generation for 30 min. We observed an increase in free radical generation in cGCs and mGCs from 10 min on. Although cytoplasmic and mitochondrial free radical levels are indistinguishable in the physiological state in both cGCs and mGCs, the free radical changes measured in mitochondria were significantly larger in both cell types, suggesting mitochondria are sites of free radical generation. Furthermore, we observed later occurrence and a smaller percentage of cytoplasmic free radical change in cGCs, indicating that cGCs may be more resistant to oxidative stress.

8.
Front Mol Neurosci ; 16: 1211373, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37790884

RESUMO

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.

9.
Early Hum Dev ; 186: 105868, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37797474

RESUMO

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.


Assuntos
Fator Neurotrófico Derivado do Encéfalo , Metilação de DNA , Lactente , Recém-Nascido , Humanos , Estudos Prospectivos , Fator Neurotrófico Derivado do Encéfalo/genética , Lactente Extremamente Prematuro , Idade Gestacional , Unidades de Terapia Intensiva Neonatal , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética
10.
Epigenomics ; 15(8): 479-486, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-37309586

RESUMO

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.


What is this article about? Necrotizing enterocolitis (NEC) is a common emergency condition affecting the gastrointestinal system of preterm infants. Epigenetic changes in DNA methylation may be present in infants before the onset of NEC. DNA methylation is a natural process that can help turn genes on or off, thereby affecting their function. This study focused on measuring the amount of DNA methylation in certain genes in preterm infants who developed NEC. What were the results? This study included 24 preterm infants with NEC and 45 matched healthy controls. The researchers isolated human DNA from stool samples, and the amount of DNA methylation of four specific genes was measured. They found that one of the genes, CTDSPL2, had significantly higher DNA methylation in infants who later developed NEC than in healthy infants. What do the results of the study mean? In this study, researchers found that CTDSPL2 showed a higher level of DNA methylation in stool samples of infants who later developed NEC. The effect of this change remains unclear, but may affect the way cells grow and respond to injury or infection, which could contribute to the development of NEC. Measuring DNA methylation in stool samples provides a noninvasive method for identifying DNA methylation changes in preterm infants. Comparing the amount of DNA methylation in healthy infants with that in preterm infants at risk of NEC may help predict the risk of developing NEC.


Assuntos
Enterocolite Necrosante , Recém-Nascido Prematuro , Humanos , Lactente , Recém-Nascido , Metilação de DNA , Enterocolite Necrosante/genética , Fezes
11.
Pediatr Res ; 94(4): 1365-1372, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37208432

RESUMO

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 (rsSO2). Using a piglet model of asphyxia, we aimed to correlate changes in rsSO2 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 rsSO2 and calculated mean rsSO2 and variability of rsSO2 (rsCoVar = 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 rsSO2 and gene expression were observed. However, lower rsCoVar 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 rsSO2. Our findings may advance future research and clinical practice regarding resuscitation strategies of preterm infants.


Assuntos
Hipóxia , Recém-Nascido Prematuro , Animais , Humanos , Recém-Nascido , Animais Recém-Nascidos , Expressão Gênica , Inflamação/complicações , Intestinos , Oxigênio , Suínos , Distribuição Aleatória , Modelos Animais de Doenças
12.
Int J Mol Sci ; 24(8)2023 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-37108049

RESUMO

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.


Assuntos
Pré-Eclâmpsia , Humanos , Gravidez , Camundongos , Feminino , Masculino , Animais , Pré-Eclâmpsia/metabolismo , Placenta/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Feto/metabolismo , Camundongos Transgênicos , Aumento de Peso , Retardo do Crescimento Fetal/genética
13.
J Dev Orig Health Dis ; 14(1): 146-151, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-35748176

RESUMO

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.


Assuntos
Metilação de DNA , Pré-Eclâmpsia , Animais , Feminino , Humanos , Masculino , Camundongos , Gravidez , Encéfalo/metabolismo , Expressão Gênica , Fígado/metabolismo , Pré-Eclâmpsia/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular
14.
Clin Epigenetics ; 14(1): 170, 2022 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-36503539

RESUMO

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-R2 = 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.


Assuntos
Distonia , Distúrbios Distônicos , Humanos , Metilação de DNA , Serotonina , Distonia/genética , Distonia/complicações , Distúrbios Distônicos/complicações , Distúrbios Distônicos/genética , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética
15.
Oxid Med Cell Longev ; 2022: 1070968, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36466095

RESUMO

Ovarian aging is associated with a decrease in fecundity. Increased oxidative stress of granulosa cells (GCs) is an important contributor. We thus asked whether there is an oxidative stress-related gene signature in GCs associated with ovarian aging. Public nonhuman primate (NHP) single-cell transcriptome was processed to identify GC cluster. Then, a GC signature for ovarian aging was established based on six oxidative stress-related differentially expressed genes (MAPK1, STK24, AREG, ATG7, ANXA1, and PON2). Receiver operating characteristic (ROC) analysis confirmed good discriminating capacity in both NHP single-cell and human bulk transcriptome datasets. Gene expression levels were investigated using qPCR in the human ovarian granulosa-like tumor cell line (KGN) and mouse GCs. In an oxidative stress model, KGN cells were treated with menadione (7.5 µM, 24 h) to induce oxidative stress, after which upregulation of MAPK1, STK24, ATG7, ANXA1, and PON2 and downregulation of AREG were observed (p < 0.05). In an aging model, KGN cells were continuously cultured for 3 months, leading to increased expressions of all genes (p < 0.05). In GCs of reproductively aged (8-month-old) Kunming mice, upregulated expression of Mapk1, Stk24, Atg7, and Pon2 and downregulated expression of Anxa1 and Areg were observed (p < 0.01). We therefore here identify a six-gene GC signature associated with oxidative stress and ovarian aging.


Assuntos
Células da Granulosa , Ovário , Feminino , Humanos , Camundongos , Animais , Lactente , Estresse Oxidativo/genética , Envelhecimento/genética , Vitamina K 3 , Proteínas Serina-Treonina Quinases
16.
Front Endocrinol (Lausanne) ; 13: 895489, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36046788

RESUMO

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.


Assuntos
Diabetes Mellitus , Hiperglicemia , Estado Pré-Diabético , Exercício Físico , Feminino , Humanos , Menopausa/genética , Estado Pré-Diabético/genética , Estado Pré-Diabético/terapia
17.
Eur J Prev Cardiol ; 29(17): 2183-2199, 2022 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-35989414

RESUMO

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.


Assuntos
Doenças Cardiovasculares , Humanos , Prevenção Secundária , Doenças Cardiovasculares/genética , Doenças Cardiovasculares/prevenção & controle , Exercício Físico
18.
Front Pediatr ; 10: 876803, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35722484

RESUMO

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.

20.
Front Cell Neurosci ; 16: 797588, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35496908

RESUMO

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.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA