Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Physiol Rep ; 10(23): e15518, 2022 12.
Article in English | MEDLINE | ID: mdl-36461654

ABSTRACT

Intrauterine growth restriction (IUGR) and exposure to a high-fat diet (HFD) independently increase the risk of cardiovascular disease (CVD) and hyperlipidemia. In our previous studies, IUGR increased blood pressure and promoted vascular remodeling and stiffness in early life, a finding that persisted and was augmented by a maternal HFD through postnatal day (PND) 60. The impact of these findings with aging and the development of hyperlipidemia and atherosclerosis remain unknown. We hypothesized that the previously noted impact of IUGR on hypertension, vascular remodeling, and hyperlipidemia would persist. Adult female rats were fed either a regular diet (RD) or high fat diet (HFD) prior to conception through lactation. IUGR was induced by uterine artery ligation. Offspring were weaned to either RD or HFD through PND 365. For both control (C) and IUGR (I) and rats, this resulted in the following six groups per sex: offspring from RD dams weaned to an RD (CRR and IRR), or offspring from HFD dams weaned to either an RD (CHR and IHR) or to an HFD (CHH and IHH). IHH male and female rats had increased large artery stiffness, a suggestion of fatty streaks in the aorta, and persistent decreased elastin and increased collagen in the aorta and carotid arteries. Post-weaning HFD intake increased blood lipids regardless of IUGR status. IUGR increased HFD-induced mortality. We speculate that HFD-induced risk of CVD and mortality is potentiated by developmental programming of the ECM.


Subject(s)
Atherosclerosis , Cardiovascular Diseases , Female , Male , Rats , Animals , Humans , Fetal Growth Retardation/etiology , Diet, High-Fat/adverse effects , Vascular Remodeling , Uterine Artery , Atherosclerosis/etiology
2.
Am J Physiol Heart Circ Physiol ; 321(2): H382-H389, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34142888

ABSTRACT

Pulmonary hypertension (PH) is associated with structural remodeling of pulmonary arteries (PAs) because of excessive proliferation of fibroblasts, endothelial cells, and smooth muscle cells (SMCs). The peptide hormone angiotensin II (ANG II) contributes to pulmonary vascular remodeling, in part, through its ability to trigger extracellular signal-regulated kinase (ERK1/2) activation. Here, we demonstrate that the ERK1/2 phosphatase, dual-specificity phosphatase 5 (DUSP5), functions as a negative regulator of ANG II-mediated SMC proliferation and PH. In contrast to wild-type controls, Dusp5 null mice infused with ANG II developed PH and right ventricular (RV) hypertrophy. PH in Dusp5 null mice was associated with thickening of the medial layer of small PAs, suggesting an in vivo role for DUSP5 as a negative regulator of ANG II-dependent SMC proliferation. Consistent with this, overexpression of DUSP5 blocked ANG II-mediated proliferation of cultured human pulmonary artery SMCs (hPASMCs) derived from patients with idiopathic PH or from failed donor controls. Collectively, the data support a role for DUSP5 as a feedback inhibitor of ANG II-mediated ERK signaling and PASMC proliferation and suggest that disruption of this circuit leads to adverse cardiopulmonary remodeling.NEW & NOTEWORTHY Dual-specificity phosphatases (DUSPs) serve critical roles in the regulation of mitogen-activated protein kinases, but their functions in the cardiovascular system remain poorly defined. Here, we provide evidence that DUSP5, which resides in the nucleus and specifically dephosphorylates extracellular signal-regulated kinase (ERK1/2), blocks pulmonary vascular smooth muscle cell proliferation. In response to angiotensin II infusion, mice lacking DUSP5 develop pulmonary hypertension and right ventricular cardiac hypertrophy. These findings illustrate DUSP5-mediated suppression of ERK signaling in the lungs as a protective mechanism.


Subject(s)
Cell Proliferation/genetics , Dual-Specificity Phosphatases/genetics , Heart Ventricles/metabolism , Hypertension, Pulmonary/genetics , Hypertrophy, Right Ventricular/genetics , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/metabolism , Vascular Remodeling/genetics , Angiotensin II/pharmacology , Animals , Case-Control Studies , Cells, Cultured , Heart Ventricles/drug effects , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Humans , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/chemically induced , Hypertrophy, Right Ventricular/physiopathology , MAP Kinase Signaling System , Mice , Mice, Knockout , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/drug effects , Pulmonary Artery/drug effects , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Vasoconstrictor Agents/pharmacology
3.
Pediatr Res ; 90(5): 998-1008, 2021 11.
Article in English | MEDLINE | ID: mdl-33603215

ABSTRACT

BACKGROUND: The brain of chronically ventilated preterm human infants is vulnerable to collateral damage during invasive mechanical ventilation (IMV). Damage is manifest, in part, by learning and memory impairments, which are hippocampal functions. A molecular regulator of hippocampal development is insulin-like growth factor 1 (IGF1). A gentler ventilation strategy is noninvasive respiratory support (NRS). We tested the hypotheses that NRS leads to greater levels of IGF1 messenger RNA (mRNA) variants and distinct epigenetic profile along the IGF1 gene locus in the hippocampus compared to IMV. METHODS: Preterm lambs were managed by NRS or IMV for 3 or 21 days. Isolated hippocampi were analyzed for IGF1 mRNA levels and splice variants for promoter 1 (P1), P2, and IGF1A and 1B, DNA methylation in P1 region, and histone covalent modifications along the gene locus. RESULTS: NRS had significantly greater levels of IGF1 P1 (predominant transcript), and 1A and 1B mRNA variants compared to IMV at 3 or 21 days. NRS also led to more DNA methylation and greater occupancy of activating mark H3K4 trimethylation (H3K4me3), repressive mark H3K27me3, and elongation mark H3K36me3 compared to IMV. CONCLUSIONS: NRS leads to distinct IGF1 mRNA variant levels and epigenetic profile in the hippocampus compared to IMV. IMPACT: Our study shows that 3 or 21 days of NRS of preterm lambs leads to distinct IGF1 mRNA variant levels and epigenetic profile in the hippocampus compared to IMV. Preterm infant studies suggest that NRS leads to better neurodevelopmental outcomes later in life versus IMV. Also, duration of IMV is directly related to hippocampal damage; however, molecular players remain unknown. NRS, as a gentler mode of respiratory management of preterm neonates, may reduce damage to the immature hippocampus through an epigenetic mechanism.


Subject(s)
Animals, Newborn , Epigenesis, Genetic , Hippocampus/metabolism , Respiration, Artificial/methods , Somatomedins/metabolism , Animals , DNA Methylation , Female , Histones/metabolism , Male , Promoter Regions, Genetic , Sheep , Somatomedins/genetics
4.
Reprod Sci ; 26(9): 1173-1180, 2019 09.
Article in English | MEDLINE | ID: mdl-30453824

ABSTRACT

Uteroplacental insufficiency (UPI) causes intrauterine growth restriction (IUGR) and increases the risk of hypercholesterolemia and cardiovascular disease, which are leading causes of morbidity and mortality worldwide. Little is known about the mechanism through which UPI increases cholesterol. Hepatic Cholesterol 7 alpha-hydroxylase (Cyp7a1) is the rate-limiting and most highly regulated step of cholesterol catabolism to bile acids. Cholesterol 7 alpha-hydroxylase is regulated by transcription factor liver X receptor α (Lxrα) and by microRNA-122. We previously showed that microRNA-122 inhibition of Cyp7a1 translation decreased cholesterol catabolism to bile acids in female IUGR rats at the time of weaning. We hypothesized that UPI would increase cholesterol and microRNA-122 and decrease Cyp7a1 protein and hepatic bile acids in young adult female IUGR rats. To test our hypothesis, we used a rat model of IUGR induced by bilateral uterine artery ligation. Both control and IUGR offspring were exposed to a maternal high-fat diet from before conception through lactation, and all offspring were weaned to a high-fat diet on postnatal day 21. At postnatal day 60, IUGR female rats had increased total and low-density lipoprotein serum cholesterol and hepatic cholesterol, decreased Lxrα and Cyp7a1 protein, and decreased hepatic bile acids. Hepatic microRNA-122 was not changed by UPI. Our findings suggest that UPI decreased cholesterol catabolism to bile acids in young adult female rats through a mechanism independent of microRNA-122.


Subject(s)
Cholesterol/metabolism , Fetal Growth Retardation/metabolism , Liver/metabolism , Placental Insufficiency/metabolism , Animals , Bile Acids and Salts/metabolism , Cholesterol 7-alpha-Hydroxylase/metabolism , Diet, High-Fat , Female , MicroRNAs/metabolism , Pregnancy , Rats , Rats, Sprague-Dawley
5.
Sci Rep ; 7(1): 12879, 2017 10 10.
Article in English | MEDLINE | ID: mdl-29018280

ABSTRACT

Adipose tissue inflammation is a central pathological element that regulates obesity-mediated insulin resistance and type II diabetes. Evidence demonstrates that extracellular signal-regulated kinase (ERK 1/2) activation (i.e. phosphorylation) links tumor necrosis factor α (TNFα) to pro-inflammatory gene expression in the nucleus. Dual specificity phosphatases (DUSPs) inactivate ERK 1/2 through dephosphorylation and can thus inhibit inflammatory gene expression. We report that DUSP5, an ERK1/2 phosphatase, was induced in epididymal white adipose tissue (WAT) in response to diet-induced obesity. Moreover, DUSP5 mRNA expression increased during obesity development concomitant to increases in TNFα expression. Consistent with in vivo findings, DUSP5 mRNA expression increased in adipocytes in response to TNFα, parallel with ERK1/2 dephosphorylation. Genetic loss of DUSP5 exacerbated TNFα-mediated ERK 1/2 signaling in 3T3-L1 adipocytes and in adipose tissue of mice. Furthermore, inhibition of ERK 1/2 and c-Jun N terminal kinase (JNK) signaling attenuated TNFα-induced DUSP5 expression. These data suggest that DUSP5 functions in the feedback inhibition of ERK1/2 signaling in response to TNFα, which resulted in increased inflammatory gene expression. Thus, DUSP5 potentially acts as an endogenous regulator of adipose tissue inflammation; although its role in obesity-mediated inflammation and insulin signaling remains unclear.


Subject(s)
Adipocytes/metabolism , Adipocytes/pathology , Dual-Specificity Phosphatases/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Feedback, Physiological , Gene Expression Regulation , Inflammation/genetics , Tumor Necrosis Factor-alpha/metabolism , 3T3-L1 Cells , Animals , Diet, High-Fat , Dual-Specificity Phosphatases/genetics , Gene Deletion , Male , Mice , Mice, Inbred C57BL , Obesity/genetics , Obesity/pathology , Phosphorylation , RNA, Messenger/genetics , RNA, Messenger/metabolism
6.
Physiol Genomics ; 48(8): 616-25, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27317589

ABSTRACT

Nutrient deprivation suppresses protein synthesis by blocking peptide elongation. Transcriptional upregulation and activation of eukaryotic elongation factor 2 kinase (eEF2K) blocks peptide elongation by phosphorylating eukaryotic elongation factor 2. Previous studies examining placentas from intrauterine growth restricted (IUGR) newborn infants show decreased eEF2K expression and activity despite chronic nutrient deprivation. However, the effect of IUGR on hepatic eEF2K expression in the fetus is unknown. We, therefore, examined the transcriptional regulation of hepatic eEF2K gene expression in a Sprague-Dawley rat model of IUGR. We found decreased hepatic eEF2K mRNA and protein levels in IUGR offspring at birth compared with control, consistent with previous placental observations. Furthermore, the CpG island within the eEF2K promoter demonstrated increased methylation at a critical USF 1/2 transcription factor binding site. In vitro methylation of this binding site caused near complete loss of eEF2K promoter activity, designating this promoter as methylation sensitive. The eEF2K promotor in IUGR offspring also lost the protective histone covalent modifications associated with unmethylated CGIs. In addition, the +1 nucleosome was displaced 3' and RNA polymerase loading was reduced at the IUGR eEF2K promoter. Our findings provide evidence to explain why IUGR-induced chronic nutrient deprivation does not result in the upregulation of eEF2K gene transcription.


Subject(s)
Elongation Factor 2 Kinase/genetics , Fetal Growth Retardation/genetics , Protein Biosynthesis/genetics , Animals , Binding Sites/genetics , CpG Islands/genetics , Epigenesis, Genetic/genetics , Female , Fetus/metabolism , Male , Nucleosomes/genetics , Pregnancy , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Signal Transduction/genetics , Transcription, Genetic/genetics , Up-Regulation/genetics
7.
Physiol Genomics ; 47(12): 634-43, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26487705

ABSTRACT

Intrauterine growth restriction (IUGR) is a common human pregnancy complication. IUGR offspring carry significant postnatal risk for early-onset metabolic syndrome, which is associated with persistent reduction in IGF-1 protein expression. We have previously shown that preadolescent IUGR male mice have decreased hepatic IGF-1 mRNA and circulating IGF-1 protein at postnatal day 21, the age when growth hormone (GH) normally upregulates hepatic IGF-1 expression. Here we studied nucleosome occupancy and CpG methylation at a putative growth hormone-responsive element in intron 2 (in2GHRE) of the hepatic IGF-1 gene in normal, sham-operated, and IUGR mice. Nucleosome occupancy and CpG methylation were determined in embryonic stem cells (ESCs) and in liver at postnatal days 14, 21, and 42. For CpG methylation, additional time points out to 2 yr were analyzed. We confirmed the putative mouse in2GHRE was GH-responsive, and in normal mice, a single nucleosome was displaced from the hepatic in2GHRE by postnatal day 21, which exposed two STAT5b DNA binding sites. Nucleosome displacement correlated with developmentally programmed CpG demethylation. Finally, IUGR significantly altered the nucleosome-depleted region (NDR) at the in2GHRE of IGF-1 on postnatal day 21, with either complete absence of the NDR or with a shifted NDR exposing only one of two STAT5b DNA binding sites. An NDR shift was also seen in offspring of sham-operated mothers. We conclude that prenatal insult such as IUGR or anesthesia/surgery could perturb the proper formation of a well-positioned NDR at the mouse hepatic IGF-1 in2GHRE necessary for transitioning to an open chromatin state.


Subject(s)
DNA Methylation/genetics , Fetal Growth Retardation/genetics , Insulin-Like Growth Factor I/genetics , Nucleosomes/metabolism , Animals , Female , Human Growth Hormone/genetics , Humans , Mice , Pregnancy
8.
Am J Physiol Regul Integr Comp Physiol ; 309(2): R119-27, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25972460

ABSTRACT

Intrauterine growth restriction (IUGR) increases the risk for neurodevelopment delay and neuroendocrine reprogramming in both humans and rats. Neuroendocrine reprogramming involves the glucocorticoid receptor (GR) gene that is epigenetically regulated in the hippocampus. Using a well-characterized rodent model, we have previously shown that IUGR increases GR exon 1.7 mRNA variant and total GR expressions in male rat pup hippocampus. Epigenetic regulation of GR transcription may involve chromatin remodeling of the GR gene. A key chromatin remodeler is Brahma-related gene-1(Brg1), a member of the ATP-dependent SWItch/Sucrose NonFermentable (SWI/SNF) chromatin remodeling complex. Brg1 regulates gene expression by affecting nucleosome repositioning and recruiting transcriptional components to target promoters. We hypothesized that IUGR would increase hippocampal Brg1 expression and binding to GR exon 1.7 promoter, as well as alter nucleosome positioning over GR promoters in newborn male pups. Further, we hypothesized that IUGR would lead to accumulation of specificity protein 1 (Sp1) and RNA pol II at GR exon 1.7 promoter. Indeed, we found that IUGR increased Brg1 expression and binding to GR exon 1.7 promoter. We also found that increased Brg1 binding to GR exon 1.7 promoter was associated with accumulation of Sp1 and RNA pol II carboxy terminal domain pSer-5 (a marker of active transcription). Furthermore, the transcription start site of GR exon 1.7 was located within a nucleosome-depleted region. We speculate that changes in hippocampal Brg1 expression mediate GR expression and subsequently trigger neuroendocrine reprogramming in male IUGR rats.


Subject(s)
Chromatin Assembly and Disassembly , DNA Helicases/metabolism , Fetal Growth Retardation/metabolism , Hippocampus/metabolism , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Receptors, Glucocorticoid/metabolism , Transcription Factors/metabolism , Animals , Animals, Newborn , Binding Sites , DNA Helicases/genetics , Disease Models, Animal , Exons , Fetal Growth Retardation/genetics , Fetal Growth Retardation/physiopathology , Gene Expression Regulation, Developmental , Hippocampus/growth & development , Hippocampus/physiopathology , Male , Nuclear Proteins/genetics , Nucleosomes/metabolism , RNA Polymerase II/metabolism , Rats , Receptors, Glucocorticoid/genetics , Sp1 Transcription Factor/metabolism , Transcription Factors/genetics , Transcription Initiation Site , Transcription, Genetic , Up-Regulation
9.
Pediatr Res ; 78(1): 14-23, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25826117

ABSTRACT

BACKGROUND: Intrauterine growth restriction (IUGR) offspring with rapid catch-up growth are at increased risk for early obesity especially in males. Persistent insulin-like growth factor-1 (IGF-1) reduction is an important risk factor. Using a mouse model of maternal hypertension-induced IUGR, we examined IGF-1 levels, promoter DNA methylation, and histone H3 covalent modifications at birth (D1). We additionally investigated whether prenatal perturbations could reset at preadolescence (D21). METHODS: IUGR was induced via maternal thromboxane A2-analog infusion in mice. RESULTS: IUGR uniformly decreased D1 IGF-1 mRNA and protein levels with reduced promoter 1 (P1) transcription and increased P1 DNA methylation. IUGR males also had increased H3K4ac at exon 5 and 3' distal UTR. At D21, IUGR males continued to have decreased IGF-1 levels, originating from both P1 and P2 with reduced 1A variant. IUGR males also had decreased activation mark of H3K4me3 at P1 compared with sham males. In contrast, D21 IUGR females normalized their IGF-1 levels, in association with an increased activation mark of H3K4me3 at P1 compared with sham females. CONCLUSION: IUGR uniformly affected D1 hepatic IGF-1 epigenetic modifications in both sexes. However, at preadolescence, IUGR males are unable to correct for the prenatal reduction possibly due to a more perturbed IGF-1 chromatin structure.


Subject(s)
Chromatin Assembly and Disassembly , Fetal Growth Retardation/metabolism , Gene Expression Regulation, Developmental , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Animals , Blood Glucose/analysis , Body Weight , Chromatin/metabolism , DNA Methylation , Exons , Female , Fetal Growth Retardation/genetics , Histones/chemistry , Insulin/blood , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/metabolism , Promoter Regions, Genetic , Risk Factors , Sex Factors , Thromboxane A2/chemistry
10.
FASEB J ; 29(4): 1176-84, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25466885

ABSTRACT

Intrauterine growth restriction (IUGR) decreases serum IGF-1 levels. Postnatal IGF-1 expression is transcriptionally regulated by growth hormone (GH) through growth hormone response elements (GHREs). We hypothesized that IUGR disrupts the normal developmental maturation of hepatic IGF-1 intron 2 growth hormone response element (IN2GHRE) histone methylation of key lysines and DNA methylation. We also evaluated a 5' distal weak enhancer (IGF-1 5'-upstream region growth hormone response element; 5URGHRE) as a GHRE specificity control. IUGR was induced through a well-characterized model of bilateral uterine artery ligation of the pregnant rat. Offspring livers were tested at d 0 and 21. Chromatin immunoprecipitation and bisulfite sequencing quantified epigenetic characteristics. We found that distinct age-related developmental patterns of histone and DNA methylation characterize each GHRE. Development increased H3K4 trimethylation (me3) in both GHREs. However, H3K9me3 decreased with age at IN2GHRE and increased with age at 5URGHRE. IUGR altered the developmental pattern of H3K4me3 and K9me3 around the GHREs in a sex-specific manner at d 21. Developmental and IUGR-induced DNA methylation occurred in a GHRE-, CpG site-, and sex-specific manner. We conclude that IUGR disrupts developmental epigenetics around distal GHREs on the rat hepatic IGF-1 gene.


Subject(s)
Epigenesis, Genetic , Fetal Growth Retardation/genetics , Insulin-Like Growth Factor I/genetics , Animals , Animals, Newborn , Binding Sites/genetics , CpG Islands , DNA Methylation , Female , Fetal Growth Retardation/metabolism , Gene Expression Regulation, Developmental , Histones/metabolism , Liver/metabolism , Male , Pregnancy , Rats , Rats, Sprague-Dawley , Response Elements , STAT5 Transcription Factor/metabolism
11.
FASEB J ; 29(3): 807-19, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25395450

ABSTRACT

Intrauterine growth restriction (IUGR) confers heritable alterations in DNA methylation, rendering risk of adult metabolic syndrome (MetS). Because CpG methylation is coupled to intake of essential nutrients along the one-carbon pathway, we reasoned that essential nutrient supplementation (ENS) may abrogate IUGR-conferred multigenerational MetS. Pregnant Sprague-Dawley rats underwent bilateral uterine artery ligation causing IUGR in F1. Among the F2 generation, IUGR lineage rats were underweight at birth (6.7 vs. 8.0 g, P < 0.0001) and obese by adulthood (p160: 613 vs. 510 g; P < 0.0001). Dual energy X-ray absorptiometry studies revealed increased central fat mass (Δ+40 g), accompanied by dyslipidemic (>30% elevated, P < 0.05) serum triglycerides (139 mg/dl), very-LDLs (27.8 mg/dl), and fatty acids (632 µM). Hyperglycemic-euglycemic clamp studies and glucose tolerance testing revealed insulin resistance. Conversely, IUGR lineage ENS-fed rats did not manifest MetS, with significantly lower body weight (p160: 410 g), >5-fold less central fat mass, normal hepatic glucose efflux, and >70% reduced circulating triglycerides and very-LDLs compared with IUGR control-fed F2 offspring (P < 0.01). Moreover, increased methylation of the IGF-1 P2 transcriptional start site among IUGR lineage F2 offspring was reversed in ENS (P < 0.04). This is an initial demonstration that supplementation along the one-carbon pathway abrogates adult morbidity and associated epigenomic modifications of IGF-1 in a rodent model of multigenerational MetS.


Subject(s)
DNA Methylation , Dietary Supplements , Fetal Growth Retardation/physiopathology , Metabolic Syndrome/prevention & control , Prenatal Exposure Delayed Effects/prevention & control , Absorptiometry, Photon , Animals , Blood Glucose/metabolism , Female , Glucose Tolerance Test , Insulin-Like Growth Factor I/genetics , Metabolic Syndrome/etiology , Polymerase Chain Reaction , Pregnancy , Promoter Regions, Genetic/genetics , Rats , Rats, Sprague-Dawley
12.
Pediatr Res ; 76(5): 432-40, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25119340

ABSTRACT

BACKGROUND: Intrauterine growth restriction (IUGR) increases the risk of adult-onset hypercholesterolemia. High-fat diet (HFD) consumption potentiates IUGR-induced increased cholesterol. Cholesterol is converted to bile acids by Cyp7a1 in preparation for excretion. We hypothesized that IUGR rats fed a HFD will have increased cholesterol, decreased Cyp7a1 protein levels, and decreased bile acids compared to control rats fed a HFD. METHODS: At day 21, IUGR and control pups were placed on one of three diets: a regular chow or one of two HFDs containing 1% or 2% cholesterol. Cholesterol levels and hepatic Cyp7a1 protein levels were quantified a postnatal week 28. RESULTS: Both HFDs increased serum cholesterol levels in control rats, and HFD fed IUGR rats had further increased serum cholesterol up to 35-fold. Both HFDs increased hepatic cholesterol levels, and IUGR further increased hepatic cholesterol levels up to fivefold. IUGR decreased hepatic Cyp7a1 protein up to 75%, and hepatic bile acids up to 54%. CONCLUSION: IUGR increased cholesterol and bile acids and decreased Cyp7a1 protein in rats fed a HFD without changing food intake. These findings suggest that IUGR increases the vulnerability of HFD fed rats to hypercholesterolemia via decreased cholesterol conversion to bile acids.


Subject(s)
Cholesterol/blood , Diet, High-Fat , Fetal Growth Retardation , Hypercholesterolemia/etiology , Prenatal Exposure Delayed Effects , Animals , Bile Acids and Salts/metabolism , Biomarkers/blood , Cholesterol 7-alpha-Hydroxylase/metabolism , Disease Models, Animal , Eating , Fatty Acids/blood , Female , Hypercholesterolemia/blood , Hypercholesterolemia/enzymology , Liver/enzymology , Male , Pregnancy , Rats, Sprague-Dawley , Time Factors , Up-Regulation , Weight Gain
13.
Syst Biol Reprod Med ; 59(4): 184-90, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23631676

ABSTRACT

Fetal growth restriction (FGR) is associated with impaired neurodevelopmental outcomes in affected newborns. The pathogenesis of FGR-associated neurodevelopmental impairment implicates abnormal hippocampal function. The steroid hormone estrogen and its receptor, estrogen receptor alpha (ERα), are involved in the normal programming of hippocampal development and structure. However, the impact of FGR on hippocampal estrogen and hippocampal ERα is not well characterized. We hypothesized that FGR will reduce hippocampal and serum levels of 17-beta estradiol and its receptor, ERα, in the newborn rat hippocampus. We further hypothesize that FGR will reduce hippocampal ERα levels in a region-specific manner. To test our hypotheses, we used the well characterized rat model of FGR induced by uteroplacental-insufficiency in the pregnant Sprague-Dawley rat. Hippocampi and serum were obtained from FGR and control day 0 rat pups and examined for hippocampal 17-beta estradiol, serum 17-beta estradiol, and ERα mRNA and protein levels. Immunohistochemistry was performed to examine region-specific ERα staining. FGR decreased hippocampal 17-beta estradiol levels in the hippocampi of male newborn rats but not females. Serum 17-beta estradiol levels were not affected by FGR in either gender. FGR decreased hippocampal ERα mRNA levels in males but not females. Hippocampal ERα protein levels by Western blotting were not affected by FGR. However, FGR decreased apparent ERα staining in the cornu ammonis (CA)1, CA3, and dentate gyrus regions in the hippocampi of male newborn rats but not females. We conclude that FGR affects the programming of hippocampal estrogen and hippocampal ERα levels in the newborn rat in a gender-specific manner.


Subject(s)
Estradiol/blood , Estrogen Receptor alpha/metabolism , Fetal Growth Retardation/physiopathology , Hippocampus/metabolism , Animals , Animals, Newborn , Female , Hippocampus/embryology , Immunohistochemistry , Male , Pregnancy , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
14.
Pediatr Res ; 73(5): 612-620, 2013 May.
Article in English | MEDLINE | ID: mdl-23419538

ABSTRACT

BACKGROUND: We showed that intrauterine growth restriction (IUGR) increases distal airspace wall thickness at birth (postnatal age 0; P0) in rat pups (saccular stage of lung development). However, that report did not assess whether the saccular phenotype persisted postnatally or occurred in males or females, nor did the report identify a potential molecular pathway for the saccular phenotype at P0. We hypothesized that IUGR persistently delays alveolar formation and disrupts retinoic acid receptor (RAR) mRNA and protein levels in the lung of rat pups in a postnatal age- and sex-specific manner. METHODS: IUGR was induced in pregnant rats by bilateral uterine artery ligation. Alveolar formation and expression of RARα, -ß, and -γ were quantified at P0, P6 (alveolar stage), and P21 (postalveolarization). RESULTS: IUGR increased distal airspace wall thickness in female pups at P0 only. IUGR did not affect male pups at any age. IUGR transiently increased lung RAR-ß protein abundance, which inhibits alveolar formation, at P0 in female pups. Serum retinol concentration was normal at all ages. CONCLUSION: IUGR alone is not sufficient to persistently delay postnatal alveolar formation or disrupt expression of RARs. We speculate that for IUGR to delay alveolar formation postnatally, a second insult is necessary.


Subject(s)
Fetal Growth Retardation , Lung/metabolism , Pulmonary Alveoli/embryology , Receptors, Retinoic Acid/metabolism , Animals , Female , Lung/embryology , Pregnancy , Rats
15.
J Nutr Metab ; 2012: 930364, 2012.
Article in English | MEDLINE | ID: mdl-22548154

ABSTRACT

In utero environmental adaptation may predispose to lifelong morbidity. Organisms fine-tune gene expression to achieve environmental adaptation by epigenetic alterations of histone markers of gene accessibility. One example of epigenetics is how uteroplacental insufficiency-induced intrauterine growth restriction (IUGR), which predisposes to adult onset insulin resistance, decreases postnatal IGF-1 mRNA variants and the gene elongation mark histone 3 trimethylation of lysine 36 of the IGF-1 gene (H3Me3K36). Limitations in the study of epigenetics exist due to lack of a primary transgenic epigenetic model. Therefore we examined the epigenetic profile of insulin-like growth factor 1 (IGF-1) in a well-characterized rat model of maternal hyperglycemia to determine if the epigenetic profile of IGF-1 is conserved in disparate models of in utero adaptation. We hypothesized that maternal hyperglycemia would increase IGF-1 mRNA variants and H3Me3K36. However maternal hyperglycemia decreased hepatic IGF-1 mRNA variants and H3Me3K36. This finding is intriguing given that despite different prenatal insults and growth, both maternal hyperglycemia and IUGR predispose to adult onset insulin resistance. We speculate that H3Me3K36 of the IGF-1 gene is sensitive to the glucose level of the prenatal environment, with resultant alteration of IGF-1 mRNA expression and ultimately vulnerability to adult onset insulin resistance.

16.
J Neurotrauma ; 29(11): 2075-85, 2012 Jul 20.
Article in English | MEDLINE | ID: mdl-22413999

ABSTRACT

Traumatic brain injury (TBI) is a major cause of acquired cognitive disability in childhood. Such disability may be blunted by enhancing the brain's endogenous neuroprotective response. An important endogenous neuroprotective response is the insulin-like growth factor-1 (IGF-1) mRNA variant, IGF-1B. IGF-1B mRNA, characterized by exon 5 inclusion, encodes the IGF-1 and Eb peptides. IGF-1A mRNA excludes exon 5 and encodes the IGF-1 and Ea peptides. A region in the human IGF-1B homologue acts as an exon-splicing enhancer (ESE) to increase IGF-1B mRNA. It is not known if TBI is associated with increased brain IGF-1B mRNA. Epigenetic modifications may underlie altered gene expression in the brain after TBI. We hypothesized that TBI would increase hippocampal IGF-1B mRNA in 17-day-old rats, associated with DNA methylation and/or histone modifications at the promoter site 1 (P1) or exon 5/ESE region. Hippocampi from rat pups after controlled cortical impact (CCI) were used to measure IGF-1B mRNA, DNA methylation, and histone modifications at the P1, P2, and exon5/ESE regions. In CCI hippocampi, IGF-1B mRNA peaked at post-injury day (PID) 2 (1700±320% sham), but normalized by PID 14. IGF-1A peaked at PID 3 (280±52% sham), and remained elevated at PID 14. Increased IGF-1B mRNA was associated with increased methylation at P1, and increased histone modifications associated with gene activation at P2 and exon5/ESE, together with differential methylation in the exon 5/ESE regions. We report for the first time that hippocampal IGF-1B mRNA increased after developmental TBI. We speculate that epigenetic modifications at the P2 and exon 5/ESE regions are important in the regulation of IGF-1B mRNA expression. The exon 5/ESE region may present a means for future therapies to target IGF-1B transcription after TBI.


Subject(s)
Brain Injuries/genetics , Epigenesis, Genetic/genetics , Hippocampus/metabolism , Insulin-Like Growth Factor I/genetics , Promoter Regions, Genetic , Animals , Brain Injuries/metabolism , Chromatin Immunoprecipitation , Disease Models, Animal , Exons/genetics , Insulin-Like Growth Factor I/metabolism , Male , RNA, Messenger/analysis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
17.
Pediatr Res ; 72(1): 2-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22367251

ABSTRACT

INTRODUCTION: Uteroplacental insufficiency (UPI) produces significant neurodevelopmental deficits affecting the hippocampus of intrauterine growth-restricted (IUGR) offspring. IUGR males have worse deficits as compared with IUGR females. The exact mechanisms underlying these deficits are unclear. Alterations in hippocampal cellular composition along with altered expression of neural stem cell (NSC) differentiation molecules may underlie these deficits. We hypothesized that IUGR hippocampi would be endowed with altered neuronal, astrocytic, and immature oligodendrocytic proportions at birth, with males showing greater cellular deficits. We further hypothesized that UPI would perturb rat hippocampal expression of ErbB receptors (ErbB-Rs) and neuregulin 1 (NRG1) at birth and at weaning to account for the short- and long-term IUGR neurological sequelae. METHODS: A well-established rat model of bilateral uterine artery ligation at embryonic day 19.5 was used to induce IUGR. RESULTS: As compared with gender-matched controls, IUGR offspring have altered hippocampal neuronal, astrocytic, and immature oligodendrocytic composition in a subregion- and gender-specific manner at birth. In addition, IUGR hippocampi have altered receptor type- and gender-specific ErbB-R expression at birth and at weaning. DISCUSSION: These cellular and molecular alterations may account for the neurodevelopmental complications of IUGR and for the male susceptibility to worse neurologic outcomes.


Subject(s)
Fetal Growth Retardation/physiopathology , Hippocampus/physiopathology , Placental Insufficiency/physiopathology , Receptor, ErbB-2/metabolism , Animals , Astrocytes/metabolism , Cell Differentiation/physiology , Female , Fetal Growth Retardation/etiology , Hippocampus/cytology , Ligation , Male , Microscopy, Fluorescence , Neural Stem Cells/metabolism , Neuregulin-1/metabolism , Oligodendroglia/metabolism , Pregnancy , Rats , Sex Factors , Uterine Artery/surgery
18.
Physiol Genomics ; 43(20): 1160-9, 2011 Oct 20.
Article in English | MEDLINE | ID: mdl-21828247

ABSTRACT

Intrauterine growth retardation (IUGR) predisposes humans toward hippocampal morbidities, such as impaired learning and memory. Hippocampal dual specificity phosphatase 5 (DUSP5) may be involved in these morbidities because DUSP5 regulates extracellular signal-regulated kinase phosphorylation (Erk). In the rat, IUGR causes postnatal changes in hippocampal gene expression and epigenetic characteristics. However, the impact of IUGR upon hippocampal DUSP5 expression and epigenetic characteristics is not known. We therefore hypothesized that IUGR affects hippocampal 1) DUSP5 expression, DNA CpG methylation, and histone code, and 2) erk1/2 phosphorylation in a well-characterized rat model of IUGR. We found that IUGR significantly decreased DUSP5 expression in the day of life (DOL) 0 and 21 male rat, while decreasing only DUSP5 protein levels in the DOL21 female rat. Fluorescent in situ hybridization and immunohistochemistry analyses localized the changes in DUSP5 mRNA and protein, many of which occurred in the dentate gyrus. IUGR also caused sex-specific differences in DNA CpG methylation and histone code in two sites of the hippocampal DUSP5 gene, a 5'-flanking specificity protein-1 (SP1) site and exon 2. Finally, when IUGR decreased DUSP5 protein levels, Erk phosphorylation increased. We conclude that IUGR affects hippocampal DUSP5 expression and epigenetic characteristics in a sex-specific manner.


Subject(s)
Dual-Specificity Phosphatases/genetics , Epigenesis, Genetic , Fetal Growth Retardation/enzymology , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Hippocampus/enzymology , Animals , CpG Islands/genetics , DNA Methylation/genetics , Dual-Specificity Phosphatases/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Fetal Growth Retardation/pathology , Hippocampus/pathology , Histone Code , Immunohistochemistry , In Situ Hybridization, Fluorescence , Male , Phosphorylation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats
19.
Pediatr Res ; 70(2): 123-9, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21505375

ABSTRACT

Intrauterine growth restricted (IUGR) infants have increased susceptibility to infection associated with higher risk of illness and death. Dual specificity phosphatase 1 (DUSP1), which is transcribed in the thymus, increases in quantity as T cells mature and differentiate into CD4+ cells. Little is known about how IUGR affects DUSP1 levels and T-cell subpopulations over time. We hypothesized that IUGR would decrease cell count, CD4+ and CD8+ subpopulations of T lymphocytes, and DUSP1 levels in IUGR rat thymus and spleen. Bilateral uterine artery ligation produced IUGR rats. Thymus and spleen were harvested at P0 and P21. Flow cytometry was used to compare CD4+ and CD8+ lymphocyte populations. Real-time RT-PCR and Western blotting were used to determine DUSP1 quantity. IUGR significantly decreased total cell count in P0 and P21 IUGR male and female thymus. IUGR significantly increased CD4+ cells in IUGR P0 males and females, significantly decreased CD4+ cells in P21 female thymus, and significantly altered DUSP1 levels in the IUGR female thymus at P0 and P21, although it is not yet known whether the change in DUSP1 levels is due to a change in the level per cell or to a change in cellular composition of the thymus.


Subject(s)
Cell Differentiation/immunology , Dual Specificity Phosphatase 1/metabolism , Fetal Growth Retardation/enzymology , Fetal Growth Retardation/immunology , T-Lymphocytes/immunology , Thymus Gland/metabolism , Analysis of Variance , Animals , Animals, Newborn , Blotting, Western , CD4-CD8 Ratio , Cell Count , DNA Primers/genetics , Female , Flow Cytometry , Male , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
20.
Physiol Genomics ; 43(9): 499-505, 2011 May 13.
Article in English | MEDLINE | ID: mdl-21363967

ABSTRACT

Complications of intrauterine growth restriction (IUGR) include increased pulmonary morbidities and impaired alveolar development. Normal alveolar development depends upon elastin expression and processing, as well as the formation and deposition of elastic fibers. This is true of the human and rat. In this study, we hypothesized that uteroplacental insufficiency (UPI)-induced IUGR decreases mRNA levels of elastin and genes required for elastin fiber synthesis and assembly, at birth (prealveolarization) and postnatal day 7 (midalveolarization) in the rat. We further hypothesized that this would be accompanied by reduced elastic fiber deposition and increased static compliance at postnatal day 21 (mature lung). We used a well characterized rat model of IUGR to test these hypotheses. IUGR decreases mRNA transcript levels of genes essential for elastic fiber formation, including elastin, at birth and day 7. In the day 21 lung, IUGR decreases elastic fiber deposition and increases static lung compliance. We conclude that IUGR decreases mRNA transcript levels of elastic fiber synthesis genes, before and during alveolarization leading to a reduced elastic fiber density and increased static lung compliance in the mature lung. We speculate that the mechanism by which IUGR predisposes to pulmonary disease may be via decreased lung elastic fiber deposition.


Subject(s)
Elastin/metabolism , Fetal Growth Retardation/metabolism , Lung/growth & development , Lung/metabolism , Animals , Animals, Newborn , Elastic Tissue/metabolism , Elastin/genetics , Female , Fetal Growth Retardation/genetics , Lung Compliance/genetics , Lung Compliance/physiology , Placental Insufficiency/metabolism , Pregnancy , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...