Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Phlebology ; 37(4): 296-302, 2022 May.
Article in English | MEDLINE | ID: mdl-35249404

ABSTRACT

OBJECTIVE: Bioprosthetic venous valves have yet to achieve long-term patency due to issues with calcification following implantation that is influenced by current xenograft fixation methods, most notably glutaraldehyde. The goal of this study was to investigate the effects of glutaraldehyde fixation on the functional properties of venous tissue to establish a benchmark for the evaluation of alternative fixation methods. METHODS: The degree of crosslinking was evaluated by determining shrink temperature and the stability of tissue with pronase and collagenase digestion. RESULTS: Glutaraldehyde fixation of venous tissue was confirmed by a significant difference in the shrink temperature between fresh and glutaraldehyde treated samples. Significant differences in the amount of tissue remaining following digestion were observed for venous versus cardiac tissue. CONCLUSIONS: This study demonstrates the importance of tissue-specific evaluation in the development of alternative xenograft fixation methods to improve outcomes with bioprosthetic venous valves.


Subject(s)
Bioprosthesis , Venous Valves , Benchmarking , Glutaral , Humans , Temperature , Veins
2.
Animals (Basel) ; 11(1)2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33467586

ABSTRACT

The meat quality of different pig breeds is associated with their different muscle tissue physiological processes, which involves a large variety of genes related with muscle fat and energy metabolism. Understanding the differences of biological processes of muscle after slaughter is helpful to reveal the meat quality development of different breeds. Therefore, eight native Large Black pigs (BP), with high fat content in meat, and seven cross-bred commercial pigs (CP), which had a high feed efficiency with high lean meat, were used to investigate the differences in their meat quality and RNA transcriptomes. The average daily gain (ADG) and hot carcass weight (HCW) of CP were higher than BP, but the back-fat thickness of BP was higher than CP (p < 0.05). The CP had higher a* (redness) but lower h (hue angle) than BP (p < 0.05). The metmyoglobin (MMb) percentage of CP was higher (p < 0.05) than BP. The fat content and oxygen consumption of longissimus dorsi (LD) muscles in BP were higher (p < 0.05) than CP. BP had higher monounsaturated fatty acids (MUFA) content, but CP had higher polyunsaturated fatty acids (PUFA) content (p < 0.05). The RNA-seq data highlighted 201 genes differentially expressed between the two groups (corrected false discovery rate (FDR) p < 0.05), with 75 up-regulated and 126 down-regulated genes in BP compared with CP using the fold change (FC). The real-time PCR was used to validate the results of RNA-seq for eight genes, and the genes related to lipid and energy metabolism were highly expressed in BP (p < 0.05). Based on the results, BP had superior intramuscular fat content to CP, while the growth performance of CP was better, and the transcriptomic differences between these two groups of pigs may cause the meat quality and growth performance variance.

3.
Front Physiol ; 12: 798987, 2021.
Article in English | MEDLINE | ID: mdl-35126181

ABSTRACT

At present, the worldwide prevalence of obesity has become alarmingly high with estimates foreshadowing a continued escalation in the future. Furthermore, there is growing evidence attributing an individual's predisposition for developing obesity to maternal health during gestation. Currently, 60% of pregnancies in the US are to either overweight or obese mothers which in turn contributes to the persistent rise in obesity rates. While obesity itself is problematic, it conveys an increased risk for several diseases such as diabetes, inflammatory disorders, cancer and cardiovascular disease (CVD). Additionally, as we are learning more about the mechanisms underlying CVD, much attention has been brought to the role of perivascular adipose tissue (PVAT) in maintaining cardiovascular health. PVAT regulates vascular tone and for a significant number of individuals, obesity elicits PVAT disruption and dysregulation of vascular function. Obesity elicits changes in adipocyte and leukocyte populations within PVAT leading to an inflammatory state which promotes vasoconstriction thereby aiding the onset/progression of CVD. Our current understanding of obesity, PVAT and CVD has only been examined at the individual level without consideration for a maternal programming effect. It is unknown if maternal obesity affects the propensity for PVAT remodeling in the offspring, thereby enhancing the obesity/CVD link, and what role PVAT leukocytes play in this process. This perspective will focus on the maternal contribution of the interplay between obesity, PVAT disruption and CVD and will highlight the leukocyte/PVAT interaction as a novel target to stem the tide of the current obesity epidemic and its secondary health consequences.

4.
Placenta ; 93: 74-82, 2020 04.
Article in English | MEDLINE | ID: mdl-32250742

ABSTRACT

INTRODUCTION: Maternal obesity and poor quality diets are associated with greater risk of obesity in offspring. Maternal diet and obesity influence placental gene expression and nutrient transport, but the impact of diet and obesity on global epigenetic changes in the placenta are poorly understood. We hypothesized that placental DNA methylation patterns are associated with maternal body mass index (BMI) and/or maternal diet composition. METHODS: Using reduced representation bisulfite sequencing (RRBS), we assessed genome scale DNA methylation of ~300,000 CpGs in 150 term placentas from normal weight mothers (n = 72) and overweight/obese mothers (n = 78). Maternal BMI was assessed before week 10 of gestation and maternal diet composition was assessed using 3-day food records at each trimester. RESULTS: In multivariable linear regression models, maternal BMI category (normal weight or overweight/obese), BMI (kg/m2), and maternal saturated fat consumption (g/d) were associated (p < 0.0001) with methylation of 185, 103, and 302 CpGs, respectively. Of the 56 CpGs associated with both maternal BMI category and maternal BMI (p < 0.0001), GO analysis showed biological processes related to SREBP signaling, phospholipid transport, granulocyte differentiation, and RNA pol II transcription to be affected. Maternal saturated fat intake was associated with methylation of 302 CpGs (p < 0.0001). These genes were related to chromatin remodeling, IGF receptor, PI3K, and nitric oxide synthase signaling. DISCUSSION: These data suggest that placental DNA methylation status is associated with both maternal obesity and maternal saturated fat intake, possibly contributing to maternal obesity-associated changes in placental function.


Subject(s)
Body Mass Index , DNA Methylation , Diet , Maternal Nutritional Physiological Phenomena , Placenta/metabolism , Adult , Cohort Studies , CpG Islands/genetics , Diet, Healthy , Female , Gestational Weight Gain/physiology , Humans , Infant, Newborn , Male , Mothers , Obesity/genetics , Obesity/metabolism , Overweight/genetics , Overweight/metabolism , Pregnancy , Pregnancy Complications/genetics , Pregnancy Complications/metabolism , Young Adult
5.
Obesity (Silver Spring) ; 28(3): 624-630, 2020 03.
Article in English | MEDLINE | ID: mdl-32030918

ABSTRACT

OBJECTIVE: This study investigated which antenatal and postnatal factors determine offspring adiposity during the first 2 years of life. METHODS: Participants were mother and child pairs (N = 224). Offspring percent fat mass (%FM) was obtained using quantitative nuclear magnetic resonance at 11 time points between ages 0.5 and 24 months. Independent variables included race, age, gestational weight gain, first-trimester %FM, delivery mode, gestational measures of resting energy expenditure, respiratory exchange ratio, physical activity, serum cytokines and lipids, and dietary intake for the mothers, as well as sex, birth weight and length, breastfeeding duration, and physical activity at age 2 years for the children. Linear mixed models were used to construct the best-fitted models for the entire cohort and for each sex. RESULTS: Maternal %FM (P = 0.006), high-density lipoprotein (HDL) (P < 0.001), and breastfeeding duration (P = 0.023) were positively associated with female offspring adiposity, whereas maternal dietary fiber intake (P = 0.016) had a negative association. Birth weight (P = 0.004), maternal HDL (P = 0.013), and breastfeeding duration (P = 0.015) were all positively associated with male offspring adiposity. CONCLUSIONS: Antenatal and postnatal factors differentially impact male and female offspring adiposity during the first 2 years of life.


Subject(s)
Adiposity/physiology , Obesity, Maternal/complications , Adult , Child, Preschool , Cohort Studies , Female , Humans , Infant , Infant, Newborn , Male , Pregnancy
6.
Pediatr Obes ; 15(4): e12596, 2020 04.
Article in English | MEDLINE | ID: mdl-31856430

ABSTRACT

BACKGROUND: Maternal obesity increases offspring's obesity risk. However, studies have not often considered maternal metabolic and exercise patterns as well as paternal adiposity as potential covariates. OBJECTIVE: To assess the relationship between parental and newborn adiposity. METHODS: Participants were mother-child pairs (n = 209) and mother-father-offspring triads (n = 136). Parental (during gestation) and offspring (2 weeks old) percent fat mass (FM) were obtained using air displacement plethysmography. Maternal race, age, resting energy expenditure (indirect calorimetry), physical activity (accelerometry), gestational weight gain (GWG), gestational age (GA), delivery mode, infant's sex and infant feeding method were incorporated in multiple linear regression analyses. The association between parental FM and offspring insulin-like growth factor 1 (IGF-1) was assessed at age 2 years. RESULTS: Maternal adiposity was positively-associated with male (ß = 0.11, P = .015) and female (ß = 0.13, P = .008) infant FM, whereas paternal adiposity was negatively-associated with male newborn adiposity (ß = -0.09, P = .014). Breastfeeding, female sex, GA and GWG positively associated with newborn adiposity. Vaginal and C-section delivery methods associated with greater adiposity than vaginal induced delivery method. Plasma IGF-1 of 2-year-old boys and girls positively associated with their respective fathers' and mothers' FM. CONCLUSIONS: Maternal and paternal adiposity differentially associate with newborn adiposity. The mechanisms of this finding remain to be determined.


Subject(s)
Adiposity , Body Composition , Parents , Adult , Child, Preschool , Female , Gestational Age , Humans , Infant, Newborn , Insulin-Like Growth Factor I/analysis , Male , Pregnancy , Weight Gain
7.
Sci Rep ; 8(1): 16502, 2018 11 07.
Article in English | MEDLINE | ID: mdl-30405201

ABSTRACT

The contributions of maternal diet and obesity in shaping offspring microbiome remain unclear. Here we employed a mouse model of maternal diet-induced obesity via high-fat diet feeding (HFD, 45% fat calories) for 12 wk prior to conception on offspring gut microbial ecology. Male and female offspring were provided access to control or HFD from weaning until 17 wk of age. Maternal HFD-associated programming was sexually dimorphic, with male offspring from HFD dams showing hyper-responsive weight gain to postnatal HFD. Likewise, microbiome analysis of offspring cecal contents showed differences in α-diversity, ß-diversity and higher Firmicutes in male compared to female mice. Weight gain in offspring was significantly associated with abundance of Lachnospiraceae and Clostridiaceae families and Adlercreutzia, Coprococcus and Lactococcus genera. Sex differences in metagenomic pathways relating to lipid metabolism, bile acid biosynthesis and immune response were also observed. HFD-fed male offspring from HFD dams also showed worse hepatic pathology, increased pro-inflammatory cytokines, altered expression of bile acid regulators (Cyp7a1, Cyp8b1 and Cyp39a1) and serum bile acid concentrations. These findings suggest that maternal HFD alters gut microbiota composition and weight gain of offspring in a sexually dimorphic manner, coincident with fatty liver and a pro-inflammatory state in male offspring.


Subject(s)
Diet, High-Fat/adverse effects , Fatty Liver/etiology , Fatty Liver/metabolism , Gastrointestinal Microbiome , Maternal Exposure/adverse effects , Animals , Bile Acids and Salts/metabolism , Biodiversity , Biomarkers , Fatty Liver/pathology , Female , Lipid Metabolism , Male , Metabolic Networks and Pathways , Metagenome , Metagenomics/methods , Mice , Phylogeny , Sex Characteristics , Weight Gain
8.
Article in English | MEDLINE | ID: mdl-28971605

ABSTRACT

One mechanism by which the female sex may protect against elevated coronary vascular tone is inhibition of Ca2+ entry into arterial smooth muscle cells (ASMCs). In vitro findings confirm that high estrogen concentrations directly inhibit voltage-dependent Cav 1.2 channels in coronary ASMCs. For this study, we hypothesized that the nonacute, in vitro exposure of coronary arteries to a low concentration of 17ß-estradiol (17ßE) reduces the expression of Cav 1.2 channel proteins in coronary ASMCs. Segments of the right coronary artery obtained from sexually mature female pigs were mounted for isometric tension recording. As expected, our results indicate that high concentrations (≥10 µmol/L) of 17ßE acutely attenuated Ca2+ -dependent contractions to depolarizing KCl stimuli. Interestingly, culturing coronary arteries for 24 h in a 10,000-fold lower concentration (1 nmol/L) of 17ßE also attenuated KCl-induced contractions and reduced the contractile response to the Cav 1.2 agonist, FPL64176, by 50%. Western blots revealed that 1 nmol/L 17ßE decreased protein expression of the pore-forming α1C subunit (Cav α) of the Cav 1.2 channel by 35%; this response did not depend on an intact endothelium. The 17ßE-induced loss of Cav α protein in coronary arteries was prevented by the estrogen ERα/ERß antagonist, ICI 182,780, whereas the GPER antagonist, G15, did not prevent it. There was no effect of 1 nmol/L 17ßE on Cav α transcript expression. We conclude that 17ßE reduces Cav 1.2 channel abundance in isolated coronary arteries by a posttranscriptional process. This unrecognized effect of estrogen may confer physiological protection against the development of abnormal Ca2+ -dependent coronary vascular tone.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium/metabolism , Coronary Vessels/cytology , Estradiol/pharmacology , Muscle Contraction/drug effects , Animals , Cells, Cultured , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Female , Gene Expression Regulation/drug effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Swine
9.
Placenta ; 57: 194-203, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28864012

ABSTRACT

INTRODUCTION: Maternal obesity (OB) and excessive gestational weight gain (GWG) are strong independent contributors that augment obesity risk in offspring. However, direct evidence of epigenetic changes associated with maternal habitus remains sparse. METHODS: We utilized Bisulfite Amplicon Sequencing (BSAS) to conduct targeted DNA methylation association analysis of maternal obesity and excessive GWG with DNA methylation of select metabolism-related and imprinted genes. Umbilical cord (UC) tissue from infants born to normal weight and overweight/obese women from the Glowing study were utilized (n = 78). RESULTS: In multivariable linear regression adjusted for relevant confounders, Institute on Medicine (IOM) GWG category and infant sex were significantly associated with UC IGFBP1 methylation, while gestation length was significantly associated with UC PRKAA1 methylation. In addition, infant fat mass (%) at 2 weeks of age was significantly associated with umbilical cord methylation of RAPTOR. While regression tree analysis confirmed findings from multivariable models demonstrating that maternal early pregnancy BMI and IOM GWG category are associated with fetal UC DNA methylation patterns for select metabolic and imprinted genes, in general, effect sizes were quite small and statistical significance was not maintained when accounting for multiple testing. DISCUSSION: Our findings suggest that maternal obesity and excessive GWG are weakly correlated with offspring DNA methylation patterns at birth.


Subject(s)
DNA Methylation , Obesity/metabolism , Pregnancy Complications/metabolism , Umbilical Cord/metabolism , Weight Gain , AMP-Activated Protein Kinases/metabolism , Cohort Studies , Female , Humans , Infant, Newborn , Insulin-Like Growth Factor Binding Protein 1/metabolism , Male , Pregnancy , Proteins/metabolism , Regression Analysis , Regulatory-Associated Protein of mTOR/metabolism
10.
PLoS One ; 12(4): e0175675, 2017.
Article in English | MEDLINE | ID: mdl-28414763

ABSTRACT

OBJECTIVE: Non-alcoholic fatty liver disease (NAFLD) is an important co-morbidity associated with obesity and a precursor to steatohepatitis. However, the contributions of gestational and early life influences on development of NAFLD and NASH remain poorly appreciated. METHODS: Two independent studies were performed to examine whether maternal over-nutrition via exposure to high fat diet (HFD) leads to exacerbated hepatic responses to post-natal HFD and methionine choline deficient (MCD) diets in the offspring. Offspring of both control diet- and HFD-fed dams were weaned onto control and HFD, creating four groups. RESULTS: When compared to their control diet-fed littermates, offspring of HF-dams weaned onto HFD gained greater body weight; had increased relative liver weight and showed hepatic steatosis and inflammation. Similarly, this group revealed significantly greater immune response and pro-fibrogenic gene expression via RNA-seq. In parallel, 7-8 week old offspring were challenged with either control or MCD diets for 3 weeks. Responses to MCD diets were also exacerbated due to maternal HFD as seen by gene expression of classical pro-fibrogenic genes. Quantitative genome-scale DNA methylation analysis of over 1 million CpGs showed persistent epigenetic changes in key genes in tissue development and metabolism (Fgf21, Ppargc1ß) with maternal HFD and in cell adhesion and communication (VWF, Ephb2) in the combination of maternal HFD and offspring MCD diets. Maternal HFD also influenced gut microbiome profiles in offspring leading to a decrease in α-diversity. Linear regression analysis revealed association between serum ALT levels and Coprococcus, Coriobacteriacae, Helicobacterioceae and Allobaculum. CONCLUSION: Our findings indicate that maternal HFD detrimentally alters epigenetic and gut microbiome pathways to favor development of fatty liver disease and its progressive sequelae.


Subject(s)
Diet, High-Fat/adverse effects , Non-alcoholic Fatty Liver Disease/etiology , Prenatal Exposure Delayed Effects/etiology , Animals , Choline/administration & dosage , Choline Deficiency/complications , DNA Methylation , Disease Models, Animal , Epigenesis, Genetic , Female , Gastrointestinal Microbiome/genetics , Gene Expression Profiling , Liver/metabolism , Male , Maternal Nutritional Physiological Phenomena , Methionine/administration & dosage , Methionine/deficiency , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/microbiology , Overnutrition/complications , Pregnancy , Prenatal Exposure Delayed Effects/genetics , Prenatal Exposure Delayed Effects/microbiology
11.
Mol Cell Endocrinol ; 435: 7-19, 2016 11 05.
Article in English | MEDLINE | ID: mdl-27392497

ABSTRACT

The consequences of excessive maternal weight and adiposity at conception for the offspring are now well recognized. Maternal obesity increases the risk of overweight and obesity even in children born with appropriate-for-gestational age (AGA) birth weights. Studies in animal models have employed both caloric excess and manipulation of macronutrients (especially high-fat) to mimic hypercaloric intake present in obesity. Findings from these studies show transmission of susceptibility to obesity, metabolic dysfunction, alterations in glucose homeostasis, hepatic steatosis, skeletal muscle metabolism and neuroendocrine changes in the offspring. This review summarizes the essential literature in this area in both experimental and clinical domains and focuses on the translatable aspects of these experimental studies. Moreover this review highlights emerging mechanisms broadly explaining maternal obesity-associated developmental programming. The roles of early developmental alterations and placental adaptations are also reviewed. Increasing evidence also points to changes in the epigenome and other emerging mechanisms such as alterations in the microbiome that may contribute to persistent changes in the offspring. Finally, we examine potential interventions that have been employed in clinical cohorts.


Subject(s)
Disease Models, Animal , Fetal Development , Obesity/complications , Pregnancy Complications/epidemiology , Animals , Epigenomics , Female , Humans , Maternal Nutritional Physiological Phenomena , Obesity/genetics , Obesity/therapy , Pregnancy , Prenatal Exposure Delayed Effects/epidemiology , Prenatal Exposure Delayed Effects/genetics
12.
Biomed Res Int ; 2016: 2365609, 2016.
Article in English | MEDLINE | ID: mdl-28105413

ABSTRACT

Nonshivering thermogenesis is the process of biological heat production in mammals and is primarily mediated by brown adipose tissue (BAT). Through ubiquitous expression of uncoupling protein 1 (Ucp1) on the mitochondrial inner membrane, BAT displays uncoupling of fuel combustion and ATP production in order to dissipate energy as heat. Because of its crucial role in regulating energy homeostasis, ongoing exploration of BAT has emphasized its therapeutic potential in addressing the global epidemics of obesity and diabetes. The recent appreciation that adult humans possess functional BAT strengthens this prospect. Furthermore, it has been identified that there are both classical brown adipocytes residing in dedicated BAT depots and "beige" adipocytes residing in white adipose tissue depots that can acquire BAT-like characteristics in response to environmental cues. This review aims to provide a brief overview of BAT research and summarize recent findings concerning the physiological, cellular, and developmental characteristics of brown adipocytes. In addition, some key genetic, molecular, and pharmacologic targets of BAT/Beige cells that have been reported to have therapeutic potential to combat obesity will be discussed.


Subject(s)
Adenosine Triphosphate/metabolism , Adipocytes, Brown/metabolism , Adipose Tissue, Brown/metabolism , Obesity/metabolism , Obesity/therapy , Thermogenesis , Uncoupling Protein 1/metabolism , Adipocytes, Brown/pathology , Adipose Tissue, Brown/pathology , Animals , Humans , Obesity/pathology
13.
Placenta ; 36(12): 1342-51, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26515927

ABSTRACT

INTRODUCTION: Syncytialization is a process essential to the genesis and vitality of the decisive maternal-fetal interface, the syncytiotrophoblast. While the role of specific genes important in syncytial fusion is appreciated, an integrated global analysis of syncytialization is absent. METHODS: We leveraged a variety of approaches (RNA-seq, genome-scale DNA methylation and ChIP-seq) to assemble a genome-wide transcriptomic and epigenomic view of syncytialization in BeWo cells. RESULTS: RNA-seq analysis of expression profiles revealed alterations in ∼3000 genes over the 3 day time-course of forskolin, including identification of several previously unrecognized genes to be involved in syncytialization. These genes were enriched for cell differentiation, morphogenesis, blood vessel and placental labyrinth development and steroid hormone response. Genome-scale DNA methylation via reduced representation bisulfite sequencing (RRBS) showed altered methylation of a number of CpGs associated with cell differentiation and commitment. Finally, genome-wide localization of seven key histone marks encompassing permissive (H3K4me3, H3K9ac, H3K27ac), enhancer (H3K4me1), elongation (H3K36me3) and repressive (H3K27me3, H3K9me3) states was performed via ChiP-seq. These analyses clearly revealed that syncytialization was associated with a gain in transcriptionally permissive/active marks (H3K4me3, K9ac, K27ac and K36me3) among genes that are either constitutive or upregulated in syncytialization. DISCUSSION: Overall, these results provide a novel resource to elucidate the underlying epigenetic mechanisms coordinating transcriptional changes associated with syncytialization in BeWo cells.


Subject(s)
Epigenesis, Genetic , Placenta/metabolism , Placentation/genetics , Transcriptome , Trophoblasts/cytology , Cell Fusion , DNA Methylation , Epigenomics , Escherichia coli Proteins , Female , Histones/metabolism , Humans , Pregnancy
14.
Obesity (Silver Spring) ; 23(5): 1047-54, 2015 May.
Article in English | MEDLINE | ID: mdl-25919924

ABSTRACT

OBJECTIVE: To study potential effects of maternal body composition on central nervous system (CNS) development of newborn infants. METHODS: Diffusion tensor imaging (DTI) was used to evaluate brain white matter development in 2-week-old, full-term, appropriate for gestational age (AGA) infants from uncomplicated pregnancies of normal-weight (BMI < 25 at conception) or obese ( BMI = 30 at conception) and otherwise healthy mothers. Tract-based spatial statistics (TBSS) analyses were used for voxel-wise group comparison of fractional anisotropy (FA), a sensitive measure of white matter integrity. DNA methylation analyses of umbilical cord tissue focused on genes known to be important in CNS development were also performed. RESULTS: Newborns from obese women had significantly lower FA values in multiple white matter regions than those born of normal-weight mothers. Global and regional FA values negatively correlated (P < 0.05) with maternal fat mass percentage. Linear regression analysis followed by gene ontology enrichment showed that methylation status of 68 CpG sites representing 57 genes with GO terms related to CNS development was significantly associated with maternal adiposity status. CONCLUSIONS: These results suggest a negative association between maternal adiposity and white matter development in offspring.


Subject(s)
Adiposity , Obesity/complications , White Matter/growth & development , White Matter/pathology , Adult , Anisotropy , Brain/growth & development , Brain/pathology , Child Development/physiology , Diffusion Tensor Imaging/methods , Female , Humans , Infant , Infant, Newborn , Pregnancy , Pregnancy Complications/pathology , Regression Analysis
15.
Plant Foods Hum Nutr ; 70(1): 56-62, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25535004

ABSTRACT

Highbush (cultivated) and lowbush (wild) are the two major blueberry species in the US market. Eight phenolic acids were detected and quantified from these two species by HPLC-MS. Chlorogenic acid was found to be the predominant phenolic acid in both species, with 0.44 mg/g fresh weight in lowbush blueberries and 0.13 mg/g fresh weight in highbush blueberries. Total phenolic content in lowbush blueberries is over three times higher than that of highbush blueberries. The phenolic acid mixtures representing those in the two species were prepared by using authentic standards to assess their contribution to total antioxidant and anti-inflammatory activities of the whole berries. Neither lowbush nor highbush blueberry phenolic acid mixture contributed significantly to the total antioxidant capacity of their relevant whole berries measured by oxygen radical absorbance capacity (ORAC). Both phenolic acid mixtures were able to enter the cell and showed in cell antioxidant activities from the cell based antioxidant protection of erythrocytes (CAP-e) assay. Lowbush blueberry phenolic acid mixture was found to show anti-inflammatory activities by inhibiting the nuclear factor-κB (NF-κB) activation and the production of inflammatory cytokines (TNF-α and IL-6) at the high dose.


Subject(s)
Anti-Inflammatory Agents/chemistry , Antioxidants/chemistry , Chlorogenic Acid/pharmacology , Fruit/chemistry , Hydroxybenzoates/pharmacology , Blueberry Plants/chemistry , Blueberry Plants/classification , Cell Culture Techniques , Chromatography, High Pressure Liquid , Erythrocytes/drug effects , Fruit/classification , Humans , Interleukin-6/metabolism , NF-kappa B/metabolism , Reactive Oxygen Species/chemistry , Tumor Necrosis Factor-alpha/metabolism
16.
Exp Biol Med (Maywood) ; 239(10): 1340-51, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24951473

ABSTRACT

The umbilical cord (UC) matrix is a source of multipotent mesenchymal stem cells (MSCs) that have adipogenic potential and thus can be a model to study adipogenesis. However, existing variability in adipocytic differentiation outcomes may be due to discrepancies in methods utilized for adipogenic differentiation. Additionally, functional characterization of UCMSCs as adipocytes has not been described. We tested the potential of three well-established adipogenic cocktails containing IBMX, dexamethasone, and insulin (MDI) plus indomethacin (MDI-I) or rosiglitazone (MDI-R) to stimulate adipocyte differentiation in UCMSCs. MDI, MDI-I, and MDI-R treatment significantly increased peroxisome proliferator-activated receptor gamma (PPARγ) and CCAAT-enhancer binding protein alpha (C/EBPα) mRNA and induced lipid droplet formation. However, MDI-I had the greatest impact on mRNA expression of PPARγ, C/EBPα, FABP4, GPD1, PLIN1, PLIN2, and ADIPOQ and lipid accumulation, whereas MDI showed the least. Interestingly, there were no treatment group differences in the amount of PPARγ protein. However, MDI-I treated cells had significantly more C/EBPα protein compared to MDI or MDI-R, suggesting that indomethacin-dependent increased C/EBPα may contribute to the adipogenesis-inducing potency of MDI-I. Additionally, bone morphogenetic protein 4 (BMP4) treatment of UCMSCs did not enhance responsiveness to MDI-induced differentiation. Finally to characterize adipocyte function, differentiated UCMSCs were stimulated with insulin and downstream signaling was assessed. Differentiated UCMSCs were responsive to insulin at two weeks but showed decreased sensitivity by five weeks following differentiation, suggesting that long-term differentiation may induce insulin resistance. Together, these data indicate that UCMSCs undergo adipogenesis when differentiated in MDI, MDI-I, and MDI-R, however the presence of indomethacin greatly enhances their adipogenic potential beyond that of rosiglitazone. Furthermore, our results suggest that insulin signaling pathways of differentiated UCMSCs are functionally similar to adipocytes.


Subject(s)
Adipogenesis , Cell Differentiation , Mesenchymal Stem Cells/physiology , Umbilical Cord/cytology , Culture Media/chemistry , Female , Gene Expression Profiling , Genotype , Humans , Phenotype , Pregnancy , RNA, Messenger/analysis , RNA, Messenger/genetics
17.
Pediatr Res ; 76(2): 202-10, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24819376

ABSTRACT

BACKGROUND: Maternal obesity is associated with unfavorable outcomes, which may be reflected in the as yet undiscovered gene expression profiles of the umbilical cord (UC). METHODS: UCs from 12 lean (pregravid BMI < 24.9) and 10 overweight/obese (pregravid BMI ≥ 25) women without gestational diabetes were collected for gene expression analysis using Human Primeview microarrays. Metabolic parameters were assayed in mother's plasma and cord blood. RESULTS: Although offspring birth weight and adiposity (at 2 wk) did not differ between groups, expression of 232 transcripts was affected in UC from overweight/obese compared with those of lean mothers. Gene-set enrichment analysis revealed an upregulation of genes related to metabolism, stimulus and defense response, and inhibitory to insulin signaling in the overweight/obese group. We confirmed that EGR1, periostin, and FOSB mRNA expression was induced in UCs from overweight/obese mothers, while endothelin receptor B, KLF10, PEG3, and EGLN3 expression was decreased. Messenger RNA expression of EGR1, FOSB, MEST, and SOCS1 were positively correlated (P < 0.05) with mother's first-trimester body fat mass (%). CONCLUSION: Our data suggest a positive association between maternal obesity and changes in UC gene expression profiles favoring inflammation and insulin resistance, potentially predisposing infants to develop metabolic dysfunction later on in life.


Subject(s)
Gene Expression Regulation/physiology , Inflammation/physiopathology , Insulin Resistance/physiology , Maternal Nutritional Physiological Phenomena/physiology , Obesity/physiopathology , Umbilical Cord/physiopathology , Adiposity/physiology , Adult , Analysis of Variance , Anthropometry , Blotting, Western , Cell Adhesion Molecules/metabolism , DNA Primers/genetics , Early Growth Response Protein 1/metabolism , Female , Gene Expression Profiling , Human Umbilical Vein Endothelial Cells , Humans , Insulin/blood , Leptin/blood , Microarray Analysis , Proto-Oncogene Proteins c-fos/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Umbilical Cord/metabolism
18.
Am J Physiol Endocrinol Metab ; 305(1): E1-14, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23632636

ABSTRACT

Obesity is associated with low-grade chronic inflammation, which contributes to cellular dysfunction promoting metabolic disease. Obesity during pregnancy leads to a proinflammatory milieu in the placenta; however, the underlying causes for obesity-induced placental inflammation remain unclear. Here, we examine the mechanisms by which saturated fatty acids and inflammatory cytokines induce inflammation in placental trophoblasts. We conducted global transcriptomic profiling in BeWo cells following palmitate and/or TNFα treatment and gene/protein expression analyses of MAPK pathways and characterized downstream transcription factors directly regulating inflammatory cytokines. Microarray analysis revealed increased expression of genes regulating inflammation, stress response, and immediate early response in cytotrophoblasts in response to palmitic acid (PA), TNFα, or a combination of both (PA + TNFα). Both gene ontology and gene set enrichment analysis revealed MAPK and EGR-1 signaling to be upregulated in BeWo cells, which was confirmed via immunoblotting. Importantly, activation of JNK signaling was necessary for increased proinflammatory cytokine (IL-6, TNFα, and IL-8) and EGR1 mRNA. Consistent with the requirement of JNK signaling, ChIP analysis confirmed the recruitment of c-Jun and other MAPK-responsive immediate early factors on the EGR1 promoter. Moreover, recruitment of EGR-1 on cytokine promoters (IL-6, TNFα, and IL-8) and an impaired proinflammatory response following knockdown of EGR-1 suggested it as a central component of the mechanism facilitating inflammatory gene expression. Finally, akin to in vitro findings, term placenta from obese women also had both increased JNK and p38 signaling and greater EGR-1 protein relative to lean women. Our results demonstrate that lipotoxic insults induce inflammation in placental cells via activation of JNK/EGR-1 signaling.


Subject(s)
Early Growth Response Protein 1/immunology , Lipid Metabolism/immunology , Obesity/immunology , Placenta/immunology , Pregnancy Complications/immunology , Activating Transcription Factor 3/immunology , Activating Transcription Factor 3/metabolism , Cell Line , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Female , Humans , Infant, Newborn , Interleukin-6/genetics , Interleukin-6/immunology , Interleukin-6/metabolism , Interleukin-8/genetics , Interleukin-8/immunology , Interleukin-8/metabolism , Lipid Metabolism/genetics , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/immunology , Male , Palmitates/pharmacology , Placenta/cytology , Pregnancy , Serum Response Factor/immunology , Serum Response Factor/metabolism , Transcriptome/drug effects , Transcriptome/immunology , Trophoblasts/cytology , Trophoblasts/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
19.
Pharmacol Res ; 70(1): 126-38, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23376354

ABSTRACT

Ion channels are multimeric, transmembrane proteins that selectively mediate ion flux across the plasma membrane in a variety of cells including vascular smooth muscle cells (VSMCs). The dynamic interplay of Ca(2+) and K(+) channels on the plasma membrane of VSMCs plays a pivotal role in modulating the vascular tone of small arteries and arterioles. The abnormally-elevated arterial tone observed in hypertension thus points to an aberrant expression and function of Ca(2+) and K(+) channels in the VSMCs. In this short review, we focus on the three well-studied ion channels in VSMCs, namely the L-type Ca(2+) (CaV1.2) channels, the voltage-gated K(+) (KV) channels, and the large-conductance Ca(2+)-activated K(+) (BK) channels. First, we provide a brief overview on the physiological role of vascular CaV1.2, KV and BK channels in regulating arterial tone. Second, we discuss the current understanding of the expression changes and regulation of CaV1.2, KV and BK channels in the vasculature during hypertension. Third, based on available proof-of-concept studies, we describe the potential therapeutic approaches targeting these vascular ion channels in order to restore blood pressure to normotensive levels.


Subject(s)
Antihypertensive Agents/pharmacology , Calcium Channels/metabolism , Hypertension/drug therapy , Hypertension/metabolism , Muscle, Smooth, Vascular/drug effects , Potassium Channels/metabolism , Animals , Calcium Channels/genetics , Humans , Ion Channel Gating/drug effects , Molecular Targeted Therapy , Muscle, Smooth, Vascular/metabolism , Potassium Channels/genetics , Protein Subunits
20.
Hypertension ; 61(1): 137-42, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23129698

ABSTRACT

Voltage-gated L-type Ca(2+) (Ca(v)1.2) channels in vascular smooth muscle cells are a predominant Ca(2+) influx pathway that mediates arterial tone. Channel biogenesis is accomplished when the pore-forming α(1C) subunit coassembles with regulatory Ca(v)ß subunits intracellularly, and the multiprotein Ca(v)1.2 channel complex translocates to the plasma membrane to form functional Ca(2+) channels. We hypothesized that the main Ca(v)ß isoform in vascular smooth muscle cells, Ca(v)ß3, is required for the upregulation of arterial Ca(v)1.2 channels during the development of hypertension, an event associated with abnormal Ca(2+)-dependent tone. Ca(v)1.2 channel expression and function were compared between second-order mesenteric arteries of C57BL/6 wild-type (WT) and Ca(v)ß3(-/-) mice infused with saline (control) or angiotensin II (Ang II) for 2 weeks to induce hypertension. The mesenteric arteries of Ang II-infused WT mice showed increased Ca(v)1.2 channel expression and accentuated Ca(2+)-mediated contractions compared with saline-infused WT mice. In contrast, Ca(v)1.2 channels failed to upregulate in mesenteric arteries of Ang II-infused Ca(v)ß3(-/-) mice, and Ca(2+)-dependent reactivity was normal in these arteries. Basal systolic blood pressure was not significantly different between WT and Ca(v)ß3(-/-) mice (98 ± 2 and 102 ± 3 mm Hg, respectively), but the Ca(v)ß3(-/-) mice showed a blunted pressor response to Ang II infusion. Two weeks after the start of Ang II administration, the systolic blood pressure of Ca(v)ß3(-/-) mice averaged 149 ± 4 mm Hg compared with 180 ± 5 mm Hg in WT mice. Thus, the Ca(v)ß3 subunit is a critical regulatory protein required to upregulate arterial Ca(v)1.2 channels and fully develop Ang II-dependent hypertension in C57BL/6 mice.


Subject(s)
Angiotensin II , Calcium Channels, L-Type/metabolism , Hypertension/metabolism , Mesenteric Arteries/metabolism , Up-Regulation/genetics , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Calcium/metabolism , Calcium Channels, L-Type/genetics , Hypertension/chemically induced , Hypertension/genetics , Mice , Mice, Inbred C57BL , Muscle Contraction/physiology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...