Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 23
Filtrer
1.
Reprod Toxicol ; 128: 108657, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-39002939

RÉSUMÉ

Air pollution (AP) is detrimental to pregnancies including increasing risk factors of gestational diabetes mellitus. We hypothesized that exposure to AP causes cardiovascular and metabolic disruption thereby altering placental gene expression, which in turn affects the placental phenotype and thereby embryonic/fetal development. To test this hypothesis, we investigated the impact of intra-nasal instilled AP upon gestational day 16-19 maternal mouse cardiovascular and metabolic status, placental nutrient transporters, and placental-fetal size and morphology. To further unravel mechanisms, we also examined placental total DNA 5'-hydroxymethylation and bulk RNA sequenced gene expression profiles. AP exposed pregnant mice and fetuses were tachycardic with a reduction in maternal left ventricular fractional shortening and increased uterine artery with decreased umbilical artery systolic peak velocities. In addition, they were hyperglycemic, glucose intolerant and insulin resistant, with changes in placental glucose (Glut3) and fatty acid (Fatp1 & Cd36) transporters, and a spatial disruption of cells expressing Glut10 that imports L-dehydroascorbic acid in protecting against oxidative stress. Placentas revealed inflammatory cellular infiltration with associated cellular edema and necrosis, with dilated vascular spaces and hemorrhage. Placental and fetal body weights decreased in mid-gestation with a reduction in brain cortical thickness emerging in late gestation. Placental total DNA 5'-hydroxymethylation was 2.5-fold higher, with perturbed gene expression profiles involving key metabolic, inflammatory, transcriptional, cellular polarizing and processing genes and pathways. We conclude that gestational exposure to AP incites a maternal inflammatory response resulting in features mimicking maternal gestational diabetes mellitus with altered placental DNA 5'-hydroxymethylation, gene expression, and associated injury.

2.
Front Neurosci ; 18: 1363094, 2024.
Article de Anglais | MEDLINE | ID: mdl-38576870

RÉSUMÉ

Introduction: Serotonin (5-HT) is critical for neurodevelopment and the serotonin transporter (SERT) modulates serotonin levels. Perturbed prenatal and postnatal dietary exposures affect the developing offspring predisposing to neurobehavioral disorders in the adult. We hypothesized that the postnatal brain 5-HT-SERT imbalance associated with gut dysbiosis forms the contributing gut-brain axis dependent mechanism responsible for such ultimate phenotypes. Methods: Employing maternal diet restricted (IUGR, n=8) and high fat+high fructose (HFhf, n=6) dietary modifications, rodent brain serotonin was assessed temporally by ELISA and SERT by quantitative Western blot analysis. Simultaneously, colonic microbiome studies were performed. Results: At early postnatal (P) day 2 no changes in the IUGR, but a ~24% reduction in serotonin (p = 0.00005) in the HFhf group occurred, particularly in the males (p = 0.000007) revealing a male versus female difference (p = 0.006). No such changes in SERT concentrations emerged. At late P21 the IUGR group reared on HFhf (IUGR/HFhf, (n = 4) diet revealed increased serotonin by ~53% in males (p = 0.0001) and 36% in females (p = 0.023). While only females demonstrated a ~40% decrease in serotonin (p = 0.010), the males only trended lower without a significant change within the HFhf group (p = 0.146). SERT on the other hand was no different in HFhf or IUGR/RC, with only the female IUGR/HFhf revealing a 28% decrease (p = 0.036). In colonic microbiome studies, serotonin-producing Bacteriodes increased with decreased Lactobacillus at P2, while the serotonin-producing Streptococcus species increased in IUGR/HFhf at P21. Sex-specific changes emerged in association with brain serotonin or SERT in the case of Alistipase, Anaeroplasma, Blautia, Doria, Lactococcus, Proteus, and Roseburia genera. Discussion: We conclude that an imbalanced 5-HT-SERT axis during postnatal brain development is sex-specific and induced by maternal dietary modifications related to postnatal gut dysbiosis. We speculate that these early changes albeit transient may permanently alter critical neural maturational processes affecting circuitry formation, thereby perturbing the neuropsychiatric equipoise.

3.
Nutrients ; 15(1)2023 Jan 01.
Article de Anglais | MEDLINE | ID: mdl-36615874

RÉSUMÉ

Intra-Uterine Growth Restriction (IUGR) is a risk factor for many adult-onset chronic diseases, such as diabetes and obesity. These diseases are associated with intestinal microbiome perturbations (dysbiosis). The establishment of an intestinal microbiome begins in utero and continues postnatally (PN). Hypercaloric diet-induced dysbiosis is a major driver of childhood obesity. We hypothesized that different postnatal diets superimposed on IUGR will alter the postnatal intestinal microbiome. We compared four experimental rat groups: (1) Ad lib fed regular chow diet pre- and postnatally (CON), (2-3) IUGR induced by maternal caloric restriction prenatally followed postnatally (PN) by either (2) the control diet (IUGR-RC) or (3) High-Fat-high-fructose (IUGR-HFhf) diet, and lastly (4) HFhf ad lib pre- and postnatally (HFhf). Fecal samples were collected from dams and male and female rat offspring at postnatal day 2, 21, and adult day 180 for 16S rRNA gene sequencing. Maternal diet induced IUGR led to dysbiosis of the intestinal microbiome at PN21. Postnatal HFhf diet significantly reduced microbial diversity and worsened dysbiosis reflected by an increased Gammaproteobacteria/Clostridia ratio. Dysbiosis arising from a mismatch between IUGR and a postnatal HFhf diet may contribute to increased risk of the IUGR offspring for subsequent detrimental health problems.


Sujet(s)
Microbiome gastro-intestinal , Obésité pédiatrique , Enfant , Humains , Animaux , Rats , Mâle , Femelle , Dysbiose/complications , ARN ribosomique 16S/génétique , Obésité pédiatrique/complications , Retard de croissance intra-utérin/étiologie , Régime alimentaire , Alimentation riche en graisse/effets indésirables
4.
Appl Biochem Biotechnol ; 194(11): 5474-5505, 2022 Nov.
Article de Anglais | MEDLINE | ID: mdl-35789986

RÉSUMÉ

The enzymatic and bio-enzymatic saccharification of waste broken rice (79.8% of starch) was successfully carried out to produce a reducible sugar solution (160 g/L). Bioethanol of concentration 71.2 g/L (9.0% v/v) was prepared by fermentation of reducing sugar solution, using the commercially available waste brewer's yeast (Saccharomyces cerevisiae). The fermentation process parameters were optimized through response surface methodology (RSM) and hybrid artificial neural network-genetic algorithm (ANN-GA) for optimizing the ethanol concentration. The hybrid ANN-GA model predicted a maximum concentration of 71.9 g/L with a deviation of only 0.97% from the experimental value (71.2 g/L). Four different kinetic models were attempted to fit the experimental time evolution of concentrations with the kinetic parameters estimated by the Levenberg-Marquardt optimization technique. The 4th order Runge-Kutta algorithm was implemented through a C program module. The accuracy of each model was checked against coefficient of determination R2, adjusted R2, the absolute mean deviation (AMD), and root mean square deviation (RMSD). The Andrew-Levenspiel kinetics produced the best performance criteria at two initial substrate concentrations of 160 and 170 g/L. Finally, the FTIR analysis of 781.2 g/L (98.5% v/v) bioethanol (concentrated by two-stage vacuum distillation followed by treatment with 3A molecular sieve) showed a favorable blending possibility with the commercial gasoline (petrol) as a green fuel.


Sujet(s)
Oryza , Cinétique , Essence , Éthanol , Saccharomyces cerevisiae , Amidon , Glucides , Sucres
5.
Sci Rep ; 12(1): 8438, 2022 05 19.
Article de Anglais | MEDLINE | ID: mdl-35589747

RÉSUMÉ

The placenta is a heterogeneous organ whose development involves complex interactions of trophoblasts with decidual, vascular, and immune cells at the fetal-maternal interface. It maintains a critical balance between maternal and fetal homeostasis. Placental dysfunction can lead to adverse pregnancy outcomes including intra-uterine growth restriction, pre-eclampsia, or pre-term birth. Exposure to environmental pollutants contributes to the development of placental abnormalities, with poorly understood molecular underpinning. Here we used a mouse (C57BL/6) model of environmental pollutant exposure by administration of a particulate matter (SRM1649b at 300 µg/day/mouse) suspension intra-nasally beginning 2 months before conception and during gestation, in comparison to saline-exposed controls. Placental transcriptomes, at day 19 of gestation, were determined using bulk RNA-seq from whole placentas of exposed (n = 4) and control (n = 4) animals and scRNAseq of three distinct placental layers, followed by flow cytometry analysis of the placental immune cell landscape. Our results indicate a reduction in vascular placental cells, especially cells responsible for structural integrity, and increase in trophoblast proliferation in animals exposed to particulate matter. Pollution-induced inflammation was also evident, especially in the decidual layer. These data indicate that environmental exposure to air pollutants triggers changes in the placental cellular composition, mediating adverse pregnancy outcomes.


Sujet(s)
Polluants atmosphériques , Maladies du placenta , Polluants atmosphériques/toxicité , Animaux , Caduques , Femelle , Humains , Souris , Souris de lignée C57BL , Matière particulaire/toxicité , Placenta , Grossesse , Trophoblastes
6.
Dev Neurosci ; 43(1): 27-42, 2021.
Article de Anglais | MEDLINE | ID: mdl-33774619

RÉSUMÉ

Perturbed maternal diet and prenatal exposure to air pollution (AP) affect the fetal brain, predisposing to postnatal neurobehavioral disorders. Glucose transporters (GLUTs) are key in fueling neurotransmission; deficiency of the neuronal isoform GLUT3 culminates in autism spectrum disorders. Along with the different neurotransmitters, serotonin (5-HT) and oxytocin (OXT) are critical for the development of neural connectivity. Serotonin transporter (SERT) modulates synaptic 5-HT levels, while the OXT receptor (OXTR) mediates OXT action. We hypothesized that perturbed brain GLUT1/GLUT3 regulated 5-HT-SERT imbalance, which serves as a contributing factor to postnatal neuropsychiatric phenotypes, with OXT/OXTR providing a counterbalance. Employing maternal diet restriction (intrauterine growth restriction [IUGR]), high-fat (HF) dietary modifications, and prenatal exposure to simulated AP, fetal (E19) murine brain 5-HT was assessed by ELISA with SERT and OXTR being localized by immunohistochemistry and measured by quantitative Western blot analysis. IUGR with lower head weights led to a 48% reduction in male and female fetal brain GLUT3 with no change in GLUT1, when compared to age- and sex-matched controls, with no significant change in OXTR. In addition, a ∼50% (p = 0.005) decrease in 5-HT and SERT concentrations was displayed in fetal IUGR brains. In contrast, despite emergence of microcephaly, exposure to a maternal HF diet or AP caused no significant changes. We conclude that in the IUGR during fetal brain development, reduced GLUT3 is associated with an imbalanced 5-HT-SERT axis. We speculate that these early changes may set the stage for altering the 5HT-SERT neural axis with postnatal emergence of associated neurodevelopmental disorders.


Sujet(s)
Polluants atmosphériques , Transporteur de glucose de type 3 , Récepteurs à l'ocytocine , Transporteurs de la sérotonine , Sérotonine , Animaux , Encéphale , Régime alimentaire , Femelle , Transporteurs de glucose par diffusion facilitée , Transporteur de glucose de type 1 , Mâle , Souris , Ocytocine , Grossesse
7.
3 Biotech ; 11(1): 28, 2021 Jan.
Article de Anglais | MEDLINE | ID: mdl-33442526

RÉSUMÉ

Reducible sugar solution has been produced from waste broken rice by a novel saccharification process using a combination of bio-enzyme (bakhar) and commercial enzyme (α-amylase). The reducible sugar solution thus produced is a promising raw material for the production of bioethanol using the fermentation process. Response surface methodology (RSM) and Artificial neural network-genetic algorithm (ANN-GA) have been used separately to optimize the multivariable process parameters for maximum yield of the total reducing sugar (TRS) in saccharification process. The maximum yield (0.704 g/g) of TRS is predicted by the ANN-GA model at a temperature of 93 °C, saccharification time of 250 min, 6.5 pH and 1.25 mL/kg of enzyme dosages, while the RSM predicts the maximum yield of 0.7025 g/g at a little different process conditions. The fresh experimental validation of the said model predictions by ANN-GA and RSM is found to be satisfactory with the relative mean error of 2.4% and 3.8% and coefficients of determination of 0.997 and 0.996.

8.
Bioresour Technol ; 299: 122592, 2020 Mar.
Article de Anglais | MEDLINE | ID: mdl-31869631

RÉSUMÉ

Biological pretreatment of polysaccharidic wastes (PWs) is a cost-effective and environmentally friendly approach to improve the digestibility and utilization of these valuable substrates in dual-stage biohythane production. In order to reduce the prolonged incubation time and loss of carbohydrate during the pretreatment of PWs with Aspergillus fumigatus, a systematic optimization using Taguchi methodology resulted in an unprecedented recovery of soluble carbohydrates (362.84 mg g-1) within 5 days. The disruption and fragmentation of lignocellulosic structures in PWs, and possible saccharification of cellulose and hemicellulose components, increased its digestibility. A dual-stage biohythane production with pretreated PWs showed increased yield (214.13 mL g-1 VSadded), which was 56% higher than the corresponding value with the untreated PWs. This resulted in 47% higher energy recovery as biohythane in pretreated biomass compared to untreated biomass. Optimized fungal pretreatment is, therefore, an effective method to improve the digestibility of PWs and its subsequent conversion to biohythane.


Sujet(s)
Aspergillus fumigatus , Cellulose , Biomasse , Glucides
9.
J Nutr Biochem ; 62: 192-201, 2018 12.
Article de Anglais | MEDLINE | ID: mdl-30308381

RÉSUMÉ

We examined the effect of a high-fat diet (HFD) vs. control diet (CD) upon pregestational and gestational wild-type (wt) and glucose transporter (glut)3 heterozygous (glut3+/-) female mice and observed an increase in pregestational body weights, white adiposity (wt > glut3+/-), circulating cholesterol, and high-density lipoproteins, with glucose intolerance in both genotypes. The HFD-exposed offspring displayed reduced birth weight with catch up to CD-fed in wt vs. an increased birth weight persisting as such at weaning by day 21 in glut3+/- mice. To decipher the mechanism behind this genotype-specific difference in the HFD offspring's phenotype, we first examined placental macronutrient transporters and noted HFD-induced increase in CD36 in wt with no change in other FATPs, sodium-coupled neutral amino acid transporters and system L amino acid transporter in both genotypes. In contrast, while placental Glut1 increased in both the genotypes, only Glut3 increased in the glut3+/- genotype in response to HFD. Hence, we next assessed glut3+/- embryonic (ES) cells under differing stressors of low glucose, hypoxia and inhibition of oxidative phosphorylation. Reduced Glut3-mediated glucose uptake in glut3+/- vs. wt ES cells culminated in deficient growth. We conclude that maternal HFD affects the in utero growth potential of the offspring by altering placental CD36 and Glut1 concentrations. In contrast, a differential effect on placental Glut3 concentrations between glut3+/- and wt genotypes is evident, with an increase occurring in the glut3+/- genotype alone. Deficient Glut3 in ES cells interferes with glucose uptake, cell survival and growth being further exaggerated with low glucose, hypoxia and inhibition of oxidative phosphorylation.


Sujet(s)
Adiposité/physiologie , Alimentation riche en graisse/effets indésirables , Transporteur de glucose de type 3/génétique , Obésité/étiologie , Placenta/métabolisme , Animaux , Animaux nouveau-nés , Blastocyste/métabolisme , Poids , Antigènes CD36/métabolisme , Femelle , Transporteur de glucose de type 1/métabolisme , Transporteur de glucose de type 3/métabolisme , Hétérozygote , Souris de lignée C57BL , Souches mutantes de souris , Nutriments/métabolisme , Taille d'organe , Phosphorylation oxydative , Grossesse
10.
J Neurosci ; 38(44): 9579-9599, 2018 10 31.
Article de Anglais | MEDLINE | ID: mdl-30232223

RÉSUMÉ

We created a neural-specific conditional murine glut3 (Slc2A3) deletion (glut3flox/flox/nestin-Cre+) to examine the effect of a lack of Glut3 on neurodevelopment. Compared with age-matched glut3flox/flox = WT and heterozygotes (glut3flox/+/nestin-Cre+), we found that a >90% reduction in male and female brain Glut3 occurred by postnatal day 15 (PN15) in glut3flox/flox/nestin-Cre+ This genetic manipulation caused a diminution in brain weight and cortical thickness at PN15, a reduced number of dendritic spines, and fewer ultrasonic vocalizations. Patch-clamp recordings of cortical pyramidal neurons revealed increased frequency of bicuculline-induced paroxysmal discharges as well as reduced latency, attesting to a functional synaptic and cortical hyperexcitability. Concomitant stunting with lower glucose concentrations despite increased milk intake shortened the lifespan, failing rescue by a ketogenic diet. This led to creating glut3flox/flox/CaMK2α-Cre+ mice lacking Glut3 in the adult male limbic system. These mice had normal lifespan, displayed reduced IPSCs in cortical pyramidal neurons, less anxiety/fear, and lowered spatial memory and motor abilities but heightened exploratory and social responses. These distinct postnatal and adult phenotypes, based upon whether glut3 gene is globally or restrictively absent, have implications for humans who carry copy number variations and present with neurodevelopmental disorders.SIGNIFICANCE STATEMENT Lack of the key brain-specific glucose transporter 3 gene found in neurons during early postnatal life results in significant stunting, a reduction in dendritic spines found on neuronal processes and brain size, heightened neuronal excitability, along with a shortened lifespan. When occurring in the adult and limited to the limbic system alone, lack of this gene in neurons reduces the fear of spatial exploration and socialization but does not affect the lifespan. These features are distinct heralding differences between postnatal and adult phenotypes based upon whether the same gene is globally or restrictively lacking. These findings have implications for humans who carry copy number variations pertinent to this gene and have been described to present with neurodevelopmental disorders.


Sujet(s)
Encéphale/métabolisme , Comportement d'exploration/physiologie , Délétion de gène , Transporteur de glucose de type 3/déficit , Transporteur de glucose de type 3/génétique , Phénotype , Facteurs âges , Animaux , Animaux nouveau-nés , Encéphale/anatomopathologie , Épines dendritiques/génétique , Épines dendritiques/métabolisme , Épines dendritiques/anatomopathologie , Femelle , Souris , Souris de lignée C57BL , Souris knockout , Grossesse , Isoformes de protéines/déficit , Isoformes de protéines/génétique
11.
PLoS One ; 13(3): e0193583, 2018.
Article de Anglais | MEDLINE | ID: mdl-29590129

RÉSUMÉ

BACKGROUND: Intrauterine growth restriction (IUGR) results from a lack of nutrients transferred to the developing fetus, particularly oxygen and glucose. Increased expression of the cytoprotective mitochondrial peptide, humanin (HN), and the glucose transporter 8, GLUT8, has been reported under conditions of hypoxic stress. However, the presence and cellular localization of HN and GLUT8 in IUGR-related placental pathology remain unexplored. Thus, we undertook this study to investigate placental expression of HN and GLUT8 in IUGR-affected versus normal pregnancies. RESULTS: We found 1) increased HN expression in human IUGR-affected pregnancies on the maternal aspect of the placenta (extravillous trophoblastic (EVT) cytoplasm) compared to control (i.e. appropriate for gestational age) pregnancies, and a concomitant increase in GLUT8 expression in the same compartment, 2) HN and GLUT8 showed a protein-protein interaction by co-immunoprecipitation, 3) elevated HN and GLUT8 levels in vitro under simulated hypoxia in human EVT cells, HTR8/SVneo, and 4) increased HN expression but attenuated GLUT8 expression in vitro under serum deprivation in HTR8/SVneo cells. CONCLUSIONS: There was elevated HN expression with cytoplasmic localization to EVTs on the maternal aspect of the human placenta affected by IUGR, also associated with increased GLUT8 expression. We found that while hypoxia increased both HN and GLUT8, serum deprivation increased HN expression alone. Also, a protein-protein interaction between HN and GLUT8 suggests that their interaction may fulfill a biologic role that requires interdependency. Future investigations delineating molecular interactions between these proteins are required to fully uncover their role in IUGR-affected pregnancies.


Sujet(s)
Retard de croissance intra-utérin/métabolisme , Transporteurs de glucose par diffusion facilitée/métabolisme , Protéines et peptides de signalisation intracellulaire/métabolisme , Placenta/métabolisme , Adulte , Cytoplasme/métabolisme , Femelle , Retard de croissance intra-utérin/génétique , Retard de croissance intra-utérin/anatomopathologie , Transporteurs de glucose par diffusion facilitée/génétique , Humains , Protéines et peptides de signalisation intracellulaire/génétique , Mâle , Placenta/anatomopathologie , Grossesse , Transport des protéines , ARN messager/génétique , ARN messager/métabolisme , Trophoblastes/anatomopathologie , Régulation positive
12.
Reprod Sci ; 25(4): 523-539, 2018 04.
Article de Anglais | MEDLINE | ID: mdl-28693373

RÉSUMÉ

Inherent genetic programming and environmental factors affect fetal growth in utero. Epidemiologic data in growth-altered fetuses, either intrauterine growth restricted (IUGR) or large for gestational age (LGA), demonstrate that these newborns are at increased risk of cardiometabolic disease in adulthood. There is growing evidence that the in utero environment leads to epigenetic modification, contributing to eventual risk of developing heart disease or diabetes. In this study, we used reduced representation bisulfite sequencing to examine genome-wide DNA methylation variation in placental samples from offspring born IUGR, LGA, and appropriate for gestational age (AGA) and to identify differential methylation of genes important for conferring risk of cardiometabolic disease. We found that there were distinct methylation signatures for IUGR, LGA, and AGA groups and identified over 500 differentially methylated genes (DMGs) among these group comparisons. Functional and gene network analyses revealed expected relationships of DMGs to placental physiology and transport, but also identified novel pathways with biologic plausibility and potential clinical importance to cardiometabolic disease. Specific loci for DMGs of interest had methylation patterns that were strongly associated with anthropometric presentations. We further validated altered gene expression of these specific DMGs contributing to vascular and metabolic diseases (SLC36A1, PTPRN2, CASZ1, IL10), thereby establishing transcriptional effects toward assigning functional significance. Our results suggest that the gene expression and methylation state of the human placenta are related and sensitive to the intrauterine environment, as it affects fetal growth patterns. We speculate that these observed changes may affect risk for offspring in developing adult cardiometabolic disease.


Sujet(s)
Poids de naissance/génétique , Méthylation de l'ADN , Développement foetal/génétique , Placenta/métabolisme , Épigenèse génétique , Femelle , Retard de croissance intra-utérin/génétique , Retard de croissance intra-utérin/métabolisme , Expression des gènes , Régulation de l'expression des gènes , Humains , Nouveau-né , Grossesse
13.
Nutr Res ; 36(10): 1055-1067, 2016 10.
Article de Anglais | MEDLINE | ID: mdl-27865347

RÉSUMÉ

Intrauterine growth restriction (IUGR) affects up to 10% of pregnancies and often results in short- and long-term sequelae for offspring. The mechanisms underlying IUGR are poorly understood, but it is known that healthy placentation is essential for nutrient provision to fuel fetal growth, and is regulated by immunologic inputs. We hypothesized that in pregnancy, maternal food restriction (FR) resulting in IUGR would decrease the overall immunotolerant milieu in the placenta, leading to increased cellular stress and death. Our specific objectives were to evaluate (1) key cytokines (eg, IL-10) that regulate maternal-fetal tolerance, (2) cellular processes (autophagy and endoplasmic reticulum [ER] stress) that are immunologically mediated and important for cellular survival and functioning, and (3) the resulting IUGR phenotype and placental histopathology in this animal model. After subjecting pregnant mice to mild and moderate FR from gestational day 10 to 19, we collected placentas and embryos at gestational day 19. We examined RNA sequencing data to identify immunologic pathways affected in IUGR-associated placentas and validated messenger RNA expression changes of genes important in cellular integrity. We also evaluated histopathologic changes in vascular and trophoblastic structures as well as protein expression changes in autophagy, ER stress, and apoptosis in the mouse placentas. Several differentially expressed genes were identified in FR compared with control mice, including a considerable subset that regulates immune tolerance, inflammation, and cellular integrity. In summary, maternal FR decreases the anti-inflammatory effect of IL-10 and suppresses placental autophagic and ER stress responses, despite evidence of dysregulated vascular and trophoblast structures leading to IUGR.


Sujet(s)
Autophagie , Stress du réticulum endoplasmique , Retard de croissance intra-utérin/étiologie , Troubles nutritionnels du foetus/étiologie , Interleukine-10/métabolisme , Placenta , Phénomènes physiologiques nutritionnels prénatals , Animaux , Apoptose , Vaisseaux sanguins , Consommation alimentaire , Ration calorique , Femelle , Retard de croissance intra-utérin/métabolisme , Troubles nutritionnels du foetus/métabolisme , Tolérance immunitaire , Inflammation/étiologie , Inflammation/métabolisme , Souris de lignée C57BL , Mères , Placenta/immunologie , Placenta/métabolisme , Placenta/anatomopathologie , Grossesse , ARN messager/métabolisme , Analyse de séquence d'ARN , Trophoblastes
14.
Endocrinology ; 157(10): 4041-4054, 2016 Oct.
Article de Anglais | MEDLINE | ID: mdl-27494059

RÉSUMÉ

We examined the effect of mild (Mi; ∼25%) and moderate (Mo; ∼50%) maternal calorie restriction (MCR) vs ad libitum-fed controls on placental glucose and leucine transport impacting fetal growth potential. We observed in MiMCR a compensatory increase in transplacental (TP) glucose transport due to increased placental glucose transporter isoform (GLUT)-3 but no change in GLUT1 protein concentrations. This change was paralleled by increased glut3 mRNA and 5-hydroxymethylated cytosines with enhanced recruitment of histone 3 lysine demethylase to the glut3 gene locus. To assess the biologic relevance of placental GLUT1, we also examined glut1 heterozygous null vs wild-type mice and observed no difference in placental GLUT3 and TP or intraplacental glucose and leucine transport. Both MCR states led to a graded decrease in TP and intraplacental leucine transport, with a decline in placental L amino acid transporter isoform 2 (LAT2) concentrations and increased microRNA-149 (targets LAT2) and microRNA-122 (targets GLUT3) expression in MoMCR alone. These changes were accompanied by a step-wise reduction in uterine and umbilical artery Doppler blood flow with decreased fetal left ventricular ejection fraction and fractional shortening. We conclude that MiMCR transactivates placental GLUT3 toward preserving TP glucose transport in the face of reduced leucine transport. This contrasts MoMCR in which a reduction in placental GLUT3 mediated glucose transport with a reciprocal increase in miR-122 expression was encountered. A posttranscriptional reduction in LAT2-mediated leucine transport also occurred with enhanced miR-149 expression. Both MCR states, although not affecting placental GLUT1, resulted in uteroplacental insufficiency and fetal growth restriction with compromised cardiovascular health.


Sujet(s)
Restriction calorique/effets indésirables , Placenta/métabolisme , Insuffisance placentaire/étiologie , Phénomènes physiologiques nutritionnels prénatals , Système y+ de transport d'acides aminés/métabolisme , Animaux , Développement embryonnaire , Femelle , Retard de croissance intra-utérin/étiologie , Retard de croissance intra-utérin/métabolisme , Chaines légères de l'antigène CD98/métabolisme , Glucose/métabolisme , Transporteur de glucose de type 1/métabolisme , Transporteur de glucose de type 3/métabolisme , Leucine/métabolisme , Souris de lignée C57BL , Placenta/anatomopathologie , Circulation placentaire , Insuffisance placentaire/métabolisme , Grossesse
15.
Trop Gastroenterol ; 36(3): 188-91, 2015.
Article de Anglais | MEDLINE | ID: mdl-27522739

RÉSUMÉ

Boerhaave's syndrome is the most sinister cause of esophageal perforation. Clinical presentation is vague. Diagnostic delays are frequent. As condition is rare therefore no consensus exists on management. A wide variety of management options are described in literature, each with its advantages and disadvantages. We present our experience of managing these cases which presented after 24 hr. Of onset of symptoms with emphasis on primary reinforced repair as first line surgical option.


Sujet(s)
Perforation de l'oesophage/thérapie , Maladies du médiastin/thérapie , Adolescent , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Perforation de l'oesophage/imagerie diagnostique , Femelle , Humains , Mâle , Maladies du médiastin/imagerie diagnostique , Tomodensitométrie
16.
J Nutr Biochem ; 25(2): 259-66, 2014 Feb.
Article de Anglais | MEDLINE | ID: mdl-24445052

RÉSUMÉ

Diminished transplacental glucose transport plays an important role in prenatal calorie restriction (CR) induced reduction in fetal growth. Fetal growth restriction (FGR) has an impact in shaping the adult phenotype with transgenerational implications. To understand the mechanisms underlying prenatal CR-induced transplacental glucose transport, we examined the epigenetic regulation of placental glucose transporter (Glut1 and Glut3) expression. We restricted calories by 50% in C57BL6 pregnant mice from gestational days 10 to 19 (CR; n=8) vs. controls (CON; n=8) and observed a 50% diminution in placental Glut3 expression (P<.05) with no effect on Glut1 expression by reverse transcription and quantitative real-time polymerase chain reaction (PCR). CR enhanced DNA methylation of a CpG island situated ~1000 bp upstream from the transcriptional start site of the glut3 gene, with no such effect on the glut1 gene as assessed by methylation-sensitive PCR and bisulfite sequencing. Chromatin immunoprecipitation (ChIP) assays demonstrated enhanced MeCP2 binding to the CpG island of the glut3 gene in response to CR vs. CON (P<.05). Sequential ChIP demonstrated that enhanced MeCP2 binding of the glut3-(m)CpG island enhanced histone deacetylase 2 recruitment (P<.05) but interfered with Sp1 binding (P<.001), although it did not affect Sp3 or Creb/pCreb interaction. We conclude that late-gestation CR enhanced DNA methylation of the placental glut3 gene. This epigenetic change augmented specific nuclear protein-DNA complex formation that was associated with prenatal CR-induced reduction of placental glut3 expression and thereby transplacental glucose transport. This molecular complex provides novel targets for developing therapeutic interventions aimed at reversing FGR.


Sujet(s)
Restriction calorique , Méthylation de l'ADN , Transporteur de glucose de type 3/métabolisme , Methyltransferases/métabolisme , Placenta/métabolisme , Animaux , Séquence nucléotidique , Ilots CpG , Amorces ADN , Femelle , Souris , Grossesse , Réaction de polymérisation en chaine en temps réel
17.
Indian J Exp Biol ; 51(7): 556-66, 2013 Jul.
Article de Anglais | MEDLINE | ID: mdl-23898555

RÉSUMÉ

An efficient conversion of lignocellulose into fermentable sugars is a key step in producing bioethanol in a cost effective and eco-friendly manner. Alternative source like water hyacinth biomass (WHB) (Eichhornia crassipes) may be used as a supplement for the routine feedstocks. The enzyme loading for optimum yield of total reducing sugar was investigated and the enzyme-substrate interaction optimised. The maximal reducing sugar and xylose yield was obtained using cellulase and xylanase loading of 46.12 and 289.98 U/g and 2.26% (w/v) substrate loading. The efficiencies of ethanol production from the WHB hydrolysate are very less and the maximal ethanol yield was 3.4969 g/L when Pichia stiptis was used, followed by 3.4496 and 3.1349 g/L for Candida shehatae and Saccharomyces cerevisiae.


Sujet(s)
Biomasse , Glucides , Cellulases/métabolisme , Eichhornia/métabolisme , Endo-1,4-beta xylanases/métabolisme , Éthanol/métabolisme , Candida/métabolisme , Fermentation , Hydrolyse , Lignine/métabolisme , Pichia/métabolisme , Saccharomyces cerevisiae/métabolisme , Spectroscopie infrarouge à transformée de Fourier , Diffraction des rayons X
18.
Physiol Genomics ; 45(14): 565-76, 2013 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-23695884

RÉSUMÉ

Maternal nutrient restriction causes the development of adult onset chronic diseases in the intrauterine growth restricted (IUGR) fetus. Investigations in mice have shown that either protein or calorie restriction during pregnancy leads to glucose intolerance, increased fat mass, and hypercholesterolemia in adult male offspring. Some of these phenotypes are shown to persist in successive generations. The molecular mechanisms underlying IUGR remain unclear. The placenta is a critical organ for mediating changes in the environment and the development of embryos. To shed light on molecular mechanisms that might affect placental responses to differing environments we examined placentas from mice that had been exposed to different diets. We measured gene expression and whole genome DNA methylation in both male and female placentas of mice exposed to either caloric restriction or ad libitum diets. We observed several differentially expressed pathways associated with IUGR phenotypes and, most importantly, a significant decrease in the overall methylation between these groups as well as sex-specific effects that are more pronounced in males. In addition, a set of significantly differentially methylated genes that are enriched for known imprinted genes were identified, suggesting that imprinted loci may be particularly susceptible to diet effects. Lastly, we identified several differentially methylated microRNAs that target genes associated with immunological, metabolic, gastrointestinal, cardiovascular, and neurological chronic diseases, as well as genes responsible for transplacental nutrient transfer and fetal development.


Sujet(s)
Restriction calorique/effets indésirables , Méthylation de l'ADN/génétique , Retard de croissance intra-utérin/génétique , Placenta/métabolisme , Animaux , Femelle , Mâle , Souris de lignée C57BL , Grossesse , Caractères sexuels
19.
Endocrinology ; 153(8): 3995-4007, 2012 Aug.
Article de Anglais | MEDLINE | ID: mdl-22700768

RÉSUMÉ

Calorie restriction (CR) decreased placenta and fetal weights in wild-type (wt) and glucose transporter (Glut) 3 heterozygous null (glut3(+/-)) mice. Because placental nutrient transport is a primary energy determinant of placentofetal growth, we examined key transport systems. Maternal CR reduced intra- and transplacental glucose and leucine transport but enhanced system A amino acid transport in wt mice. These transport perturbations were accompanied by reduced placental Glut3 and leucine amino acid transporter (LAT) family member 2, no change in Glut1 and LAT family member 1, but increased sodium coupled neutral amino acid transporter (SNAT) and SNAT2 expression. We also noted decreased total and active phosphorylated forms of mammalian target of rapamycin, which is the intracellular nutrient sensor, the downstream total P70S6 kinase, and pS6 ribosomal protein with no change in total and phosphorylated 4E-binding protein 1. To determine the role of placental Glut3 in mediating CR-induced placental transport changes, we next investigated the effect of gestational CR in glut3(+/-) mice. In glut3(+/-) mice, a key role of placental Glut3 in mediating transplacental and intraplacental glucose transport was established. In addition, reduced Glut3 results in a compensatory increase of leucine and system A transplacental transport. On the other hand, diminished Glut3-mediated intraplacental glucose transport reduced leucine transport and mammalian target of rapamycin and preserved LAT and enhancing SNAT. CR in glut3(+/-) mice further reduced transplacental glucose transport and enhanced system A amino acid transport, although the increased leucine transport was lost. In addition, increased Glut3 was seen and preserved Glut1, LAT, and SNAT. These placental changes collectively protect survival of wt and glut3(+/-) fetuses against maternal CR-imposed reduction of macromolecular nutrients.


Sujet(s)
Acides aminés/métabolisme , Restriction calorique , Transporteur de glucose de type 3/métabolisme , Glucose/métabolisme , Placenta/métabolisme , Animaux , Femelle , Transporteur de glucose de type 1/génétique , Transporteur de glucose de type 1/métabolisme , Transporteur de glucose de type 3/génétique , Hétérozygote , Souris , Grossesse
20.
Am J Physiol Endocrinol Metab ; 297(4): E836-48, 2009 Oct.
Article de Anglais | MEDLINE | ID: mdl-19690067

RÉSUMÉ

Monosaccharides enter cells by slow translipid bilayer diffusion by rapid, protein-mediated, cation-dependent cotransport and by rapid, protein-mediated equilibrative transport. This review addresses protein-mediated, equilibrative glucose transport catalyzed by GLUT1, the first equilibrative glucose transporter to be identified, purified, and cloned. GLUT1 is a polytopic, membrane-spanning protein that is one of 13 members of the human equilibrative glucose transport protein family. We review GLUT1 catalytic and ligand-binding properties and interpret these behaviors in the context of several putative mechanisms for protein-mediated transport. We conclude that no single model satisfactorily explains GLUT1 behavior. We then review GLUT1 topology, subunit architecture, and oligomeric structure and examine a new model for sugar transport that combines structural and kinetic analyses to satisfactorily reproduce GLUT1 behavior in human erythrocytes. We next review GLUT1 cell biology and the transcriptional and posttranscriptional regulation of GLUT1 expression in the context of development and in response to glucose perturbations and hypoxia in blood-tissue barriers. Emphasis is placed on transgenic GLUT1 overexpression and null mutant model systems, the latter serving as surrogates for the human GLUT1 deficiency syndrome. Finally, we review the role of GLUT1 in the absence or deficiency of a related isoform, GLUT3, toward establishing the physiological significance of coordination between these two isoforms.


Sujet(s)
Transporteurs de glucose par diffusion facilitée/métabolisme , Séquence d'acides aminés , Animaux , Erreurs innées du métabolisme glucidique/génétique , Catalyse , Diffusion , Érythrocytes/métabolisme , Femelle , Transporteurs de glucose par diffusion facilitée/déficit , Transporteurs de glucose par diffusion facilitée/génétique , Transporteur de glucose de type 1/métabolisme , Humains , Isomérie , Cinétique , Modèles moléculaires , Données de séquences moléculaires , Mutation/physiologie , Grossesse , Conformation des protéines , Spécificité du substrat
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE