Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Int J Gynaecol Obstet ; 164(2): 750-757, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37589399

ABSTRACT

OBJECTIVE: Poor intrauterine growth has negative impacts for child growth and development and disproportionately affects children living in low-resource settings. In the present study, we investigated relationships between placental pathologies and indicators of poor intrauterine growth. METHODS: We enrolled a longitudinal cohort of 279 mother-infant pairs from Leyte, the Philippines. Placental measures included characteristics, pathological findings, and immunohistochemistry. At birth, intrauterine growth was assessed using anthropometric measures, weight-for-gestational age, and the clinical assessment of nutritional status score (CANSCORE) for determining fetal malnutrition. Multivariate linear regression and log-binomial regression models were applied, controlling for potential confounding factors. RESULTS: Maternal vascular malperfusion (MVM) was related to reduced birthweight (P < 0.0001), birth length (P = 0.002), head circumference (P = 0.001), and weight-to-length ratio (P = 0.016). MVM increased the risk for preterm delivery (P = 0.0005) and small for gestational age (SGA) (P = 0.016). Acute chorioamnionitis (P = 0.013) and MVM (P = 0.021) both led to an increased risk for fetal malnutrition defined by CANSORE<25. Villous tissue activated caspase-3 was associated with lower birth length (P = 0.0006), higher weight-to-length ratio (P = 0.004), reduced risks for SGA (P = 0.011) and low weight-to-length ratio for gestational age (P = 0.004). CONCLUSION: The present study applied comprehensive measures for intrauterine growth and demonstrates that low placental weight and placental pathology, chiefly MVM, contribute to poor intrauterine growth. A better understanding of the mechanistic role of specific placental pathologies on adverse newborn outcomes will provide opportunities for reducing incidence of poor intrauterine growth and associated long-term morbidities.


Subject(s)
Fetal Nutrition Disorders , Placenta , Infant, Newborn , Child , Pregnancy , Female , Humans , Placenta/blood supply , Pregnancy Outcome/epidemiology , Mothers , Fetal Nutrition Disorders/pathology , Philippines/epidemiology , Retrospective Studies , Fetal Growth Retardation/epidemiology , Fetal Growth Retardation/etiology
2.
Curr Pediatr Rev ; 19(2): 187-196, 2023.
Article in English | MEDLINE | ID: mdl-35792119

ABSTRACT

AIMS: This case-control study was conducted to identify maternal and placental risk factors of small-for-gestational-age (SGA) and fetal malnutrition. METHODS: Cases comprised 104 consecutively delivered SGA neonates (determined as per INTERGROWTH- 21st standard). An equal number of next-born gestation and gender-matched appropriatefor- gestational age (AGA) neonates served as controls. Maternal risk factors were enquired, and placentae were evaluated by clinical and histopathological examination. Nutrition of the neonates was assessed by the clinical assessment of nutrition (CAN) score. Univariate and multivariate logistic regression analysis was done to identify the maternal and placental risk factors. RESULTS: The prevalence of SGA in the present study was 23.9%. Maternal fever [adjusted Odds Ratio (aOR), 95% confidence interval (CI), 16.3 (3.5-124.1); p = 0.001], presence of placental syncytial knots [aOR (95% CI), 2.9 (1.1-9.1); p = 0.04] and placental calcifications [aOR (95% CI), 3(1.1- 8.7); p = 0.03], were identified as independent predictors of SGA using multivariate logistic regression analysis. Malnutrition (SCORE <25) affected 64% of SGA and 16.3% of AGA neonates. The only risk factor significantly associated with malnourished SGA was prematurity, whereas malnourished AGA was significantly associated with prematurity and fetal distress. In-hospital morbidities significantly higher in SGA were perinatal asphyxia, respiratory distress, need for respiratory support, polycythemia, hypoglycemia, and feeding intolerance. Mortality before discharge was 4.8% and 3.8% in SGA and AGA population, respectively (p > 0.05). Neonatal outcomes were comparable among well-nourished, malnourished SGA and AGA groups. CONCLUSION: Maternal fever, placental syncytial knots, and calcifications were independent risk factors of SGA, whereas prematurity and fetal distress were responsible for malnutrition.


Subject(s)
Fetal Nutrition Disorders , Placenta , Infant, Newborn , Pregnancy , Female , Humans , Placenta/pathology , Gestational Age , Case-Control Studies , Fetal Nutrition Disorders/pathology , Fetal Distress/complications , Fetal Distress/pathology , Fetal Growth Retardation/epidemiology , Risk Factors
3.
Clin Sci (Lond) ; 135(9): 1103-1126, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33899910

ABSTRACT

Poor maternal nutrition in pregnancy affects fetal development, predisposing offspring to cardiometabolic diseases. The role of mitochondria during fetal development on later-life cardiac dysfunction caused by maternal nutrient reduction (MNR) remains unexplored. We hypothesized that MNR during gestation causes fetal cardiac bioenergetic deficits, compromising cardiac mitochondrial metabolism and reserve capacity. To enable human translation, we developed a primate baboon model (Papio spp.) of moderate MNR in which mothers receive 70% of control nutrition during pregnancy, resulting in intrauterine growth restriction (IUGR) offspring and later exhibiting myocardial remodeling and heart failure at human equivalent ∼25 years. Term control and MNR baboon offspring were necropsied following cesarean-section, and left ventricle (LV) samples were collected. MNR adversely impacted fetal cardiac LV mitochondria in a sex-dependent fashion. Increased maternal plasma aspartate aminotransferase, creatine phosphokinase (CPK), and elevated cortisol levels in MNR concomitant with decreased blood insulin in male fetal MNR were measured. MNR resulted in a two-fold increase in fetal LV mitochondrial DNA (mtDNA). MNR resulted in increased transcripts for several respiratory chain (NDUFB8, UQCRC1, and cytochrome c) and adenosine triphosphate (ATP) synthase proteins. However, MNR fetal LV mitochondrial complex I and complex II/III activities were significantly decreased, possibly contributing to the 73% decreased ATP content and increased lipid peroxidation. MNR fetal LV showed mitochondria with sparse and disarranged cristae dysmorphology. Conclusion: MNR disruption of fetal cardiac mitochondrial fitness likely contributes to the documented developmental programming of adult cardiac dysfunction, indicating a programmed mitochondrial inability to deliver sufficient energy to cardiac tissues as a chronic mechanism for later-life heart failure.


Subject(s)
Fetal Nutrition Disorders/metabolism , Maternal Nutritional Physiological Phenomena , Mitochondria, Heart/metabolism , Adenine Nucleotides/metabolism , Animals , Female , Fetal Nutrition Disorders/pathology , Mitochondria, Heart/ultrastructure , Oxidative Stress , Papio , Pregnancy
4.
Front Endocrinol (Lausanne) ; 11: 591559, 2020.
Article in English | MEDLINE | ID: mdl-33324346

ABSTRACT

Excessive dietary fat intake has extensive impacts on several physiological systems and can lead to metabolic and nonmetabolic disease. In animal models of ingestion, exposure to a high fat diet during pregnancy predisposes offspring to increase intake of dietary fat and causes increase in weight gain that can lead to obesity, and without intervention, these physiological and behavioral consequences can persist for several generations. The hypothalamus is a region of the brain that responds to physiological hunger and fullness and contains orexigenic neuropeptide systems that have long been associated with dietary fat intake. The past fifteen years of research show that prenatal exposure to a high fat diet increases neurogenesis of these neuropeptide systems in offspring brain and are correlated to behavioral changes that induce a pro-consummatory and obesogenic phenotype. Current research has uncovered several potential molecular mechanisms by which excessive dietary fat alters the hypothalamus and involve dietary fatty acids, the immune system, gut microbiota, and transcriptional and epigenetic changes. This review will examine the current knowledge of dietary fat-associated changes in the hypothalamus and the potential pathways involved in modifying the development of orexigenic peptide neurons that lead to changes in ingestive behavior, with a special emphasis on inflammation by chemokines.


Subject(s)
Dietary Fats/adverse effects , Eating , Fetal Nutrition Disorders/pathology , Hypothalamus/pathology , Inflammation Mediators/metabolism , Inflammation/pathology , Prenatal Exposure Delayed Effects/pathology , Animals , Female , Fetal Nutrition Disorders/etiology , Fetal Nutrition Disorders/metabolism , Humans , Hypothalamus/metabolism , Inflammation/etiology , Inflammation/metabolism , Neuropeptides/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/etiology , Prenatal Exposure Delayed Effects/metabolism
5.
PLoS One ; 15(7): e0235840, 2020.
Article in English | MEDLINE | ID: mdl-32702025

ABSTRACT

OBJECTIVES: Maternal protein malnutrition is associated with impaired fetal growth, and lifetime consequences for the offspring. Our group has previously developed a model of protein-restriction in the non-human primate, which was associated with fetal growth restriction, stillbirth, decreased placental perfusion, and evidence of fetal hypoxia, suggesting perturbed vascular development. Our objective was to histologically characterize the micro-anatomic alterations associated with adverse pregnancy outcomes taking an approach that permits investigation of the 3D vascular structure and surrounding histology without the requirement for 3D vascular casting or relying on 2D stereology which both have methodological limitations. METHODS: Rhesus macaques were assigned in the pre-gestational period to a control diet that contained 26% protein, or study diet containing 13% protein (50% PR diet). Placental tissue was collected at delivery and processed using a clarification, immunohistochemistry, and confocal microscopy protocol published previously by our group. Three dimensional reconstructions and quantitative assessment of the vascular micro-anatomy was performed using analysis software (Imaris®) and statistical analysis accounted for maternal and fetal confounders. RESULTS: In unadjusted analysis, when comparing those pregnancies on a 50% PR diet (n = 4) with those on a control diet (n = 4), protein-restriction diet was associated with decreased maternal pre-pregnancy weight (difference of -1.975kg, 95% CI -3.267 to -0.6826). When controlling for maternal pre-pregnancy weight, fetal sex, and latency from tissue collection to imaging, a gestational protein-restriction diet was associated with decreases in total vascular length, total vascular surface area, total vascular volume, and vascular density. CONCLUSION: In this pilot study, a gestational protein-restriction diet altered the placental micro-vasculature with decreased vascular caliber and density, which may be related to the observed adverse pregnancy outcomes and perturbed placental perfusion previously demonstrated in this model.


Subject(s)
Diet, Protein-Restricted/adverse effects , Fetal Growth Retardation/pathology , Fetal Nutrition Disorders/pathology , Placenta/pathology , Animals , Disease Models, Animal , Female , Fetal Growth Retardation/etiology , Fetal Nutrition Disorders/etiology , Humans , Infant, Small for Gestational Age , Macaca mulatta/embryology , Macaca mulatta/physiology , Pilot Projects , Placental Circulation , Pregnancy , Stillbirth
6.
Physiol Rep ; 8(3): e14359, 2020 02.
Article in English | MEDLINE | ID: mdl-32026612

ABSTRACT

We hypothesized that late gestation malnutrition differentially affects expandability of adipose tissues to predispose for early postnatal visceral adiposity. Twin-lambs born to dams fed HIGH (150%/110% of required energy/protein, respectively), NORM (100% of requirements) or LOW (50% of NORM) diets during the last trimester were used. Postnatally, lambs were raised on moderate (CONV) or high-carbohydrate-high-fat (HCHF) diets. Adipose tissues were sampled at autopsy at 6 months of age (~puberty) to characterize cellularity, adipocyte cross-sectional area and gene expression patterns. HIGH and LOW compared to NORM lambs had reduced intrinsic (under CONV diet) cellularity in subcutaneous and mesenteric (particularly LOW), and reduced obesity-induced (under HCHF diet) hyperplasia in subcutaneous, mesenteric and perirenal (particularly HIGH) adipose tissues. This corresponded with more pronounced HCHF diet-induced hypertrophy in mesenteric (particularly LOW), perirenal (particularly HIGH) and subcutaneous (particularly HIGH) adipose tissues, and tissue-specific reductions in mRNA expressions for lipid metabolism, angiogenesis and adipose development. Gene expression for inflammation and lipid metabolism markers were increased and decreased, respectively, in HCHF lambs (HCHF lambs became obese) in all tissues. Both prenatal over- and undernutrition predisposed for abdominal adiposity and extreme perirenal hypertrophy due to reduced intrinsic (observed under CONV diet) cellularity and impaired ability of subcutaneous, mesenteric and perirenal adipose tissues to expand by hyperplasia rather than hypertrophy on an obesogenic (HCHF) diet.


Subject(s)
Fetal Nutrition Disorders/metabolism , Intra-Abdominal Fat/metabolism , Lipid Metabolism , Obesity/metabolism , Adiposity , Animals , Diet, High-Fat/adverse effects , Female , Fetal Nutrition Disorders/pathology , Intra-Abdominal Fat/pathology , Male , Obesity/etiology , Obesity/pathology , Sheep
7.
Neuroscience ; 380: 14-26, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29631020

ABSTRACT

Brain structural connectivity is known to be altered in cases of intrauterine growth restriction and premature birth, although the specific effect of maternal nutritional restriction, a common burden in human populations, has not been assessed yet. Here we analyze the effects of maternal undernutrition during pregnancy and lactation by establishing three experimental groups of female mice divided according to their diet: control (Co), moderate calorie-protein restriction (MCP) and severe protein restriction (SP). Nutritionally restricted dams gained relatively less weight during pregnancy and the body weight of the offspring was also affected by maternal undernutrition, showing global growth restriction. We performed magnetic resonance imaging (MRI) of the offspring's brains after weaning and analyzed their connectivity patterns using complex graph theory. In general, changes observed in the MCP group were more subtle than in SP. Results indicated that brain structures were not homogeneously affected by early nutritional stress. In particular, the growth of central brain regions, such as the temporo-parietal cortex, and long integrative myelinated tracts were relatively preserved, while the frequency of short tracts was relatively reduced. We also found a differential effect on network parameters: network degree, clustering, characteristic path length and small-worldness remained mainly unchanged, while the rich-club index was lower in nutritionally restricted animals. Rich-club decrease reflects an impairment in the structure by which brain regions with large number of connections tend to be more densely linked among themselves. Overall, the findings presented here support the hypothesis that chronic nutritional stress produces long-term changes in brain structural connectivity.


Subject(s)
Brain/pathology , Fetal Nutrition Disorders/pathology , Neural Pathways/pathology , Prenatal Exposure Delayed Effects/pathology , Animals , Brain/growth & development , Female , Fetal Growth Retardation/etiology , Fetal Growth Retardation/pathology , Magnetic Resonance Imaging , Mice , Mice, Inbred C57BL , Neural Pathways/growth & development , Pregnancy , Prenatal Nutritional Physiological Phenomena
8.
Gen Comp Endocrinol ; 246: 258-269, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28041790

ABSTRACT

Experimental data demonstrated the negative impact of maternal protein malnutrition (MPM) on rat prostate development, but the mechanism behind the impairment of prostate growth has not been well understood. Male Sprague Dawley rats, borned to dams fed a normal protein diet (CTR group, 17% protein diet), were compared with those borned from dams fed a low protein diet (6% protein diet) during gestation (GLP group) or gestation and lactation (GLLP). The ventral prostate lobes (VP) were removed at post-natal day (PND) 10 and 21, and analyzed via different methods. The main findings were low birth weight, a reduction in ano-genital distance (AGD, a testosterone-dependent parameter), and an impairment of prostate development. A delay in prostate morphogenesis was associated with a reduced testosterone levels and angiogenic process through downregulation of aquaporin-1 (AQP-1), insulin/IGF-1 axis and VEGF signaling pathway. Depletion of the microvascular network, which occurs in parallel to the impairment of proliferation and differentiation of the epithelial cells, affects the bidirectional flux between blood vessels impacting prostatic development. In conclusion, our data support the hypothesis that a reduction in microvascular angiogenesis, especially in the subepithelial compartment, is associated to the impairment of prostate morphogenesis in the offspring of MPM dams.


Subject(s)
Fetal Development , Fetal Nutrition Disorders/pathology , Microvessels/embryology , Neovascularization, Pathologic/pathology , Prostate/pathology , Protein-Energy Malnutrition/physiopathology , Animals , Animals, Newborn , Blotting, Western , Female , Insulin/blood , Insulin-Like Growth Factor I/metabolism , Lactation/physiology , Male , Pregnancy , Prostate/blood supply , Prostate/metabolism , Rats , Rats, Sprague-Dawley , Testosterone/blood , Vascular Endothelial Growth Factor A/metabolism
9.
Int J Clin Exp Pathol ; 7(6): 3101-11, 2014.
Article in English | MEDLINE | ID: mdl-25031729

ABSTRACT

Epidemiological studies have demonstrated that intrauterine growth restriction (IUGR) increases the risk for respiratory morbidity from infancy, throughout childhood and into adulthood. Chronic restriction of nutrients causes abnormalities in the airways and lungs of offspring, but whether IUGR adversely impacts fetal pulmonary vascular development and underlying mechanisms remain under investigation. In this study, we investigated the effects of protein malnutrition in utero on pulmonary alveolarization and vascular growth of the fetal lung and placentae. Pregnant rats were feed with an isocaloric low-protein diet (8% protein) until delivery. Placenta and fetal lungs were harvested on 20th day of gestation (term 21 days of gestation). Lung index (lung weight as a percentage of body weight), total DNA and protein, radial alveolar count, arteriolar wall thickness, lung maturity and angiogenic factor VEGF were assessed. The lung was hypoplastic in IUGR fetus, evidenced by reduction in lung weight, DNA and protein content. Protein restriction in utero led to higher glycogen levels, but reduced number of alveoli as confirmed by the measurement of radial alveolar counts. IUGR fetus had significantly reduced VEGF, Flk-1 levels in lung but no changes in Flt-1 mRNA. Furthermore, IUGR was associated with increased lung miR-126-3p levels, which modulated the expression of angiogenic factor. In contrast, with regard to the placenta, IUGR fetus presented with decreased expression of VEGF, with no changes in VEGF receptors and expression-regulating miRNAs. This work suggested that VEGF signaling defect plays an important role in the defective lung development, which may explain the increased incidence of respiratory infections in IUGR patients.


Subject(s)
Fetal Growth Retardation/metabolism , Fetal Growth Retardation/pathology , Pulmonary Alveoli/blood supply , Pulmonary Alveoli/embryology , Pulmonary Alveoli/pathology , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Animals , Disease Models, Animal , Female , Fetal Nutrition Disorders/metabolism , Fetal Nutrition Disorders/pathology , Fetus , Immunoblotting , Lung/blood supply , Lung/embryology , Lung/metabolism , Pregnancy , Prenatal Exposure Delayed Effects , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction
10.
Am J Phys Anthropol ; 154(4): 544-53, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24888714

ABSTRACT

Nutritional imbalance is one of the main sources of stress in both extant and extinct human populations. Restricted availability of nutrients is thought to disrupt the buffering mechanisms that contribute to developmental stability and canalization, resulting in increased levels of fluctuating asymmetry (FA) and phenotypic variance among individuals. However, the literature is contradictory in this regard. This study assesses the effect of prenatal nutritional stress on FA and among-individual variance in cranial shape and size using a mouse model of maternal protein restriction. Two sets of landmark coordinates were digitized in three dimensions from skulls of control and protein restricted specimens at E17.5 and E18.5. We found that, by the end of gestation, maternal protein restriction resulted in a significant reduction of skull size. Fluctuating asymmetry in size and shape exceeded the amount of measurement error in all groups, but no significant differences in the magnitude of FA were found between treatments. Conversely, the pattern of shape asymmetry was affected by the environmental perturbation since the angles between the first eigenvectors extracted from the covariance matrix of shape asymmetric component of protein restricted and control groups were not significantly different from the expected for random vectors. In addition, among-individual variance in cranial shape was significantly higher in the protein restricted than the control group at E18.5. Overall, the results obtained from a controlled experiment do not support the view of fluctuating asymmetry of cranial structures as a reliable index for inferring nutritional stress in human populations.


Subject(s)
Fetal Nutrition Disorders/pathology , Fetus/pathology , Skull/pathology , Animals , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Organ Size , Phenotype , Skull/growth & development
11.
BMC Genomics ; 15: 49, 2014 Jan 21.
Article in English | MEDLINE | ID: mdl-24447410

ABSTRACT

BACKGROUND: Maternal undernutrition leads to an increased risk of metabolic disorders in offspring including obesity and insulin resistance, thought to be due to a programmed thrifty phenotype which is inappropriate for a subsequent richer nutritional environment. In a rat model, both male and female offspring of undernourished mothers are programmed to become obese, however postnatal leptin treatment gives discordant results between males and females. Leptin treatment is able to rescue the adverse programming effects in the female offspring of undernourished mothers, but not in their male offspring. Additionally, in these rats, postnatal leptin treatment of offspring from normally-nourished mothers programmes their male offspring to develop obesity in later life, while there is no comparable effect in their female offspring. RESULTS: We show by microarray analysis of the female liver transcriptome that both maternal undernutrition and postnatal leptin treatment independently induce a similar thrifty transcriptional programme affecting carbohydrate metabolism, amino acid metabolism and oxidative stress genes. Paradoxically, however, the combination of both stimuli restores a more normal transcriptional environment. This demonstrates that "leptin reversal" is a global phenomenon affecting all genes involved in fetal programming by maternal undernourishment and leptin treatment. The thrifty transcriptional programme was associated with pro-inflammatory markers and downregulation of adaptive immune mediators, particularly MHC class I genes, suggesting a deficit in antigen presentation in these offspring. CONCLUSIONS: We propose a revised model of developmental programming reconciling the male and female observations, in which there are two competing programmes which collectively drive liver transcription. The first element is a thrifty metabolic phenotype induced by early life growth restriction independently of leptin levels. The second is a homeostatic set point calibrated in response to postnatal leptin surge, which is able to over-ride the metabolic programme. This "calibration model" for the postnatal leptin surge, if applicable in humans, may have implications for understanding responses to catch-up growth in infants. Additionally, the identification of an antigen presentation deficit associated with metabolic thriftiness may relate to a previously observed correlation between birth season (a proxy for gestational undernutrition) and infectious disease mortality in rural African communities.


Subject(s)
Fetal Nutrition Disorders/genetics , Leptin/pharmacology , Liver/drug effects , Amino Acids/metabolism , Animals , Carbohydrate Metabolism/genetics , Diet , Disease Models, Animal , Female , Fetal Development , Fetal Nutrition Disorders/metabolism , Fetal Nutrition Disorders/pathology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Inflammation Mediators/metabolism , Liver/metabolism , Male , Obesity/metabolism , Obesity/pathology , Oxidative Stress/genetics , Phenotype , Pregnancy , Rats , Rats, Wistar , Transcriptome/drug effects
12.
Life Sci ; 92(13): 763-74, 2013 Apr 19.
Article in English | MEDLINE | ID: mdl-23439325

ABSTRACT

AIMS: Maternal malnutrition by low protein diet is associated with an increased incidence of metabolic disorders and decreased male fertility in adult life. This study aimed to assess the impact of maternal protein malnutrition (MPM) on prostate growth, tissue organization and lesion incidence with aging. MAIN METHODS: Wistar rat dams were distributed into two groups, which were control (NP; fed a normal diet containing 17% protein) or a restricted protein diet (RP, fed a diet containing 6% protein) during gestation. After delivery all mothers and offspring received a normal diet. Biometrical parameters, hormonal levels and prostates were harvested at post-natal days (PND) 30, 120 and 360. KEY FINDINGS: MPM promoted low birth weight, decreased ano-genital distance (AGD) and reduced androgen plasma levels of male pups. Prostatic lobes from RP groups presented reduced glandular weight, epithelial cell height and alveolar diameter. The epithelial cell proliferation and collagen deposition were increased in RP group. Incidences of epithelial dysplasia and prostatitis were higher in the RP offspring than in the NP offspring at PND360. SIGNIFICANCE: Our findings show that MPM delays prostate development, growth and maturation until adulthood, probably as a result of low testosterone stimuli. The higher incidence of cellular dysplasia and prostatitis suggests that MPM increases prostate susceptibility to diseases with aging.


Subject(s)
Diet, Protein-Restricted/adverse effects , Fetal Nutrition Disorders/pathology , Prostate/growth & development , Prostate/pathology , Prostatitis/etiology , Prostatitis/pathology , Aging , Animals , Animals, Newborn , Apoptosis , Body Weight , Collagen/analysis , Eating , Female , Fetal Nutrition Disorders/blood , Fetal Nutrition Disorders/physiopathology , Male , Pregnancy , Prostatitis/blood , Prostatitis/physiopathology , Rats , Rats, Wistar , Receptors, Androgen/analysis , Testosterone/blood
14.
J Paediatr Child Health ; 48(10): 926-30, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22897800

ABSTRACT

AIM: The aim of this study was to determine the frequency, risk factors and anthropometric measurements of fetally malnourished, liveborn singleton term neonates. METHODS: The computed delivery room data of 11.741 liveborn singleton term neonates was used to compare malnourished and nourished newborns. RESULTS: Of the total subjects, 577 (4.9%) were malnourished. There were no differences between the groups with regard to gender distribution, Apgar scores, maternal parity, smoking during pregnancy and type of delivery. Maternal age and neonatal gestational age (GA) were significantly lower in malnourished newborns (P < 0.001). Birthweight (BW), birth length (BL) and head circumference (HC) were significantly lower in the malnourished group compared with well-nourished group (P < 0.001). Mean BW (g) was 2724.7 ± 17.0 in the malnourished group versus 3234.3 ± 3.8 in the well-nourished group; BL (cm) was 47.8 ± 0.1 in malnourished versus 49.5 ± 0.0 in well-nourished neonates; HC (cm) was 33.25 ± 0.1 in the malnourished versus 34.3 ± 0.0 in the well-nourished group. Between the groups, there were significant differences in the ratio of small, appropriate and large for GA (P < 0.001). Of the malnourished newborns, 35.5% were small for GA, 63.3% were appropriate for GA and 1.2% were large for GA. CONCLUSION: Fetal malnutrition (FM) still exists despite the advances in current obstetric care. Neonates of adolescent mothers and of low GA are particularly at risk for FM. The BW, BL and HC of fetally malnourished neonates are lower than that of well-nourished neonates. Like term singleton appropriate and small for GA neonates, term singleton large for GA neonates could also have been fetally malnourished.


Subject(s)
Fetal Nutrition Disorders , Adolescent , Adult , Birth Weight , Body Height , Case-Control Studies , Databases, Factual , Female , Fetal Nutrition Disorders/epidemiology , Fetal Nutrition Disorders/etiology , Fetal Nutrition Disorders/pathology , Gestational Age , Head/anatomy & histology , Humans , Infant, Newborn , Infant, Small for Gestational Age , Live Birth , Logistic Models , Male , Maternal Age , Pregnancy , Risk Factors , Term Birth , Turkey/epidemiology , Young Adult
15.
Int J Dev Neurosci ; 30(6): 445-50, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22819790

ABSTRACT

The aim of this study was to determine whether intrauterine malnutrition (IUM) produces a change in the expression of tryptophan-5-hydroxylase (TPH) 1 and/or 2 as the primary mechanism to explain the observed chronic cerebral acceleration of the synthesis of 5-hydroxytryptamine (5-HT). We used an IUM model and controls with ages of 1, 15 and 21 days. The brainstem was obtained to determine L-tryptophan, 5-HT and TPH activity. Expression of TPH1 and TPH2 via specific antibodies for each was also evaluated by immunocytochemistry and Western blot. Malnourished offspring had a significant elevation of L-Trp, TPH activity and 5-HT in the brainstem. Both isoforms (1 and 2) of TPH were expressed from birth in both groups; however, TPH1 expression was significantly higher in offspring with IUM in relation to the controls. Importantly, these malnourished offspring showed reduced expression of TPH2 compared to controls. It was confirmed that IUM produces an increase in 5-HT in the brainstem and also showed increased expression of TPH1 at birth, with decreased expression of TPH2. These findings together allow us to propose that chronic elevation of synthesis of 5-HT observed in the brain of the offspring with IUM is probably due to a change in the expression and activity of TPH1 induced from fetal life.


Subject(s)
Brain Stem/metabolism , Fetal Nutrition Disorders/pathology , Gene Expression Regulation, Developmental/physiology , Tryptophan Hydroxylase/metabolism , Age Factors , Animals , Animals, Newborn , Body Weight/physiology , Brain Stem/growth & development , Disease Models, Animal , Female , Male , Pregnancy , Rats , Rats, Wistar , Serotonin/metabolism
16.
Prog. obstet. ginecol. (Ed. impr.) ; 54(5): 231-234, mayo 2011. tab
Article in Spanish | IBECS | ID: ibc-142942

ABSTRACT

Para determinar la población de riesgo de síndrome de Down a la que debe recomendarse realizar una te ́cnica invasiva para obtener un cariotipo fetal existen varias pruebas de cribado. En nuestro centro se realiza con el cribado del primer trimestre (edad materna + - translucencia nucal +b-HCG + PAPP-A). Cuando éste no es posible, se realiza el doble test (AFP+b- HCG) como cribado de segundo trimestre. Los resultados del doble test en nuestro centro son insatisfactorios ya que tenemos una tasa de detección del 0%, con una tasa de falsos positivos del 7,7%. Por ello consideramos que es necesaria la utilización de otros criterios alternativos para la selección de las mujeres de riesgo de síndrome de Down en el segundo trimestre (AU)


Several screening tests are available to identify the population at risk of Down syndrome. This population should then be recommended to undergo an invasive technique to obtain a fetal karyotype. In our hospital, screening is done by the combined test (maternal age + nuchal translucency + b subunit of human chorionic gonadotropin [b-HCG] + pregnancy- associated plasma-A [PAPP-A]) during the first trimester of pregnancy. When this test is not feasible, we request the double test (alpha-fetoprotein [AFP] + b-HCG) as a second trimester screening test. The results of the double test in our hospital were unsatisfactory because the detection rate was 0% with a false positive rate of 7.7%. Therefore, we believe alternative criteria should be used to select women at risk of having a child with Down syndrome in the second trimester (AU)


Subject(s)
Female , Humans , Pregnancy , Clinical Chemistry Tests/methods , Maternal Serum Screening Tests/methods , Maternal Serum Screening Tests , Down Syndrome/genetics , Down Syndrome/pathology , Pregnant Women/psychology , Fetal Nutrition Disorders/genetics , Fetal Therapies/methods , Fetal Therapies/standards , Clinical Chemistry Tests , Maternal Serum Screening Tests/classification , Maternal Serum Screening Tests/standards , Down Syndrome/complications , Down Syndrome/metabolism , Fetal Nutrition Disorders/pathology , Fetal Therapies/instrumentation , Fetal Therapies
17.
Acta Med Port ; 24(5): 799-804, 2011.
Article in Portuguese | MEDLINE | ID: mdl-22525632

ABSTRACT

BACKGROUND: Many epidemiological studies suggest that the intra uterine environment is extremely important in determining the future of individual health. Changes in maternal nutritional status, reflected in birth weight may program the offspring to the development of diseases in adulthood. Studies with animals exposed to intrauterine malnutrition have suggested a reduction in the number of clusters in addition to increased blood pressure. OBJECTIVE: To review the literature morphophysiology changes in the kidney of adult Wistar rats exposed to malnutrition during intrauterine life. METHODS: We performed a search in the following databases: PubMed, MEDLINE, SCIENCE DIRECT, LILACS. The main search terms were malnutrition and renal function in Portuguese and English. We included original articles involving albino rats. We excluded review articles as well as those involving humans. RESULTS: According to Franco et al (2009) renal function and the number of glomeruli reduced by poor on intrauterine development, predisposing offspring to kidney disease in adulthood. According to Chen (2009) ultra-glomerular structure is not affected by maternal malnutrition suggesting that this factor does not contribute to the pathogenesis of hypertension after maternal malnutrition. Viana-son et al (2009) believes that stress on placents, causes disruption on sodium pumps in the proximal tubules of the kidney, resulting hypertension. CONCLUSION: The intrauterine malnutrition appears to interfere with the programming of kidney function with changes in glomerular morphophysiology, however, its mechanisms remain uncertain. We suggest further studies of the type and randomized clinical trials aimed at understanding the factors that trigger this process.


Subject(s)
Fetal Nutrition Disorders/pathology , Fetal Nutrition Disorders/physiopathology , Kidney/pathology , Kidney/physiopathology , Malnutrition/pathology , Malnutrition/physiopathology , Animals , Rats , Rats, Wistar
19.
Cerebellum ; 9(1): 136-44, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19838760

ABSTRACT

Copper is required for activity of several key enzymes and for optimal mammalian development, especially within the central nervous system. Copper-deficient (CuD) animals are visibly ataxic, and previous studies in rats have demonstrated impaired motor function through behavioral experiments consistent with altered cerebellar development. Perinatal copper deficiency was produced in Holtzman rat dams by restricting dietary copper during the last two thirds of gestation and lactation. Male offspring were evaluated at postnatal day 25. Compared to cerebella from copper-adequate pups, the CuD pups had larger Purkinje cell (PC) size and irregularities in the Purkinje cell monolayer. These results suggest that the ataxic behavioral phenotype of CuD rats may result from disrupted inhibitory pathways in the cerebellum. A similar PC phenotype is seen in Menkes disease and in mottled mouse mutants with genetic copper deficiency, suggesting that copper deficiency and not just specific loss of ATP7A function is responsible.


Subject(s)
Cerebellum/growth & development , Cerebellum/pathology , Copper/deficiency , Fetal Nutrition Disorders/pathology , Neurogenesis/physiology , Purkinje Cells/pathology , Animals , Cell Count , Cell Differentiation/physiology , Cell Size , Cerebellum/metabolism , Disease Models, Animal , Female , Fetal Nutrition Disorders/metabolism , Fetal Nutrition Disorders/physiopathology , Food, Formulated/adverse effects , Humans , Infant Nutrition Disorders/metabolism , Infant Nutrition Disorders/pathology , Infant Nutrition Disorders/physiopathology , Infant, Newborn , Male , Pregnancy , Purkinje Cells/metabolism , Rats , Rats, Sprague-Dawley
20.
Pediatr Int ; 51(4): 494-7, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19400817

ABSTRACT

BACKGROUND: The aim of the present study was to investigate placental pathologies in malnourished term neonates. METHODS: A group of term newborns was evaluated at birth for fetal malnutrition (FM) using the Clinical Assessment of Nutritional Status (CANS) score. The study group consisted of 37 malnourished neonates and 13 well-nourished newborns, including their placentas. Infants with FM were subdivided into two groups: appropriate for gestational age (AGA) and small for gestational age (SGA). RESULTS: The proportion of subjects having antepartum complications was 18.9% in the FM group, whereas none was found in the control group. Similarly, a significant portion, 83.8% of the study group, had gross placental abnormalities, while the control group had none. The placental microscopic findings in the study group included perivillous fibrin deposition, calcification, necrosis, retroplacental hematoma, and infarction. In contrast, the control group had only perivillous fibrin deposition and calcification. Placental histopathological findings between FM term AGA and SGA neonates were also compared, and no statistically significant differences were found (P < 0.05). CONCLUSIONS: Placental findings in malnourished term AGA neonates are not different from findings documented in malnourished SGA cases, but they are different from those in well-nourished term AGA newborns.


Subject(s)
Fetal Nutrition Disorders/pathology , Placenta/pathology , Female , Humans , Infant, Newborn , Nutritional Status , Pregnancy , Prospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL
...