Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
1.
Mol Cell Endocrinol ; 592: 112316, 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38880278

RESUMEN

This study investigated the impact of maternal protein restriction (MPR) and early postnatal sugar consumption (SUG) on the liver health of adult male descendant rats. Male offspring of mothers fed a normal protein diet (NPD) or a low protein diet (LPD) were divided into four groups: Control (CTR), Sugar Control (CTR + SUG), LPD during gestation and lactation (GLLP), and LPD with sugar (GLLP + SUG). Sugar consumption (10% glucose diluted in water) began after weaning on day 21 (PND 21), and at 90 days (PND 90), rats were sacrificed for analysis. Sugar intake reduced food intake and increased water consumption in CTR + SUG and GLLP + SUG compared to CTR and GLLP. GLLP and GLLP + SUG groups showed lower body weight and total and retroperitoneal fat compared to CTR and CTR + SUG. CTR + SUG and GLLP + SUG groups exhibited hepatocyte vacuolization associated with increased hepatic glycogen content compared to CTR and GLLP. Hepatic catalase activity increased in GLLP compared to CTR. Proteomic analysis identified 223 differentially expressed proteins (DEPs) among experimental groups. While in the GLLP group, the DEPs enriched molecular pathways related to cellular stress, glycogen metabolic pathways were enriched in the GLLP + SUG and CTR + SUG groups. The association of sugar consumption amplifies the effects of MPR, deregulating molecular mechanisms related to metabolism and the antioxidant system.

2.
Arch Med Res ; 55(4): 103002, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38735235

RESUMEN

BACKGROUND: Maternal obesity (MO) has been shown to adversely affect metabolic, oxidative, reproductive, and cognitive function in offspring. However, it is unclear whether lifestyle modification can ameliorate the metabolic and organ dysfunction programmed by MO and prevent the effects of metabolic syndrome in adulthood. This study aimed to evaluate whether moderate voluntary exercise in the offspring of rats born to obese mothers can ameliorate the adverse effects of MO programming on metabolism and liver function in mid-adulthood. METHODS: Offspring of control (CF1) and MOF1 mothers were fed with a control diet from weaning. Adult males and females participated in 15 min exercise sessions five days/week. Metabolic parameters were analyzed before and after the exercise intervention. Liver oxidative stress biomarkers and antioxidant enzymes were analyzed before and after the intervention. RESULTS: Males showed that CF1ex ran more than MOF1ex and increased the distance covered. In contrast, females in both groups ran similar distances and remained constant but ran more distance than males. At PND 300 and 450, male and female MOF1 had higher leptin, triglycerides, insulin, and HOMA-IR levels than CF1. However, male MOF1ex had lower triglycerides, insulin, and HOMA-IR levels than MOF1. Improvements in liver fat and antioxidant enzymes were observed in CF1ex and MOF1ex males and females compared to their respective CF1 and MOF1 groups. CONCLUSION: These findings suggest that moderate voluntary exercise, even when started in mid-adulthood, can improve metabolic outcomes and delay accelerated metabolic aging in MO-programmed rats in a sex-dependent manner.


Asunto(s)
Envejecimiento , Obesidad Materna , Condicionamiento Físico Animal , Animales , Femenino , Masculino , Ratas , Embarazo , Envejecimiento/metabolismo , Obesidad Materna/metabolismo , Estrés Oxidativo , Ratas Wistar , Hígado/metabolismo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Obesidad/metabolismo , Obesidad/terapia , Obesidad/fisiopatología
3.
Mol Cell Endocrinol ; 588: 112223, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38556160

RESUMEN

Maternal malnutrition can alter developmental biology, programming health and disease in offspring. The increase in sugar consumption during the peripubertal period, a worldwide concern, also affects health through adulthood. Studies have shown that maternal exposure to a low protein diet (LPD) is associated with an increase in prostate disease with aging. However, the combined effects of maternal LPD and early postnatal sugar consumption on offspring prostate disorders were not investigated. The effects on aging were evaluated using a maternal gestational model with lactational LPD (6% protein) and sugar consumption (10%) from postnatal day (PND) 21-90, associating the consequences on ventral prostate (VP) rats morphophysiology on PND540. An increase was shown in mast cells and in the VP of the CTR + SUG and Gestational and Lactational Low Protein (GLLP) groups. In GLLP + SUG, a significant increase was shown in TGF-ß1 expression in both the systemic and intra-prostatic forms, and SMAD2/3p had increased. The study identified maternal LPD and sugar consumption as risk factors for prostatic homeostasis in senility, activating the TGFß1-SMAD2/3 pathway, a signaling pathway with potential markers for prostatic disorders.


Asunto(s)
Desnutrición , Fenómenos Fisiologicos Nutricionales Maternos , Efectos Tardíos de la Exposición Prenatal , Próstata , Enfermedades de la Próstata , Animales , Masculino , Femenino , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Enfermedades de la Próstata/patología , Enfermedades de la Próstata/etiología , Enfermedades de la Próstata/metabolismo , Desnutrición/complicaciones , Próstata/metabolismo , Próstata/patología , Ratas , Inflamación/patología , Inflamación/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta1/genética , Dieta con Restricción de Proteínas/efectos adversos , Proteína Smad2/metabolismo , Ratas Wistar , Proteína smad3/metabolismo , Proteína smad3/genética , Transducción de Señal , Animales Recién Nacidos , Mastocitos/metabolismo
4.
Arch Med Res ; 55(3): 102983, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38492326

RESUMEN

Maternal obesity predisposes offspring (F1) to cardiovascular disease. To evaluate basal heart function and ischemia-reperfusion (IR) responses in F1 males and females of obese mothers, female Wistar rats (F0) were fed chow or an obesogenic (MO) diet from weaning through pregnancy and lactation. Non-sibling F1 males and females were weaned to chow at postnatal day (PND) 21 and euthanized at PND 550. Offspring of MO mothers (MOF1) rarely survive beyond PND 650. Hearts were immediately isolated from euthanized F1s and subjected to 30 min ischemia with 20 min reperfusion. Retroperitoneal fat, serum triglycerides, glucose, insulin, and insulin resistance were measured. Baseline left ventricular developed pressure (LVDP) was lower in male and female MOF1 than in controls. After global ischemia, LVDP in control (C) male and female F1 recovered 78 and 83%, respectively, while recovery in MO male and female F1 was significantly lower at 28 and 52%, respectively. Following the IR challenge, MO hearts showed a higher functional susceptibility to reperfusion injury, resulting in lower cardiac reserve than controls in both sexes. Female hearts were more resistant to IR. Retroperitoneal fat was increased in male MOF1 vs. CF1. Circulating triglycerides and insulin resistance were increased in male and female MOF1 vs. CF1. These data show that MO programming reduces F1 cardiac reserve associated with age-related insulin resistance in a sex-specific manner.


Asunto(s)
Resistencia a la Insulina , Efectos Tardíos de la Exposición Prenatal , Humanos , Ratas , Femenino , Embarazo , Masculino , Animales , Anciano , Resistencia a la Insulina/fisiología , Ratas Wistar , Obesidad , Insulina , Triglicéridos , Dieta Alta en Grasa , Isquemia , Reperfusión
5.
J Dev Orig Health Dis ; 15: e4, 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38500346

RESUMEN

The aim of this study was to analyse the expression of genes related to the regulation of energy metabolism in skeletal muscle tissue by comparing male offspring in two age groups [at 110 and 245 postnatal days (pnd)] from a mother with obesity induced by a high-fat diet and (-)-epicatechin (Epi) administration. Four groups of six male offspring from different litters were randomly selected for the control groups [C and offspring of mothers with maternal obesity (MO)] or Epi intervention groups. We evaluated the effect of Epi on gastrocnemius tissue by analysing the mRNA and protein expression levels of Fndc5/irisin, Pgc-1α, Ucp3, and Sln. Epi significantly increased the Pgc-1α protein in the MO group of offspring at 110 pnd (p < 0.036, MO vs. MO+Epi), while at 245 pnd, Epi increased Fndc5/irisin mRNA expression in the MO+Epi group versus the MO group (p = 0.006).No differences were detected in Fndc5/irisin, Ucp3 or Sln mRNA or protein levels (including Pgc-1α mRNA) in the offspring at 110 pnd or in Pgc-1α, Ucp3, or Sln mRNA or protein levels (including Fndc5/irisin protein) at 245 pnd among the experimental groups. In conclusion, (-)-epicatechin treatment increased Fndc5/irisin mRNA expression and Pgc-α protein levels in the gastrocnemius muscle of offspring at postnatal days 110 and 245. Furthermore, it is suggested that the flavonoid effect in a model of obesity and its impact on thermogenesis in skeletal muscle are regulated by a different pathway than Fndc5/irisin.


Asunto(s)
Catequina , Obesidad Materna , Humanos , Embarazo , Ratas , Masculino , Femenino , Animales , Catequina/farmacología , Fibronectinas/genética , Fibronectinas/metabolismo , Fibronectinas/farmacología , Músculo Esquelético/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/farmacología , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Obesidad Materna/metabolismo , ARN Mensajero/genética
6.
Appetite ; 190: 107030, 2023 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-37678585

RESUMEN

Evidence about the association between breastfeeding and its duration with growth, appetite and satiety indicators, and adiposity in low and middle-income countries facing nutritional transition is scarce. The aim of this study was to evaluate the association between longitudinal patterns of breastfeeding (exclusive [EBF] and continued [CBF]) with adiposity and growth, and the mediating role of appetite and satiety indicators in these associations in Mexican children during the first 2 years of life. Information from 378 mother-child pairs from the MAS-Lactancia birth cohort was analysed. Information was collected at birth and at months 1, 3, 6, 9, 12, 18 and 24 of life. Duration of EBF and CBF was computed. Linear mixed models were used to assess the association of EBF and CBF with growth and adiposity. Path analysis was used for mediation analysis. Compared with the reference group (EBF duration <1 month), males with >3 to ≤6 months of EBF had less abdominal circumference (ß = -0.66, p = 0.05), Z-score weight-for-length (ß = -0.17, p = 0.19) and length-for-age (ß = -0.49, p < 0.01). Participants without CBF beyond 6 months had higher BMI Z-score (ß = 0.19, p < 0.01), abdominal circumference (ß = 0.62, p < 0.01) and skinfold sum (ß = 0.80, p = 0.09), and o difference in length-for-age. For EBF, mediation was confirmed for satiety responsiveness on the association with BMI Z-Score, for food fussiness for the association with abdominal circumference and length-for-age Z-score, and enjoyment of food on the association with length-for-age Z-score. For CBF, mediation was confirmed for food fussiness in the association with length-for-age. This study suggests that a longer exposure to EBF and CBF is associated with lower adiposity in children under 2 years of age, and that this association could be partially mediated by appetite and satiety indicators.

7.
Biology (Basel) ; 12(9)2023 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-37759566

RESUMEN

We investigated whether maternal obesity affects the hepatic mitochondrial electron transport chain (ETC), sirtuins, and antioxidant enzymes in young (110 postnatal days (PND)) and old (650PND) male and female offspring in a sex- and age-related manner. Female Wistar rats ate a control (C) or high-fat (MO) diet from weaning, through pregnancy and lactation. After weaning, the offspring ate the C diet and were euthanized at 110 and 650PND. The livers were collected for RNA-seq and immunohistochemistry. Male offspring livers had more differentially expressed genes (DEGs) down-regulated by both MO and natural aging than females. C-650PND vs. C-110PND and MO-110PND vs. C-110PND comparisons revealed 1477 DEGs in common for males (premature aging by MO) and 35 DEGs for females. Analysis to identify KEGG pathways enriched from genes in common showed changes in 511 and 3 KEGG pathways in the male and female livers, respectively. Mitochondrial function pathways showed ETC-related gene down-regulation. All ETC complexes, sirtuin2, sirtuin3, sod-1, and catalase, exhibited gene down-regulation and decreased protein expression at young and old ages in MO males vs. C males; meanwhile, MO females down-regulated only at 650PND. Conclusions: MO accelerates the age-associated down-regulation of ETC pathway gene expression in male offspring livers, thereby causing sex-dependent oxidative stress, premature aging, and metabolic dysfunction.

8.
J Biosci ; 482023.
Artículo en Inglés | MEDLINE | ID: mdl-37309173

RESUMEN

Maternal obesity programs the offspring to metabolic dysfunction. However, the effects of maternal obesity on skeletal muscle programming and ageing have been little explored. To determine if maternal obesity is a detriment to the progress of age-related muscle strength loss in the offspring (F1), we evaluated the indicators of muscle strength, adiposity, and metabolism at young adult and senior adult ages of maternal obesity F1 (MOF1) males and females from a high-fat diet-induced maternal obesity model in rat. Controls were agematched siblings whose mothers were fed a standard maternal diet (CF1). Combinatorial data analysis was performed with body weight (BW), forelimb grip strength (FGS), FGS adjusted with BW, body fat, adiposity index, and serum triacylglycerols, cholesterol, glucose, insulin, and homeostatic model assessment for insulin resistance variables, to identify discriminant traits of variation among F1 groups. During ageing, maternal obesity caused glucose and cholesterol metabolic dysfunctions in male F1, whereas adiposity-associated skeletal strength loss and fatty acid alterations were present in female offspring. In conclusion, offspring programming-ageing effects due to maternal obesity impact metabolism and skeletal muscle strength loss at later ages in a sex-dependent manner.


Asunto(s)
Obesidad Materna , Animales , Femenino , Masculino , Embarazo , Ratas , Envejecimiento , Peso Corporal , Glucosa , Fuerza Muscular , Obesidad
9.
Nutrients ; 15(10)2023 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-37242132

RESUMEN

We investigated whether excessive retroperitoneal adipose tissue (AT) expansion programmed by maternal obesity (MO) affects adipocyte size distribution and gene expression in relation to adipocyte proliferation and differentiation in male and female offspring (F1) from control (F1C) and obese (F1MO) mothers. Female Wistar rats (F0) ate a control or high-fat diet from weaning through pregnancy and lactation. F1 were weaned onto a control diet and euthanized at 110 postnatal days. Fat depots were weighed to estimate the total AT. Serum glucose, triglyceride, leptin, insulin, and the insulin resistance index (HOMA-IR) were determined. Adipocyte size and adipogenic gene expression were examined in retroperitoneal fat. Body weight, retroperitoneal AT and adipogenesis differed between male and female F1Cs. Retroperitoneal AT, glucose, triglyceride, insulin, HOMA-IR and leptin were higher in male and female F1MO vs. F1C. Small adipocytes were reduced in F1MO females and absent in F1MO males; large adipocytes were increased in F1MO males and females vs. F1C. Wnt, PI3K-Akt, and insulin signaling pathways in F1MO males and Egr2 in F1MO females were downregulated vs. F1C. MO induced metabolic dysfunction in F1 through different sex dimorphism mechanisms, including the decreased expression of pro-adipogenic genes and reduced insulin signaling in males and lipid mobilization-related genes in females.


Asunto(s)
Leptina , Obesidad Materna , Humanos , Ratas , Femenino , Animales , Masculino , Embarazo , Madres , Fosfatidilinositol 3-Quinasas/metabolismo , Ratas Wistar , Obesidad/etiología , Obesidad/metabolismo , Obesidad Materna/metabolismo , Glucosa/metabolismo , Insulina , Dieta Alta en Grasa/efectos adversos , Triglicéridos , Tejido Adiposo/metabolismo
10.
J Nutr Biochem ; 117: 109350, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37044135

RESUMEN

Several studies have shown the beneficial effects of (-)-epicatechin (Epi) in metabolic profile and that this flavanol is a biased ligand of the apelin receptor. The apelinergic system is expressed in adipocytes and has been related to obesity and metabolic disorders. The study aim was to evaluate the effect of Epi on apelin, on its receptor and on proteins involved in lipolysis, lipogenesis, and adipogenesis in the retroperitoneal adipose tissue of male rats descended from obese mothers. We evaluated the effect of Epi in the retroperitoneal adipose tissue of four groups of male offspring, analyzing mRNA expression and protein levels of apelin and its Apj receptor. We also analyzed, by Western Blot, the levels of AMPKα, ACC, C/EBPα, ATGL, Fas, and FABP4 of the AP2 proteins. Epi significantly elevated apelin mRNA expression and protein levels as well as its Apj receptor. Besides, the flavanol significantly promoted AMPKα phosphorylation with the concomitant reduction of Fas, and the increase of the ATGL protein. In contrast, there was an increase in the inactive phosphorylated form of ACC and a decrease in the phosphorylated active form of C/EBPα. Similarly, Epi treatment induced a reduction in the fatty acid-binding protein 4 in the C+Epi and MO+Epi groups. In conclusion, Epi increases the expression of the apelinergic system and the active phosphorylated form of AMPKα; likewise, it modifies the expression level or active form of proteins involved in lipolysis, lipogenesis and adipogenesis in the retroperitoneal adipose tissue of male offspring of obese mothers.


Asunto(s)
Catequina , Obesidad Materna , Ratas , Masculino , Femenino , Animales , Humanos , Embarazo , Receptores de Apelina/metabolismo , Apelina/metabolismo , Metabolismo de los Lípidos , Catequina/farmacología , Obesidad/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
11.
Nutrients ; 15(5)2023 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-36904238

RESUMEN

The steroids corticosterone and dehydroepiandrosterone (DHEA) perform multiple life course functions. Rodent life-course circulating corticosterone and DHEA trajectories are unknown. We studied life course basal corticosterone and DHEA in offspring of rats fed protein-restricted (10% protein, R) or control (20% protein, C), pregnancy diet first letter, and/or lactation second letter, producing four offspring groups-CC, RR, CR, and RC. We hypothesize that 1. maternal diet programs are sexually dimorphic, offspring life course steroid concentrations, and 2. an aging-related steroid will fall. Both changes differ with the plastic developmental period offspring experienced R, fetal life or postnatally, pre-weaning. Corticosterone was measured by radioimmunoassay and DHEA by ELISA. Steroid trajectories were evaluated by quadratic analysis. Female corticosterone was higher than male in all groups. Male and female corticosterone were highest in RR, peaked at 450 days, and fell thereafter. DHEA declined with aging in all-male groups. DHEA: corticosterone fell in three male groups but increased in all-female groups with age. In conclusion, life course and sexually dimorphic steroid developmental programming-aging interactions may explain differences in steroid studies at different life stages and between colonies experiencing different early-life programming. These data support our hypotheses of sex and programming influences and aging-related fall in rat life course serum steroids. Life course studies should address developmental programming-aging interactions.


Asunto(s)
Corticosterona , Dieta con Restricción de Proteínas , Embarazo , Ratas , Animales , Femenino , Masculino , Ratas Wistar , Envejecimiento/metabolismo , Deshidroepiandrosterona
12.
Biomedicines ; 11(3)2023 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-36979631

RESUMEN

Robust data in animals show that sucralose intake during gestation can predispose the offspring to weight gain, metabolic disturbances, and low-grade systemic inflammation; however, concluding information remains elusive in humans. In this cross-sectional, prospective study, we examined the birth weight, glucose and insulin cord blood levels, monocyte subsets, and inflammatory cytokine profile in 292 neonates at term from mothers with light sucralose ingestion (LSI) of less than 60 mg sucralose/week or heavy sucralose intake (HSI) of more than 36 mg sucralose/day during pregnancy. Mothers in the LSI (n = 205) or HSI (n = 87) groups showed no differences in age, pregestational body mass index, blood pressure, and glucose tolerance. Although there were no differences in glucose, infants from HSI mothers displayed significant increases in birth weight and insulin compared to newborns from LSI mothers. Newborns from HSI mothers showed a substantial increase in the percentage of inflammatory nonclassical monocytes compared to neonates from LSI mothers. Umbilical cord tissue of infants from HSI mothers exhibited higher IL-1 beta and TNF-alpha with lower IL-10 expression than that found in newborns from LSI mothers. Present results demonstrate that heavy sucralose ingestion during pregnancy affects neonates' anthropometric, metabolic, and inflammatory features.

14.
Reprod Sci ; 30(8): 2416-2428, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-36849856

RESUMEN

We used uncontrolled maternal diabetes as a model to provoke fetal growth restriction in the female in the first generation (F1) and to evaluate reproductive outcomes and the possible changes in metabolic systems during pregnancy, as well as the repercussions at birth in the second generation (F2). For this, nondiabetic and streptozotocin-induced severely diabetic Sprague-Dawley rats were mated to obtain female pups (F1), which were classified as adequate (AGA) or small (SGA) for gestational weight. Afterward, we composed two groups: F1 AGA from nondiabetic dams (Control) and F1 SGA from severely diabetic dams (Restricted) (n minimum = 10 animals/groups). At adulthood, these rats were submitted to the oral glucose tolerance test, mated, and at day 17 of pregnancy, blood samples were collected to determine glucose and insulin levels for assessment of insulin resistance. At the end of the pregnancy, the blood and liver samples were collected to evaluate redox status markers, and reproductive, fetal, and placental outcomes were analyzed. Maternal diabetes was responsible for increased SGA rates and a lower percentage of AGA fetuses (F1 generation). The restricted female pups from severely diabetic dams presented rapid neonatal catch-up growth, glucose intolerance, and insulin resistance status before and during pregnancy. At term pregnancy of F1 generation, oxidative stress status was observed in the maternal liver and blood samples. In addition, their offspring (F2 generation) had lower fetal weight and placental efficiency, regardless of gender, which caused fetal growth restriction and confirmed the fetal programming influence.


Asunto(s)
Diabetes Gestacional , Resistencia a la Insulina , Humanos , Ratas , Embarazo , Animales , Femenino , Placenta/metabolismo , Ratas Wistar , Retardo del Crecimiento Fetal/etiología , Retardo del Crecimiento Fetal/metabolismo , Ratas Sprague-Dawley , Diabetes Gestacional/metabolismo , Glucemia/metabolismo
15.
Br J Nutr ; 130(5): 783-792, 2023 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-36412162

RESUMEN

Obese mothers' offspring develop obesity and metabolic alterations in adulthood. Poor postnatal dietary patterns also contribute to obesity and its comorbidities. We aimed to determine whether in obese mothers' offspring an adverse postnatal environment, such as high-fat diet (HFD) consumption (second hit) exacerbates body fat accumulation, metabolic alterations and adipocyte size distribution. Female Wistar rats ate chow (C-5 %-fat) or HFD (maternal obesity (MO)-25 %-fat) from weaning until the end of lactation. Male offspring were weaned on either control (C/C and MO/C, maternal diet/offspring diet) or HFD (C/HF and MO/HF) diet. At 110 postnatal days, offspring were killed. Fat depots were excised to estimate adiposity index (AI). Serum glucose, triglyceride, leptin, insulin, insulin resistance index (HOMA-IR), corticosterone and dehydroepiandrosterone (DHEA) were determined. Adipocyte size distribution was evaluated in retroperitoneal fat. Body weight was similar in C/C and MO/C but higher in C/HF and MO/HF. AI, leptin, insulin and HOMA-IR were higher in MO/C and C/HF v. C/C but lower than MO/HF. Glucose increased in MO/HF v. MO/C. C/HF and MO/C had higher triglyceride and corticosterone than C/C, but lower corticosterone than MO/HF. DHEA and the DHEA/corticosterone ratio were lower in C/HF and MO/C v. C/C, but higher than MO/HF. Small adipocyte proportion decreased while large adipocyte proportions increased in MO/C and C/HF v. C/C and exacerbated in MO/HF v. C/HF. Postnatal consumption of a HFD by the offspring of obese mothers exacerbates body fat accumulation as well as the decrease of small and the increase of large adipocytes, which leads to larger metabolic abnormalities.


Asunto(s)
Leptina , Efectos Tardíos de la Exposición Prenatal , Humanos , Ratas , Femenino , Animales , Masculino , Embarazo , Dieta Alta en Grasa/efectos adversos , Madres , Corticosterona/metabolismo , Ratas Wistar , Fenómenos Fisiologicos Nutricionales Maternos , Obesidad/etiología , Obesidad/metabolismo , Tejido Adiposo/metabolismo , Peso Corporal , Glucosa/metabolismo , Triglicéridos/metabolismo , Hipertrofia/metabolismo , Insulina/metabolismo , Deshidroepiandrosterona/metabolismo
16.
Int J Mol Sci ; 23(23)2022 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-36499183

RESUMEN

The Developmental Origins of Health and Disease (DOHaD) concept correlates early life exposure to stressor conditions with the increased incidence of non-communicable chronic diseases, including prostate cancer (PCa), throughout the life span. However, the molecular mechanisms involved in this process remain poorly understood. In this study, the deregulation of two miRNAs (rno-miR-18a-5p and rno-miR-345-3p) was described in the ventral prostate VP of old rats born to dams fed with a low protein diet (LPD) (6% protein in the diet) during gestational and lactational periods. Integrative analysis of the (VP) transcriptomic and proteomic data revealed changes in the expression profile of 14 identified predicted targets of these two DE miRNAs, which enriched terms related to post-translational protein modification, metabolism of proteins, protein processing in endoplasmic reticulum, phosphonate and phosphinate metabolism, the calnexin/calreticulin cycle, metabolic pathways, N-glycan trimming in the ER and the calnexin/calreticulin cycle, hedgehog ligand biogenesis, the ER-phagosome pathway, detoxification of reactive oxygen species, antigenprocessing-cross presentation, RAB geranylgeranylation, collagen formation, glutathione metabolism, the metabolism of xenobiotics by cytochrome P450, and platinum drug resistance. RT-qPCR validated the deregulation of the miR-18a-5p/P4HB (prolyl 4-hydroxylase subunit beta) network in the VP of older offspring as well as in the PNT-2 cells transfected with mimic miR-18a-5p. Functional in vitro studies revealed a potential modulation of estrogen receptor α (ESR1) by miR-18a-5p in PNT-2 cells, which was also confirmed in the VP of older offspring. An imbalance of the testosterone/estrogen ratio was also observed in the offspring rats born to dams fed with an LPD. In conclusion, deregulation of the miR-18a-5p/P4HB network can contribute to the developmental origins of prostate cancer in maternally malnourished offspring, highlighting the need for improving maternal healthcare during critical windows of vulnerability early in life.


Asunto(s)
MicroARNs , Neoplasias de la Próstata , Animales , Masculino , Ratas , Perfilación de la Expresión Génica , MicroARNs/genética , MicroARNs/metabolismo , Neoplasias de la Próstata/genética , Proteómica , Transcriptoma
18.
Antioxidants (Basel) ; 11(10)2022 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-36290594

RESUMEN

Maternal obesity (MO) causes maternal and fetal oxidative stress (OS) and metabolic dysfunction. We investigated whether supplementing obese mothers with resveratrol improves maternal metabolic alterations and reduces OS in the placenta and maternal and fetal liver. From weaning through pregnancy female Wistar rats ate chow (C) or a high-fat diet (MO). One month before mating until 19 days' gestation (dG), half the rats received 20 mg resveratrol/kg/d orally (Cres and MOres). At 19dG, maternal body weight, retroperitoneal fat adipocyte size, metabolic parameters, and OS biomarkers in the placenta and liver were determined. MO mothers showed higher body weight, triglycerides and leptin serum concentrations, insulin resistance (IR), decreased small and increased large adipocytes, liver fat accumulation, and hepatic upregulation of genes related to IR and inflammatory processes. Placenta, maternal and fetal liver OS biomarkers were augmented in MO. MOres mothers showed more small and fewer large adipocytes, lower triglycerides serum concentrations, IR and liver fat accumulation, downregulation of genes related to IR and inflammatory processes, and lowered OS in mothers, placentas, and female fetal liver. Maternal resveratrol supplementation in obese rats improves maternal metabolism and reduces placental and liver OS of mothers and fetuses in a sex-dependent manner.

19.
Dev Neurosci ; 44(6): 603-614, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36162387

RESUMEN

Maternal stress during pregnancy results in increased risk of developing psychiatric disorders in the offspring including anxiety, depression, schizophrenia, and autism. However, the mechanisms underlying this disease susceptibility remain largely to be determined. In this study, the involvement of the serotonin (5-HT) and kynurenine (KYN) pathways of tryptophan metabolism on the behavioral deficits induced by maternal stress during the late phase of gestation in mice was investigated. Adult offspring born to control or restraint-stressed dams were exposed to the elevated plus-maze and tail suspension tests. Metabolites of the KYN and 5-HT pathways were measured in the hippocampus and brainstem by ultra-performance liquid chromatography tandem mass spectrometry. Female, but not male, prenatally stressed (PNS) offspring displayed a depressive-like phenotype, mainly when in proestrus/diestrus, along with reduced hippocampal 5-HT levels and high 5-HT turnover rate in the hippocampus and brainstem. In contrast, male PNS mice showed enhanced anxiety-like behaviors and higher hippocampal and brainstem quinolinic acid levels compared to male offspring born to nonstressed dams. These results indicate that maternal stress affects the behavior and brain metabolism of tryptophan in the offspring in a sex-dependent manner and suggest that alterations in both the 5-HT and KYN pathways may underlie the emotional dysfunctions observed in individuals exposed to stress during in utero development.


Asunto(s)
Quinurenina , Triptófano , Embarazo , Ratones , Animales , Femenino , Quinurenina/metabolismo , Triptófano/metabolismo , Serotonina/metabolismo , Ansiedad/metabolismo , Conducta Animal
20.
Front Cell Dev Biol ; 10: 892315, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36072345

RESUMEN

Maternal obesity (MO) induces negative consequences in the offspring development. Adiposity phenotype is associated with maternal diet at early pregnancy and DNA methylation marks in the RXRα promotor at birth. Glucocorticoids play an important role in the regulation of metabolism through the activation of nuclear hormone receptors such as the RXRα protein. The aim of the study was to analyze steroid hormone changes at the end of pregnancy in the obese mother and RXRα gene methylation in the umbilical cord. For this purpose, in a well-established MO model, female Wistar rats were fed either standard chow (controls: C) or high-fat obesogenic diet (MO) before and during pregnancy to evaluate at 19 days of gestation (19 dG): 1) maternal concentration of circulating steroid hormones in MO and C groups, 2) maternal and fetal weights, 3) analysis of correlation between hormones concentration and maternal and fetal weights, 4) DNA methylation status of a single locus of RXRα gene near the early growth response (EGR-1) protein DNA binding site, and 5) RXRα mRNA and protein expressions in umbilical cords. Our results demonstrate that at 19 dG, MO body weight before and during pregnancy was higher than C; MO progesterone and corticosterone serum concentrations were higher and estradiol lower than C. There were not differences in fetal weight between male and female per group, therefore averaged data was used; MO fetal weight was lower than C. Positive correlations were found between progesterone and corticosterone with maternal weight, and estradiol with fetal weight, while negative correlation was observed between corticosterone and fetal weight. Additionally, male umbilical cords from MO were hypermethylated in RXRα gene compared to male C group, without differences in the female groups; mRNA and protein expression of RXRα were decreased in F1 male but not in female MO compared to C. In conclusion, MO results in dysregulation of circulating steroid hormones of the obese mothers and low fetal weight in the F1, modifying DNA methylation of RXRα gene as well as RXRα mRNA and protein expression in the umbilical cord in a sex-dependent manner.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...