Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Neuroendocrinology ; 113(5): 549-562, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36580896

RESUMEN

INTRODUCTION: Obesity is associated with impaired learning, but the mechanisms underlying this cognitive dysfunction are poorly understood. Moreover, whether obesity-induced learning deficits show sexual dimorphism remains controversial. Females are believed to be protected from cognitive decline by oestrogens. These hormones enhance the expression of tryptophan hydroxylase 2, the rate-limiting enzyme in the transformation of tryptophan (Trp) into serotonin which plays a significant role in learning and memory. However, several learning-regulating compounds also arise from Trp metabolism through the kynurenine pathway (KP), including kynurenic acid (KA), xanthurenic acid (XA), and NAD+. The present study aimed to determine the involvement of the KP of Trp metabolism in the regulation of learning in control and obese female rats. METHODS: The learning capabilities of control and obese rats were evaluated using the novel object recognition test. Trp and Trp-derived metabolites were quantified in the hippocampus and frontal cortex by ultra-performance liquid chromatography-tandem mass spectrometry. RESULTS: Control rats in proestrus/oestrous performed better than their control mates in metestrus/dioestrus. Likewise, while control and obese rats in dioestrus/metestrus did not show differences in learning, obese rats in proestrus/oestrous displayed decreased memory capacity along with decreased Trp concentration and reduced KA, XA, and NAD+ production in the hippocampus. These neurochemical alterations were associated with impaired expression of mRNAs coding for key enzymes of the KP. CONCLUSION: The results presented here indicate that the deleterious effects of obesity on learning are closely related to the oestrous cycle and associated with an impairment of the KP of Trp metabolism.


Asunto(s)
Quinurenina , NAD , Femenino , Ratas , Animales , Quinurenina/metabolismo , NAD/metabolismo , Triptófano/metabolismo , Encéfalo/metabolismo , Ácido Quinurénico/metabolismo , Trastornos de la Memoria , Obesidad/metabolismo
2.
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
3.
Nutr Neurosci ; 25(10): 2011-2022, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33926365

RESUMEN

AIM: Individuals undernourished in utero or during early life are at high risk of developing obesity and metabolic disorders and show an increased preference for consuming sugary and fatty food. This study aimed at determining whether impaired taste detection and signalling in the lingual epithelium and the brain might contribute to this altered pattern of food intake. METHODS: The preference for feeding fat and sweet food and the expression in circumvallate papillae and hypothalamus of genes coding for sweet and fat receptors and transducing pathways were evaluated in adult rats born to control or calorie-restricted dams. Expression in the hypothalamus and the brain's reward system of genes involved in the homeostatic and hedonic control of food intake was also determined. RESULTS: Male and female undernourished animals exhibited increased expression in taste papillae and hypothalamus of T1R1, T1R2, CD36, gustducin, TRMP5 and PLC-ß2 genes, all of which modulate sweet and fat detection and intracellular signalling. However, the severity of the effect was greater in females than in males. Moreover, male, but not female, undernourished rats consumed more standard and sweetened food than their control counterparts and presented increased hypothalamic AgRP and NPY mRNAs levels together with enhanced dopamine transporter and dopamine receptor D2 expression in the ventral tegmental area. CONCLUSIONS: Maternal undernutrition induces sex-specific changes in food preferences and gene expression in taste papillae, hypothalamus and brain reward regions. The gene expression alterations in the male offspring are in line with their preference for consuming sugary and fatty food.


Asunto(s)
Desnutrición , Gusto , Proteína Relacionada con Agouti/metabolismo , Animales , Antígenos CD36/genética , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/genética , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Femenino , Hipotálamo/metabolismo , Masculino , Desnutrición/metabolismo , Ratas , Receptores Dopaminérgicos/metabolismo
4.
Int J Obes (Lond) ; 43(6): 1231-1243, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30568270

RESUMEN

BACKGROUND/OBJECTIVES: Maternal obesity is associated with increased risk of obesity and other symptoms of the metabolic syndrome in the offspring. Nevertheless, the molecular mechanisms and cellular factors underlying this enhanced disease susceptibility remain to be determined. Here, we aimed at identifying changes in plasma lipids in offspring of obese mothers that might underpin, and serve as early biomarkers of, their enhanced metabolic disease risk. SUBJECTS/METHODS: We performed a longitudinal lipidomic profiling in plasma samples from normal weight, overweight, and obese pregnant women and their children that participated in the Prenatal Omega-3 Fatty Acid Supplementation, Growth, and Development trial conducted in Mexico. At recruitment women were aged between 18 and 35 years and in week 18-22 of pregnancy. Blood samples were collected at term delivery by venipuncture from mothers and from the umbilical cord of their newborns and from the same infants at 4 years old under non-fasting conditions. Lipidomic profiling was done using ultra-performance liquid chromatography high-resolution mass spectrometry. RESULTS: Analysis of the lipidomic data showed that overweight and obese mothers exhibited a significant reduction in the total abundance of ceramides (Cer) in plasma, mainly of Cer (d18:1/20:0), Cer (d18:1/22:0), Cer (d18:1/23:0), and Cer (d18:1/24:0), compared with mothers of normal body weight. This reduction was confirmed by the direct quantification of these and other ceramide species. Similar quantitative differences in the plasma concentration of Cer (d18:1/22:0), Cer (d18:1/23:0), and Cer (d18:1/24:0), were also found between 4-year-old children of overweight and obese mothers compared with children of mothers of normal body weight. Noteworthy, children exhibited equal daily amounts of energy and food intake independently of the BMI of their mothers. CONCLUSIONS: Maternal obesity results in long-lasting changes in plasma ceramides in the offspring suggesting that these lipids might be used as early predictors of metabolic disease risk due to maternal obesity.


Asunto(s)
Ceramidas/sangre , Lipidómica , Síndrome Metabólico/sangre , Obesidad Materna/sangre , Obesidad Infantil/sangre , Adulto , Biomarcadores/sangre , Preescolar , Susceptibilidad a Enfermedades , Femenino , Humanos , Peso Corporal Ideal , Lactante , Recién Nacido , Estudios Longitudinales , Masculino , Síndrome Metabólico/etiología , Síndrome Metabólico/fisiopatología , Obesidad Materna/complicaciones , Obesidad Materna/fisiopatología , Sobrepeso/sangre , Obesidad Infantil/etiología , Obesidad Infantil/fisiopatología , Embarazo
5.
J Neurochem ; 140(1): 68-81, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27778340

RESUMEN

Early malnutrition is a risk factor for depression and schizophrenia. Since the offspring of malnourished dams exhibit increased brain levels of serotonin (5-HT), a tryptophan-derived neurotransmitter involved in the pathophysiology of these mental disorders, it is believed that the deleterious effects of early malnutrition on brain function are due in large part to altered serotoninergic neurotransmission resulting from impaired tryptophan (Trp) metabolism. However, tryptophan is also metabolized through the kynurenine (KYN) pathway yielding several neuroactive compounds including kynurenic (KA), quinolinic (QA) and xanthurenic (XA) acids. Nevertheless, the impact of perinatal malnutrition on brain kynurenine pathway metabolism has not been examined to date. Here, we used ultra-performance liquid chromatography-tandem mass spectrometry for the simultaneous quantification of tryptophan and a set of seven compounds spanning its metabolism through the serotonin and kynurenine pathways, in the brain of embryos and adult offspring of rat dams fed a protein-restricted (PR) diet. Protein-restricted embryos showed reduced brain levels of Trp, serotonin and KA, but not of KYN, XA, or QA. In contrast, PR adult rats exhibited enhanced levels of Trp in the brainstem and cortex along with increased concentrations of 5-HT, kynurenine and XA. The levels of XA and KA were also increased in the hippocampus of adult PR rats. These results show that early protein deficiency induces selective and long-lasting changes in brain kynurenine metabolism. Given the regulatory role of KYN pathway metabolites on brain development and function, these changes might contribute to the risk of developing psychiatric disorders induced by early malnutrition.


Asunto(s)
Encéfalo/metabolismo , Ácido Quinurénico/metabolismo , Quinurenina/metabolismo , Lactancia/metabolismo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Deficiencia de Proteína/metabolismo , Factores de Edad , Animales , Encéfalo/crecimiento & desarrollo , Proteínas en la Dieta , Femenino , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal/etiología , Deficiencia de Proteína/complicaciones , Ratas , Ratas Wistar
6.
Endocr Res ; 42(3): 252-259, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28318332

RESUMEN

PURPOSE: The aim of this study was to investigate the possible relationship among insulin resistance (IR), endothelial dysfunction, and alteration of adipokines in Mexican obese adolescents and their association with metabolic syndrome (MetS). MATERIALS AND METHODS: Two hundred and twenty-seven adolescents were classified according to the body mass index (BMI) (control: N=104; obese: N=123) and homeostasis model of the assessment-insulin resistance index (HOMA-IR) (obese with IR: N=65). The circulating concentrations of leptin, adiponectin, soluble intercellular adhesion molecule-1 (sICAM-1), and IR were determined by standard methods. RESULTS: The obese adolescents with IR presented increased presence of MetS and higher circulating concentrations in sICAM-1 in comparison with the obese subjects without IR. The lowest concentrations of adiponectin were observed in the obese with IR. In multivariate linear regression models, sICAM-1 along with triglycerides, total cholesterol, and waist circumference was strongly associated with HOMA-IR (R2=0.457, P=0.008). Similarly, after adjustment for age, BMI-SDS, lipids, and adipokines, HOMA-IR remained associated with sICAM-1 (R2=0.372, P=0.008). BMI-SDS was mildly associated with leptin (R2=0.176, P=0.002) and the waist circumference was mild and independent determinant of adiponectin (R2=0.136, P=0.007). CONCLUSIONS: Our findings demonstrated that the obese adolescents, particularly the obese subjects with IR exhibited increased presence of MetS, abnormality of adipokines, and endothelial dysfunction. The significant interaction between IR and endothelial dysfunction may suggest a novel therapeutic approach to prevent or delay systemic IR and the genesis of cardiovascular diseases in obese patients.


Asunto(s)
Adipoquinas/sangre , Endotelio Vascular/fisiopatología , Resistencia a la Insulina/fisiología , Síndrome Metabólico/sangre , Obesidad Infantil/sangre , Adolescente , Niño , Comorbilidad , Femenino , Humanos , Masculino , Síndrome Metabólico/epidemiología , Síndrome Metabólico/fisiopatología , México/epidemiología , Obesidad Infantil/epidemiología , Obesidad Infantil/fisiopatología
7.
Int J Cancer ; 139(8): 1873-82, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27270969

RESUMEN

Breast cancer is a global public health problem and accumulating evidence indicates early-life exposures as relevant factors in the disease risk determination. Recent studies have shown that paternal nutrition can influence offspring health including breast cancer risk. Selenium is a micronutrient with essential role in central aspects of embryogenesis, male fertility and cancer and that has been extensively studied as a chemopreventive agent in several breast cancer experimental models. Thus, we designed an animal study to evaluate whether paternal selenium deficiency or supplementation during preconception could affect the female offspring mammary gland development and breast cancer susceptibility. Male Sprague-Dawley rats were fed AIN93-G diet containing 0.15 ppm (control diet), 0.05 ppm (deficient diet) or 1 ppm (supplemented diet) of selenium for 9 weeks and mated with control female rats. Mammary carcinogenesis was induced with 7,12-dimethylbenz[a]anthracene (DMBA) in their female offspring. Paternal selenium deficiency increased the number of terminal end buds, epithelial elongation and cell proliferation in the mammary gland of the female rat offspring and these effects were associated with higher susceptibility to DMBA-induced mammary tumors (increased incidence and higher grade tumors). On the other hand, paternal selenium supplementation did not influence any of these parameters. These results highlight the importance of father's nutrition including selenium status as a relevant factor affecting daughter's breast cancer risk and paternal preconception as a potential developmental stage to start disease preventive strategies.


Asunto(s)
Neoplasias Mamarias Experimentales/etiología , Selenio/administración & dosificación , Selenio/deficiencia , 9,10-Dimetil-1,2-benzantraceno , Animales , Carcinogénesis , Suplementos Dietéticos , Femenino , Masculino , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/inducido químicamente , Neoplasias Mamarias Experimentales/patología , Ratas , Ratas Sprague-Dawley
8.
Br J Nutr ; 112(3): 328-37, 2014 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-24823946

RESUMEN

Skeletal muscle exhibits a remarkable flexibility in the usage of fuel in response to the nutrient intake and energy demands of the organism. In fact, increased physical activity and fasting trigger a transcriptional programme in skeletal muscle cells leading to a switch from carbohydrate to lipid oxidation. Impaired metabolic flexibility has been reported to be associated with obesity and type 2 diabetes, but it is not known whether the disability to adapt to metabolic demands is a cause or a consequence of these pathological conditions. Inasmuch as a poor nutritional environment during early life is a predisposing factor for the development of metabolic diseases in adulthood, in the present study, we aimed to determine the long-term effects of maternal malnutrition on the metabolic flexibility of offspring skeletal muscle. To this end, the transcriptional responses of the soleus and extensor digitorum longus muscles to fasting were evaluated in adult rats born to dams fed a control (17 % protein) or a low-protein (8 % protein, protein restricted (PR)) diet throughout pregnancy and lactation. With the exception of reduced body weight and reduced plasma concentrations of TAG, PR rats exhibited a metabolic profile that was the same as that of the control rats. In the fed state, PR rats exhibited an enhanced expression of key regulatory genes of fatty acid oxidation including CPT1a, PGC-1α, UCP3 and PPARα and an impaired expression of genes that increase the capacity for fat oxidation in response to fasting. These results suggest that impaired metabolic inflexibility precedes and may contribute to the development of metabolic disorders associated with early malnutrition.


Asunto(s)
Dieta con Restricción de Proteínas/efectos adversos , Músculo Esquelético/metabolismo , Efectos Tardíos de la Exposición Prenatal , Transcripción Genética , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Metabolismo de los Hidratos de Carbono , Carnitina O-Palmitoiltransferasa/genética , Metabolismo Energético , Activación Enzimática , Ayuno , Ácidos Grasos/metabolismo , Femenino , Expresión Génica , Canales Iónicos/genética , Lactancia , Metabolismo de los Lípidos , Masculino , Metaboloma , Proteínas Mitocondriales/genética , Oxidación-Reducción , PPAR alfa/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Fenotipo , Embarazo , Ratas , Ratas Wistar , Factores de Transcripción/genética , Proteína Desacopladora 3
9.
Int J Health Plann Manage ; 34(1): e713-e725, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30350426

RESUMEN

Maternal obesity is one of the main public health problems at a world level. It is a multifactorial disease with multiple causes, and few studies exist on its dietary patterns, physical activity and social determinants. This work aims to identify determinants of maternal obesity in a middle income country. Research is based on a prospective cohort design. Data were collected using questionnaires applied to pregnant women. Three dietary patterns were identified, and only half of the women carry out physical activity. The regression analysis showed an association between overweight/obesity and the following variables: age 25 to 29 years old (3.8; CI 1.6-9.0), 30 to 34 years old (3.7; CI 1.2-11.6); health problems during pregnancy (2.1; CI 1.0-4.1); socio-economic income (1.73; CI 1.54-2.05); hypertension (2.7; CI 1.4-4.5); mild food insecurity (1.9; CI 1.0-3.8); moderate insecurity (3.7; CI 0.92-15.4); refined food dietary pattern (.76; CI.61-.95). The risk of increasing BMI during pregnancy mainly depends on socioeconomic and demographic variables such as age, educational level, income, food insecurity, and dietary pattern. This study's results could be used as evidences for the revision, planning, and adjustment of interventions for the prevention and management of maternal obesity, as a part of the national strategies against overweight and obesity.


Asunto(s)
Países en Desarrollo/estadística & datos numéricos , Dieta/efectos adversos , Ejercicio Físico , Obesidad Materna/etiología , Adolescente , Adulto , Dieta/estadística & datos numéricos , Femenino , Humanos , México/epidemiología , Obesidad Materna/epidemiología , Embarazo , Estudios Prospectivos , Factores de Riesgo , Determinantes Sociales de la Salud , Factores Socioeconómicos , Encuestas y Cuestionarios , Adulto Joven
10.
Acta Physiol (Oxf) ; 240(3): e14090, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38230587

RESUMEN

AIM: Offspring of obese mothers are at high risk of developing metabolic syndrome and cognitive disabilities. Impaired metabolism has also been reported in the offspring of obese fathers. However, whether brain function can also be affected by paternal obesity has barely been examined. This study aimed to characterize the learning deficits resulting from paternal obesity versus those induced by maternal obesity and to identify the underlying mechanisms. METHODS: Founder control and obese female and male Wistar rats were mated to constitute three first-generation (F1) experimental groups: control mother/control father, obese mother/control father, and obese father/control mother. All F1 animals were weaned onto standard chow and underwent a learning test at 4 months of age, after which several markers of glutamate-mediated synaptic plasticity together with the expression of miRNAs targeting glutamate receptors and the concentration of kynurenic and quinolinic acids were quantified in the hippocampus and frontal cortex. RESULTS: Maternal obesity induced a severe learning deficit by impairing memory encoding and memory consolidation. The offspring of obese fathers also showed reduced memory encoding but not impaired long-term memory formation. Memory deficits in offspring of obese fathers and obese mothers were associated with a down-regulation of genes encoding NMDA glutamate receptors subunits and several learning-related genes along with impaired expression of miR-296 and miR-146b and increased concentration of kynurenic acid. CONCLUSION: Paternal and maternal obesity impair offspring's learning abilities by affecting different processes of memory formation. These cognitive deficits are associated with epigenetic and neurochemical alterations leading to impaired glutamate-mediated synaptic plasticity.


Asunto(s)
MicroARNs , Obesidad Materna , Humanos , Adulto , Ratas , Femenino , Masculino , Embarazo , Animales , Obesidad Materna/complicaciones , Obesidad Materna/genética , Ratas Wistar , Obesidad , Padre , Encéfalo , Receptores de Glutamato/genética , Glutamatos/genética , Epigénesis Genética
11.
Int J Tryptophan Res ; 15: 11786469221111116, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35846874

RESUMEN

In addition to be a primary risk factor for type 2 diabetes and cardiovascular disease, obesity is associated with learning disabilities. Here we examined whether a dysregulation of the kynurenine pathway (KP) of tryptophan (Trp) metabolism might underlie the learning deficits exhibited by obese individuals. The KP is initiated by the enzymatic conversion of Trp into kynurenine (KYN) by indoleamine 2,3-dioxygenase (IDO). KYN is further converted to several signaling molecules including quinolinic acid (QA) which has a negative impact on learning. Wistar rats were fed either standard chow or made obese by exposure to a free choice high-fat high-sugar (fcHFHS) diet. Their learning capacities were evaluated using a combination of the novel object recognition and the novel object location tasks, and the concentrations of Trp and KYN-derived metabolites in several brain regions determined by ultra-performance liquid chromatography-tandem mass spectrometry. Male, but not female, obese rats exhibited reduced learning capacity characterized by impaired encoding along with increased hippocampal concentrations of QA, Xanthurenic acid (XA), Nicotinamide (Nam), and oxidized Nicotinamide Adenine Dinucleotide (NAD+). In contrast, no differences were detected in the serum levels of Trp or KP metabolites. Moreover, obesity enhanced the expression in the hippocampus and frontal cortex of kynurenine monooxygenase (KMO), an enzyme involved in the production of QA from kynurenine. QA stimulates the glutamatergic system and its increased production leads to cognitive impairment. These results suggest that the deleterious effects of obesity on cognition are sex dependent and that altered KP metabolism might contribute to obesity-associated learning disabilities.

12.
Nutrients ; 13(9)2021 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-34578953

RESUMEN

Maternal supplementation during pregnancy with docosahexaenoic acid (DHA) is internationally recommended to avoid postpartum maternal depression in the mother and improve cognitive and neurological outcomes in the offspring. This study was aimed at determining whether this nutritional intervention, in the rat, protects the offspring against the development of obesity and its associated metabolic disorders. Pregnant Wistar rats received an extract of fish oil enriched in DHA or saline (SAL) as placebo by mouth from the beginning of gestation to the end of lactation. At weaning, pups were fed standard chow or a free-choice, high-fat, high-sugar (fc-HFHS) diet. Compared to animals fed standard chow, rats exposed to the fc-HFHS diet exhibited increased body weight, liver weight, body fat and leptin in serum independently of saline or DHA maternal supplementation. Nevertheless, maternal DHA supplementation prevented both the glucose intolerance and the rise in serum insulin resulting from consumption of the fc-HFHS diet. In addition, animals from the DHA-fc-HFHS diet group showed decreased hepatic triglyceride accumulation compared to SAL-fc-HFHS rats. The beneficial effects on glucose homeostasis declined with age in male rats. Yet, the preventive action against hepatic steatosis was still present in 6-month-old animals of both sexes and was associated with decreased hepatic expression of lipogenic genes. The results of the present work show that maternal DHA supplementation during pregnancy programs a healthy phenotype into the offspring that was protective against the deleterious effects of an obesogenic diet.


Asunto(s)
Fenómenos Fisiológicos Nutricionales de los Animales/efectos de los fármacos , Dieta Alta en Grasa/efectos adversos , Ácidos Docosahexaenoicos/farmacología , Hígado Graso/prevención & control , Lactancia , Animales , Dieta Alta en Grasa/métodos , Suplementos Dietéticos , Modelos Animales de Enfermedad , Ácidos Docosahexaenoicos/administración & dosificación , Hígado Graso/etiología , Femenino , Fenómenos Fisiologicos Nutricionales Maternos/efectos de los fármacos , Embarazo , Ratas , Ratas Wistar
13.
Nutrients ; 13(12)2021 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-34959795

RESUMEN

We investigated if supplementing obese mothers (MO) with docosahexaenoic acid (DHA) improves milk long-chain polyunsaturated fatty acid (LCPUFA) composition and offspring anxiety behavior. From weaning throughout pregnancy and lactation, female Wistar rats ate chow (C) or a high-fat diet (MO). One month before mating and through lactation, half the mothers received 400 mg DHA kg-1 d-1 orally (C+DHA or MO+DHA). Offspring ate C after weaning. Maternal weight, total body fat, milk hormones, and milk nutrient composition were determined. Pups' milk nutrient intake was evaluated, and behavioral anxiety tests were conducted. MO exhibited increased weight and total fat, and higher milk corticosterone, leptin, linoleic, and arachidonic acid (AA) concentrations, and less DHA content. MO male and female offspring had higher ω-6/ ω-3 milk consumption ratios. In the elevated plus maze, female but not male MO offspring exhibited more anxiety. MO+DHA mothers exhibited lower weight, total fat, milk leptin, and AA concentrations, and enhanced milk DHA. MO+DHA offspring had a lower ω-6/ω-3 milk intake ratio and reduced anxiety vs. MO. DHA content was greater in C+DHA milk vs. C. Supplementing MO mothers with DHA improves milk composition, especially LCPUFA content and ω-6/ω-3 ratio reducing offspring anxiety in a sex-dependent manner.


Asunto(s)
Animales Recién Nacidos/psicología , Conducta Animal/efectos de los fármacos , Suplementos Dietéticos , Ácidos Docosahexaenoicos/administración & dosificación , Leche/química , Animales , Ansiedad/prevención & control , Ingestión de Alimentos/psicología , Ácidos Grasos Omega-3/análisis , Ácidos Grasos Omega-6/análisis , Ácidos Grasos Insaturados/análisis , Femenino , Lactancia , Masculino , Fenómenos Fisiologicos Nutricionales Maternos/efectos de los fármacos , Obesidad , Embarazo , Ratas , Ratas Wistar , Factores Sexuales
14.
Physiol Behav ; 96(3): 481-92, 2009 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-19100759

RESUMEN

Protein or calorie restriction during gestation and/or suckling induces hyperphagia and increases the susceptibility to develop obesity, glucose intolerance and hypertension in adulthood. The mechanisms by which early nutrient restriction affects the normal physiological regulation of feeding as well as to what extent the metabolic programming of hyperphagia contributes to the long-term risk of obesity and insulin resistance remain, however, to be determined. Here the temporal pattern of food intake and the behavioural satiety sequence were investigated in the offspring of Sprague-Dawley rats fed a control (C) or a low-protein (LP) diet throughout pregnancy and lactation. During the first two months of their post-natal life, protein-restricted animals exhibited hyperphagia characterized by a delayed appearance of satiety, an increase in meal size and reduced latency to eat. Protein-restricted pups also exhibited an enhanced expression of the orexigenic peptides Agouti-related protein and neuropeptide Y and decreased hypothalamic levels of the anorexigenic peptide pro-opiomelanocortin. At 8 months, LP rats still consumed larger meals than their control counterparts but they ingested daily the same amount of food as control offspring and exhibited enhanced abdominal fat and increased levels of triglycerides and fatty acids in serum. These observations indicate that the hyperphagia observed in young LP rats results from a decreased action of negative feedback signals critical to meal termination and an enhanced function of the positive signals that initiate and maintain eating. These results also suggest that perinatal malnutrition programmes obesity through a mechanism independent of its effects on feeding behaviour.


Asunto(s)
Regulación del Apetito/fisiología , Obesidad/fisiopatología , Efectos Tardíos de la Exposición Prenatal , Desnutrición Proteico-Calórica/fisiopatología , Saciedad/fisiología , Adaptación Fisiológica , Análisis de Varianza , Animales , Animales Recién Nacidos , Dieta con Restricción de Proteínas , Conducta Alimentaria/fisiología , Femenino , Hiperfagia/fisiopatología , Masculino , Desnutrición/fisiopatología , Embarazo , Ratas , Ratas Sprague-Dawley
15.
Eur J Neurosci ; 27(6): 1400-8, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18331342

RESUMEN

Early malnutrition has been associated with a high risk of developing obesity, diabetes and cardiovascular diseases in adulthood. In animals, poor perinatal nutrition produces hyperphagia and persistent increased levels of serotonin (5-HT) in the brain. Inasmuch as 5-HT is directly related to the negative regulation of food intake, here we have investigated whether the anorexic effects of 5-HT are altered by protein malnutrition. Pregnant Sprague-Dawley rats were fed ad libitum either a control (20% protein) or a low-protein (8% protein) diet throughout pregnancy and lactation. At weaning, pups received a standard diet and at 35 days their feeding behaviour was evaluated after the administration of DL-fenfluramine (DL-FEN), an anorexic compound that blocks the reuptake of 5-HT and stimulates its release. Male offspring born to protein-restricted dams exhibited significantly decreased body weight and hyperphagia compared with controls. DL-FEN dose-dependently reduced the 1 h chow intake at the onset of the dark cycle in both control and undernourished rats. However, the hypophagic effects of DL-FEN were significantly attenuated in animals submitted perinatally to protein restriction. The stimulatory action of DL-FEN on c-fos immunoreactivity within the paraventricular nucleus of the hypothalamus was also decreased in low-protein-fed rats. Further pharmacological analysis with selective 5-HT(1B) and 5-HT(2C) receptor agonist showed that the reduced anorexic effects of 5-HT in malnourished animals were coupled to a desensitization of 5-HT(1B) receptors. These observations indicate that the hyperphagia associated with metabolic programming is at least partially related to a reduced regulatory function of 5-HT on food intake.


Asunto(s)
Dieta con Restricción de Proteínas , Ingestión de Alimentos/fisiología , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Serotonina/fisiología , Animales , Animales Recién Nacidos , Peso Corporal/fisiología , Dieta con Restricción de Proteínas/métodos , Ingestión de Alimentos/efectos de los fármacos , Femenino , Hiperfagia/etiología , Hiperfagia/fisiopatología , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal/etiología , Ratas , Ratas Sprague-Dawley , Agonistas de Receptores de Serotonina/farmacología
16.
Rev Salud Publica (Bogota) ; 20(2): 245-253, 2018.
Artículo en Español | MEDLINE | ID: mdl-30570010

RESUMEN

OBJECTIVE: To identify and analyze the socioeconomic indicators of maternal obesity in Mexico and France. MATERIAL AND METHODS: Comparative study of two cohorts: EDEN (France) and NUTTSEA (Mexico). The study population consisted of women who requested prenatal consultation at week 24 of pregnancy. Data were collected using questionnaires and semi-structured interviews. The variables of interest were socioeconomic aspects, nutrition, anthropometry and food security. The quantitative analysis was performed using Stata and the qualitative analysis with Atlas-ti. RESULTS: In the EDEN cohort, 68.6% were aged 25-34 years, 73% had paid employment and 53% completed high school. In addition, 6.6% reported having difficulty accessing food and the mean gestational BMI was 23.23 ± 4.6. In the NUTTSEA cohort, 55% were in the age range 18-24 years, 15% reported having paid employment, 42% had completed secondary education, 32.1% presented a degree of food insecurity, and the mean BMI was 27.8 ± 4.8. CONCLUSIONS: The qualitative and quantitative results of both cohorts suggest that populations with greater socio-economic vulnerability are more prone to maternal obesity, which leads to determine guidelines on barriers and facilitators to strengthen programs to prevent it maternal obesity.


OBJETIVO: Identificar y analizar los indicadores socioeconómicos de obesidad materna en México y Francia. METODOLOGÍA: Estudio comparativo de dos cohortes EDEN sus siglas en francés (Etude des déterminants pré et post natals précoces de la santé et de développement de l'enfant) (Francia) y NUTTSEA (México). La población de estudio se conformó por mujeres que solicitaron consulta prenatal en semana 24 de gestación. Los datos fueron recolectados con cuestionarios y entrevistas semi-estructuradas. Las variables de interés fueron aspectos socioeconómicos, alimentación, antropometría y seguridad alimentaria. El análisis cuantitativo se realizó utilizando Stata y el análisis cualitativo con Atlas-ti. RESULTADOS: En cohorte EDEN el 68.6% resultó con edad de 25-34 años, el 73% tenían un empleo remunerado, el 53% preparatoria completa; el 6.6% refirió tener dificultad para acceder a los alimentos; la media del IMC gestacional fue de 23.23±4.6. En la cohorte NUTTSEA el 55% se encontraba en el rango de edad de 18-24 años; el 15% refirió contar con un empleo remunerado; el 42% de la población tenía secundaria completa; el 32.1% presentó un grado de inseguridad alimentaria; la media de IMC fue de 27.8±4.8. CONCLUSIONES: Los resultados cuali-cuantitativos de ambas cohortes sugieren que poblaciones con mayor vulnerabilidad socio-económica son más propensas a la obesidad materna, determinando directrices sobre barreras y facilitadores para fortalecer programas de prevención de la obesidad materna.


Asunto(s)
Obesidad/etiología , Complicaciones del Embarazo/etiología , Adolescente , Adulto , Femenino , Francia/epidemiología , Humanos , México/epidemiología , Obesidad/epidemiología , Embarazo , Complicaciones del Embarazo/epidemiología , Investigación Cualitativa , Factores de Riesgo , Factores Socioeconómicos , Adulto Joven
17.
FEBS Lett ; 580(22): 5371-6, 2006 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-16989820

RESUMEN

Gustatory papillae and associated taste buds receive and process chemical information from the environment. In mammals, their development takes place during the late phase of embryogenesis. However, the cellular factors that regulate the differentiation of taste papillae remain largely unknown. Here, we show by quantitative real time RT-PCR that both isoforms of tryptophan hydroxylase (TPH1 and TPH2), the first and rate limiting enzyme of serotonin (5-HT) synthesis, are expressed in developing circumvallate papillae. Immuno-staining experiments further indicated that TPH is localized both in gustatory fibers and in differentiated taste receptor cells. These results point to the synthesis of 5-HT in gustatory papillae, and allow one to hypothesize that the development of taste buds might be modulated by serotonin.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Regulación Enzimológica de la Expresión Génica/fisiología , Papilas Gustativas/embriología , Triptófano Hidroxilasa/biosíntesis , Animales , Diferenciación Celular/fisiología , Perfilación de la Expresión Génica/métodos , Isoenzimas/biosíntesis , Isoenzimas/genética , Ratones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Serotonina/biosíntesis , Papilas Gustativas/citología , Papilas Gustativas/enzimología , Triptófano Hidroxilasa/genética
18.
Sci Rep ; 6: 21275, 2016 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-26882991

RESUMEN

Numerous epidemiological studies indicate that malnutrition during in utero development and/or childhood induces long-lasting learning disabilities and enhanced susceptibility to develop psychiatric disorders. However, animal studies aimed to address this question have yielded inconsistent results due to the use of learning tasks involving negative or positive reinforces that interfere with the enduring changes in emotional reactivity and motivation produced by in utero and neonatal malnutrition. Consequently, the mechanisms underlying the learning deficits associated with malnutrition in early life remain unknown. Here we implemented a behavioural paradigm based on the combination of the novel object recognition and the novel object location tasks to define the impact of early protein-restriction on the behavioural, cellular and molecular basis of memory processing. Adult rats born to dams fed a low-protein diet during pregnancy and lactation, exhibited impaired encoding and consolidation of memory resulting from impaired pattern separation. This learning deficit was associated with reduced production of newly born hippocampal neurons and down regulation of BDNF gene expression. These data sustain the existence of a causal relationship between early malnutrition and impaired learning in adulthood and show that decreased adult neurogenesis is associated to the cognitive deficits induced by childhood exposure to poor nutrition.


Asunto(s)
Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/fisiopatología , Hipocampo/fisiología , Desnutrición/complicaciones , Memoria , Neurogénesis , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Dieta con Restricción de Proteínas/efectos adversos , Modelos Animales de Enfermedad , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Femenino , Expresión Génica , Discapacidades para el Aprendizaje/etiología , Discapacidades para el Aprendizaje/fisiopatología , Masculino , Ratas
19.
Behav Processes ; 125: 19-25, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26836391

RESUMEN

We tested the hypothesis that perinatal undernourishment is a factor for binge eating. At 52 days rats born from dams fed on 17% protein (Control) or 8% protein (Undernourished) were distributed into four groups, two of which continued to be fed ad libitum chow and two were submitted to three consecutive Restricted/Refeeding (R/R) cycles. According to the following schedule: Control Naïve (from mothers fed 17% protein/no restriction phase); Control Restricted (from mothers fed 17% protein/restriction phase); Undernourished Naïve (from mothers fed 8% protein/no restriction phase); and Undernourished Restricted (from mothers fed 8% protein/restriction phase). Each cycle consisted of a restriction phase (in the first four days 40% of the mean daily individual chow intake was offered for consumption), followed by a refeeding phase (4 days of chow ad libitum). After the three cycles, all animals were subjected to a feeding test (chow diet and palatable food ad libitum for 24h). During the feeding test, the Undernourished Restricted demonstrated rebound hyperphagia during 2, 4 and 6h. These results suggest the perinatal undernourishment cannot contribute to a binge eating phenotype.


Asunto(s)
Bulimia/etiología , Dieta con Restricción de Proteínas/efectos adversos , Ingestión de Alimentos , Conducta Alimentaria , Efectos Tardíos de la Exposición Prenatal , Animales , Peso Corporal , Femenino , Embarazo , Ratas
20.
Med Sci (Paris) ; 32(1): 85-92, 2016 Jan.
Artículo en Francés | MEDLINE | ID: mdl-26850612

RESUMEN

Eating behavior is strongly regulated by intrinsic physiological factors and largely influenced by the individual and cultural environments. Excessive food intake and sedentary lifestyle are the main reasons for the global epidemic of obesity. The influence of family background on eating habits makes no doubt but the fact that the nutritional, metabolic and hormonal status of the parents before conception, and of the mother during gestation and lactation, may influence the child's future eating behavior is an innovative concept that opens the way for preventive policies. In the last decades, research on human cohorts and animal models have targeted biological mechanisms (neuroanatomical, epigenetic) that give some clues on how eating behavior can be formatted by early nutrition and related sensory experience.


Asunto(s)
Conducta Alimentaria , Efectos Tardíos de la Exposición Prenatal , Fenómenos Fisiologicos de la Nutrición Prenatal , Sensación/fisiología , Adulto , Hijos Adultos , Animales , Niño , Conducta Alimentaria/fisiología , Conducta Alimentaria/psicología , Femenino , Humanos , Recién Nacido , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Efectos Tardíos de la Exposición Prenatal/psicología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA