Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 183
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Pediatr Int ; 66(1): e15724, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38156534

RESUMEN

BACKGROUND: This study investigated the prevalence of post COVID-19 condition (PCC) in Taiwanese children, explored common symptoms and behavioral manifestations in children with PCC, and examined potential associations with parental stress and family functioning. METHODS: A total of 116 children who had contracted COVID-19 and their parents participated. At 1 month after the confirmed COVID-19 infection (baseline), the parents were asked to complete a questionnaire about the post COVID-19 symptoms, the child behavior checklist (CBCL) based on the child's situation, also the impact event scale of COVID-19 (IES-C) and family APGAR based on the parents' own situation. Three months after the confirmed COVID-19 infection, parents were again asked to complete the same questionnaires as they did at baseline. RESULTS: Results showed that 44.8% of the children met the criteria for PCC, with the most common symptoms being sputum/nasal congestion, tiredness and decreased concentration. Children without PCC exhibited a significant decrease in CBCL scores during the 2-month follow-up, while no such decrease was observed in children with PCC. However, no significant correlations were found between the number of post COVID-19 symptoms, behavioral manifestations, parental stress levels, and family functioning. CONCLUSIONS: Children with PCC have a less favorable trajectory in improving their behavioral manifestations. Additionally, in the post-pandemic era, post COVID-19 symptoms in children are not necessarily correlated with parental stress and family functioning. Further research is needed to better understand the long-term implications of PCC in children and its impact on their mental well-being, as well as their families.


Asunto(s)
COVID-19 , Niño , Humanos , COVID-19/epidemiología , Padres , Conducta Infantil
2.
Int J Mol Sci ; 25(9)2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38731977

RESUMEN

Mesenchymal stem cells (MSCs) isolated from Wharton's jelly (WJ-MSCs) and adipose tissue (AD-MSCs) are alternative sources for bone marrow-derived MSCs. Owing to their multiple functions in angiogenesis, immune modulation, proliferation, migration, and nerve regeneration, MSC-derived exosomes can be applied in "cell-free cell therapy". Here, we investigated the functional protein components between the exosomes from WJ-MSCs and AD-MSCs to explain their distinct functions. Proteins of WJ-MSC and AD-MSC exosomes were collected and compared based on iTRAQ gel-free proteomics data. Results: In total, 1695 proteins were detected in exosomes. Of these, 315 were more abundant (>1.25-fold) in AD-MSC exosomes and 362 kept higher levels in WJ-MSC exosomes, including fibrinogen proteins. Pathway enrichment analysis suggested that WJ-MSC exosomes had higher potential for wound healing than AD-MSC exosomes. Therefore, we treated keratinocyte cells with exosomes and the recombinant protein of fibrinogen beta chain (FGB). It turned out that WJ-MSC exosomes better promoted keratinocyte growth and migration than AD-MSC exosomes. In addition, FGB treatment had similar results to WJ-MSC exosomes. The fact that WJ-MSC exosomes promoted keratinocyte growth and migration better than AD-MSC exosomes can be explained by their higher FGB abundance. Exploring the various components of AD-MSC and WJ-MSC exosomes can aid in their different clinical applications.


Asunto(s)
Movimiento Celular , Proliferación Celular , Exosomas , Queratinocitos , Células Madre Mesenquimatosas , Gelatina de Wharton , Exosomas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Humanos , Gelatina de Wharton/citología , Gelatina de Wharton/metabolismo , Queratinocitos/metabolismo , Queratinocitos/citología , Fibrinógeno/metabolismo , Proteómica/métodos , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Células Cultivadas , Cicatrización de Heridas , Proteoma/metabolismo
3.
Int J Mol Sci ; 24(17)2023 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-37686267

RESUMEN

Prenatal high-fat diet (HFD) or exposure to microplastics can affect the accumulation of liver fat in offspring. We sought to determine the effects of maternal HFD intake and microplastic exposure on fatty liver injury through oxidative stress in pups. Pregnant female Sprague-Dawley rats were randomly divided into maternal HFD (experimental group) or normal control diet (NCD; control group) groups with or without microplastic exposure. As a result, the following groups were established: HFD-L (HFD + microplastics, 5 µm, 100 µg/L), HFD-H (HFD + microplastics, 5 µm, 1000 µg/L), NCD-L (NCD + microplastics, 5 µm, 100 µg/L), and NCD-H (NCD + microplastics, 5 µm, 1000 µg/L). The pups were sacrificed on postnatal day 7 (PD7). Liver histology revealed increased hepatic lipid accumulation in pups in the HFD-L and HFD-H groups compared to those in the HFD, NCD-L, NCD-H, and NCD groups on PD7. Similarly, liver TUNEL staining and cellular apoptosis were found to increase in pups in the HFD-L and HFD-H groups compared to those in the HFD, NCD-L, NCD-H, and NCD groups. The expression levels of malondialdehyde, a lipid peroxidation marker, were high in the HFD, HFD-L, and HFD-H groups; however, the highest expression was observed in the HFD-H group (p < 0.05). The levels of glutathione peroxidase, an antioxidant enzyme, decreased in the HFD, HFD-L, and HFD-H groups (p < 0.05). Overall, oxidative stress with cellular apoptosis plays a vital role in liver injury in offspring after maternal intake of HFD and exposure to microplastic; such findings may shed light on future therapeutic strategies.


Asunto(s)
Dieta Alta en Grasa , Enfermedades no Transmisibles , Femenino , Masculino , Ratas , Embarazo , Animales , Ratas Sprague-Dawley , Dieta Alta en Grasa/efectos adversos , Microplásticos , Plásticos , Hígado , Estrés Oxidativo , Vitaminas
4.
Int J Mol Sci ; 23(15)2022 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-35955421

RESUMEN

Hypertension is an important public health challenge, affecting up to 30-50% of adults worldwide. Several epidemiological studies indicate that high blood pressure originates in fetal life-the so-called programming effect or developmental origin of hypertension. Iron-deficiency anemia has become one of the most prevalent nutritional problems globally. Previous animal experiments have shown that prenatal iron-deficiency anemia adversely affects offspring hypertension. However, the underlying mechanism remains unclear. We used a maternal low-iron diet Sprague Dawley rat model to study changes in blood pressure, the renal renin-angiotensin system, oxidative stress, inflammation, and sodium transporters in adult male offspring. Our study revealed that 16-week-old male offspring born to mothers with low dietary iron throughout pregnancy and the lactation period had (1) higher blood pressure, (2) increased renal cortex angiotensin II receptor type 1 and angiotensin-converting enzyme abundance, (3) decreased renal cortex angiotensin II receptor type 2 and MAS abundance, and (4) increased renal 8-hydroxy-2'-deoxyguanosine and interleukin-6 abundance. Improving the iron status of pregnant mothers could influence the development of hypertension in their offspring.


Asunto(s)
Anemia Ferropénica , Hipertensión , Deficiencias de Hierro , Efectos Tardíos de la Exposición Prenatal , Anemia Ferropénica/metabolismo , Animales , Presión Sanguínea , Femenino , Hipertensión/metabolismo , Hierro/metabolismo , Hierro de la Dieta/metabolismo , Riñón/metabolismo , Lactancia , Masculino , Fenómenos Fisiologicos Nutricionales Maternos , Estrés Oxidativo , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Angiotensina/metabolismo , Sistema Renina-Angiotensina
5.
Int J Mol Sci ; 23(14)2022 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-35887270

RESUMEN

Emerging evidence supports that hypertension can be programmed or reprogrammed by maternal nutrition. Maternal exposures during pregnancy, such as maternal nutrition or antibiotic use, could alter the offspring's gut microbiota. Short-chain fatty acids (SCFAs) are the major gut microbiota-derived metabolites. Acetate, the most dominant SCFA, has shown its antihypertensive effect. Limited information exists regarding whether maternal acetate supplementation can prevent maternal minocycline-induced hypertension in adult offspring. We exposed pregnant Sprague Dawley rats to normal diet (ND), minocycline (MI, 50 mg/kg/day), magnesium acetate (AC, 200 mmol/L in drinking water), and MI + AC from gestation to lactation period. At 12 weeks of age, four groups (n = 8/group) of male progeny were sacrificed. Maternal acetate supplementation protected adult offspring against minocycline-induced hypertension. Minocycline administration reduced plasma acetic acid level, which maternal acetate supplementation prevented. Additionally, acetate supplementation increased the protein level of SCFA receptor G protein-coupled receptor 41 in the offspring kidneys. Further, minocycline administration and acetate supplementation significantly altered gut microbiota composition. Maternal acetate supplementation protected minocycline-induced hypertension accompanying by the increases in genera Roseburia, Bifidobacterium, and Coprococcus. In sum, our results cast new light on targeting gut microbial metabolites as early interventions to prevent the development of hypertension, which could help alleviate the global burden of hypertension.


Asunto(s)
Hipertensión , Efectos Tardíos de la Exposición Prenatal , Acetatos/farmacología , Animales , Presión Sanguínea , Suplementos Dietéticos , Femenino , Humanos , Hipertensión/inducido químicamente , Hipertensión/tratamiento farmacológico , Hipertensión/metabolismo , Lactancia , Masculino , Exposición Materna/efectos adversos , Minociclina/farmacología , Embarazo , Efectos Tardíos de la Exposición Prenatal/prevención & control , Ratas , Ratas Sprague-Dawley
6.
Int J Mol Sci ; 23(22)2022 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-36430717

RESUMEN

Maternal high-fat (HF) diet exposure in utero may affect fetal development and cause metabolic problems throughout life due to lipid dysmetabolism and oxidative damage. Metformin has been suggested as a potential treatment for body weight reduction and nonalcoholic fatty liver disease, but its reprogramming effect on offspring is undetermined. This study assesses the effects of maternal metformin treatment on hepatic steatosis in offspring caused by maternal HF diet. Female rats were fed either a control or an HF diet before conception, with or without metformin treatment during gestation, and placenta and fetal liver tissues were collected. In another experiment, the offspring were fed a control diet until 120 d (adult stage). Metformin treatment during pregnancy ameliorates placental oxidative stress and enhances placental glucose transporter 1 (GLUT1), GLUT3, and GLUT4 expression levels through 5' adenosine monophosphate-activated protein kinase (AMPK) activation. Maternal metformin treatment was shown to reprogram maternal HF diet-induced changes in offspring fatty liver with the effects observed in adulthood as well. Further validation is required to develop maternal metformin therapy for clinical applications.


Asunto(s)
Metformina , Enfermedad del Hígado Graso no Alcohólico , Femenino , Ratas , Embarazo , Animales , Dieta Alta en Grasa/efectos adversos , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Placenta/metabolismo , Metformina/farmacología , Metformina/uso terapéutico , Metformina/metabolismo , Grasas de la Dieta/farmacología , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo
7.
J Pediatr ; 228: 58-65.e3, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32712283

RESUMEN

OBJECTIVES: To investigate the cardiovascular features and endothelium in neonates born to mothers with preeclampsia. STUDY DESIGN: In this combined observational cohort and case-control study, neonates born to mothers with normotension and mothers with preeclampsia were recruited at a neonatal intensive care unit of a tertiary medical center. Cardiovascular measurements by echocardiography and the clinical measures upon admission were analyzed. Vascular cell adhesion molecule-1 expression in umbilical arteries and in in vitro endothelial cell stimulation with plasma were examined. Continuous data were compared using nonparametric analysis, and their relationships were analyzed using linear regression. Binary logistic regression was performed in the model of adjustment of birth body weight and for multivariate analysis. RESULTS: In the cohort, almost all cardiovascular segments positively correlated to birth weight. Notably, neonates (n = 65) of mothers with preeclampsia had significantly larger coronary arteries at birth than neonates of mothers with normotension (n = 404) (median size of left main coronary artery 1.36 mm versus 1.08 mm, p <0.001; median size of right coronary artery, RCA 1.25 mm versus 1.0 mm, p <0.001). The size of the right coronary artery positively correlated to the maternal antepartum diastolic blood pressure (r = 0.298, P = .018) and was associated with in-hospital death (P < .001). Meanwhile, endothelial vascular cell adhesion molecule-1 expression was significantly increased in the umbilical arteries of the preeclamptic group and following preeclamptic cord-plasma stimulation. The latter also correlated with their relative coronary sizes. CONCLUSIONS: Neonates of mothers with preeclampsia had distinctive coronary dilatation at birth. Coronary size might be useful as a severity index of neonatal endothelial inflammation as a result of maternal preeclampsia.


Asunto(s)
Enfermedad de la Arteria Coronaria/etiología , Vasos Coronarios/fisiopatología , Endotelio Vascular/fisiopatología , Inflamación/diagnóstico , Preeclampsia/diagnóstico , Enfermedad de la Arteria Coronaria/diagnóstico , Enfermedad de la Arteria Coronaria/fisiopatología , Dilatación Patológica/diagnóstico , Dilatación Patológica/etiología , Dilatación Patológica/fisiopatología , Endotelio Vascular/diagnóstico por imagen , Femenino , Estudios de Seguimiento , Edad Gestacional , Humanos , Recién Nacido , Inflamación/fisiopatología , Masculino , Embarazo , Estudios Retrospectivos
8.
Lipids Health Dis ; 20(1): 100, 2021 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-34496884

RESUMEN

BACKGROUND: The deleterious effect of maternal high-fat diet (HFD) on the fetal rat liver may cause later development of non-alcoholic fatty liver disease (NAFLD). The aim of this study was to evaluate the effect of maternal HFD-induced maternal hepatic steatosis and dysbiosis on the fetal liver and intestines, and the effect of prenatal metformin in a rat model. METHODS: Sprague-Dawley rats were assigned to three groups (N = 6 in each group). Before mating, the rats were randomly assigned to HFD or normal-chow diet (NCD) group for 7 weeks. After mating, the HFD group rats were continued with high-fat diet during pregnancy and some of the HFD group rats were co-treated with metformin (HFMf) via drinking water during pregnancy. All maternal rats and their fetuses were sacrificed on gestational day 21. The liver and intestinal tissues of both maternal and fetal rats were analyzed. In addition, microbial deoxyribonucleic acid extracted from the maternal fecal samples was analyzed. RESULTS: HFD resulted in maternal weight gain during pregnancy, intrahepatic lipid accumulation, and change in the serum short-chain fatty acid profile, intestinal tight junctions, and dysbiosis in maternal rats. The effect of HFD on maternal rats was alleviated by prenatal metformin, which also ameliorated inflammation and apoptosis in the fetal liver and intestines. CONCLUSIONS: This study demonstrated the beneficial effects of prenatal metformin on maternal liver steatosis, focusing on the gut-liver axis. In addition, the present study indicates that prenatal metformin could ameliorate maternal HFD-induced inflammation and apoptosis in the fetal liver and intestines. This beneficial effect of in-utero exposure of metformin on fetal liver and intestines has not been reported. This study supports the use of prenatal metformin for pregnant obese women.


Asunto(s)
Disbiosis/prevención & control , Microbioma Gastrointestinal/efectos de los fármacos , Hipoglucemiantes/farmacología , Hígado/efectos de los fármacos , Metformina/farmacología , Enfermedad del Hígado Graso no Alcohólico/prevención & control , Obesidad/tratamiento farmacológico , Administración Oral , Animales , Apoptosis/efectos de los fármacos , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Agua Potable/administración & dosificación , Disbiosis/etiología , Disbiosis/metabolismo , Disbiosis/patología , Ácidos Grasos Volátiles/metabolismo , Femenino , Feto , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Inflamación , Intestinos/efectos de los fármacos , Intestinos/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/metabolismo , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Obesidad/etiología , Obesidad/metabolismo , Obesidad/patología , Embarazo , Ratas , Ratas Sprague-Dawley , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/metabolismo
9.
Int J Mol Sci ; 22(5)2021 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-33800916

RESUMEN

Gut microbiota-derived metabolites, in particular short chain fatty acids (SCFAs) and their receptors, are linked to hypertension. Fructose and antibiotics are commonly used worldwide, and they have a negative impact on the gut microbiota. Our previous study revealed that maternal high-fructose (HF) diet-induced hypertension in adult offspring is relevant to altered gut microbiome and its metabolites. We, therefore, intended to examine whether minocycline administration during pregnancy and lactation may further affect blood pressure (BP) programmed by maternal HF intake via mediating gut microbiota and SCFAs. Pregnant Sprague-Dawley rats received a normal diet or diet containing 60% fructose throughout pregnancy and lactation periods. Additionally, pregnant dams received minocycline (50 mg/kg/day) via oral gavage or a vehicle during pregnancy and lactation periods. Four groups of male offspring were studied (n = 8 per group): normal diet (ND), high-fructose diet (HF), normal diet + minocycline (NDM), and HF + minocycline (HFM). Male offspring were killed at 12 weeks of age. We observed that the HF diet and minocycline administration, both individually and together, causes the elevation of BP in adult male offspring, while there is no synergistic effect between them. Four groups displayed distinct enterotypes. Minocycline treatment leads to an increase in the F/B ratio, but decreased abundance of genera Lactobacillus, Ruminococcus, and Odoribacter. Additionally, minocycline treatment decreases plasma acetic acid and butyric acid levels. Hypertension programmed by maternal HF diet plus minocycline exposure is related to the increased expression of several SCFA receptors. Moreover, minocycline- and HF-induced hypertension, individually or together, is associated with the aberrant activation of the renin-angiotensin system (RAS). Conclusively, our results provide a new insight into the support of gut microbiota and its metabolite SCAFs in the developmental programming of hypertension and cast new light on the role of RAS in this process, which will help prevent hypertension programmed by maternal high-fructose and antibiotic exposure.


Asunto(s)
Antibacterianos/toxicidad , Fructosa/toxicidad , Microbioma Gastrointestinal/fisiología , Hipertensión/microbiología , Minociclina/toxicidad , Efectos Tardíos de la Exposición Prenatal , Animales , Antibacterianos/administración & dosificación , Ácidos Grasos Volátiles/metabolismo , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Gramnegativas/aislamiento & purificación , Bacterias Gramnegativas/metabolismo , Bacterias Grampositivas/efectos de los fármacos , Bacterias Grampositivas/aislamiento & purificación , Bacterias Grampositivas/metabolismo , Hipertensión/etiología , Riñón/efectos de los fármacos , Riñón/metabolismo , Lactancia , Masculino , Minociclina/administración & dosificación , Óxido Nítrico/metabolismo , Embarazo , Ratas , Ratas Sprague-Dawley , Receptores de Superficie Celular/biosíntesis , Receptores de Superficie Celular/genética , Sistema Renina-Angiotensina/fisiología
10.
Lipids Health Dis ; 19(1): 105, 2020 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-32450865

RESUMEN

BACKGROUND: Fat accumulation in the liver contributes to the development of non-alcoholic fatty liver disease (NAFLD). N-acetylcysteine (NAC) is an antioxidant, acting both directly and indirectly via upregulation of cellular antioxidants. We examined the mechanisms of liver steatosis after 12 months high fat (HF) diet and tested the ability of NAC to rescue liver steatosis. METHODS: Seven-week-old C57BL/6 (B6) male mice were administered HF diet for 12 months (HF group). Two other groups received HF diet for 12 months accompanied by NAC for 12 months (HFD + NAC(1-12)) or 6 months (HFD + NAC(1-6)). The control group was fed regular diet for 12 months (CD group). RESULTS: Liver steatosis was more pronounced in the HF group than in the CD group after 12 month feeding. NAC intake for 6 or 12 months decreased liver steatosis in comparison with HF diet (p < 0.05). Furthermore, NAC treatment also reduced cellular apoptosis and caspase-3 expression. In the unfolded protein response (UPR) pathway, the expression of ECHS1, HSP60, and HSP70 was decreased in the HFD group (p < 0.05) and rescued by NAC therapy. With regards to the endoplasmic reticulum (ER) stress, Phospho-PERK (p-PERK) and ATF4 expression was decreased in the HF group, and only the HFD + NAC(1-12), but not HFD + NAC(1-6) group, showed significant improvement. CONCLUSION: HF diet for 12 months induces significant liver steatosis via altered ER stress and UPR pathway activity, as well as liver apoptosis. NAC treatment rescues the liver steatosis and apoptosis induced by HF diet.


Asunto(s)
Acetilcisteína/uso terapéutico , Dieta Alta en Grasa , Estrés del Retículo Endoplásmico , Hígado/efectos de los fármacos , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Respuesta de Proteína Desplegada , Acetilcisteína/farmacología , Factor de Transcripción Activador 4/genética , Animales , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Apoptosis , Chaperonina 60/genética , Enoil-CoA Hidratasa/genética , Hígado Graso/tratamiento farmacológico , Hígado Graso/metabolismo , Hígado Graso/fisiopatología , Regulación de la Expresión Génica , Proteínas HSP70 de Choque Térmico/genética , Hígado/metabolismo , Hígado/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Mitocondriales/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/fisiopatología
11.
Lipids Health Dis ; 19(1): 174, 2020 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-32711539

RESUMEN

BACKGROUND: Maternal obesity is an emerging problem in the modern world. Growing evidence suggests that intrauterine high-fat (HF) exposure may predispose progeny to subsequent metabolic challenges. Progeny born to mothers who ate an HF diet also tends to eat an HF diet when growing and aggravate metabolic issues. Thus, the generational transmission of obesity is cyclical. Developing a strategy to prevent the occurrence of metabolic syndrome related to prenatal and/or postnatal HF diet is important. In this study, the reprogramming effects of maternal resveratrol treatment for the progeny with maternal HF/postnatal HF diets were investigated. METHODS: Sprague-Dawley dams were fed either a control or a high-fat/high sucrose diet (HFHS) from mating to lactation. After weaning, the progeny was fed chow or an HF diet. Four experimental groups were yielded: CC (maternal/postnatal control diet), HC (maternal HF/postnatal control diet), CH (maternal control/postnatal HFHS diet), and HH (maternal/postnatal HFHS diet). A fifth group (HRH) received a maternal HFHS diet plus maternal resveratrol treatment and a postnatal chow diet to study the effects of maternal resveratrol therapy. RESULTS: Maternal resveratrol treatment lessened the weight and adiposity of progeny that were programmed by combined prenatal and postnatal HFHS diets. Maternal resveratrol therapy ameliorated the decreased abundance of the sirtuin 1 (SIRT1) enzyme in retroperitoneal tissue and the altered leptin/soluble leptin receptor ratio of progeny. Maternal resveratrol therapy also decreased lipogenesis and increased lipolysis for progeny. CONCLUSIONS: Maternal resveratrol intervention can prevent adiposity programmed by maternal and postnatal HFHS diets by inducing lipid metabolic modulation. This study offers a novel reprogramming role for the effect of maternal resveratrol supplements against obesity.


Asunto(s)
Adiposidad/efectos de los fármacos , Resveratrol/farmacología , Análisis de Varianza , Animales , Western Blotting , Peso Corporal/efectos de los fármacos , Dieta Alta en Grasa/efectos adversos , Ingestión de Alimentos/efectos de los fármacos , Femenino , Lactancia/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Lipogénesis/efectos de los fármacos , Lipólisis/efectos de los fármacos , Masculino , Obesidad/metabolismo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sirtuina 1/metabolismo
12.
Int J Mol Sci ; 21(10)2020 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-32408716

RESUMEN

To examine the effects of maternal resveratrol in rats borne to dams with gestational high-fat diet (HFD)/obesity with or without postnatal high-fat diet. We first tested the effects of maternal resveratrol intake on placenta and male fetus brain in rats borne to dams with gestational HFD/obesity. Then, we assessed the possible priming effect of a subsequent insult, male offspring were weaned onto either a rat chow or a HFD. Spatial learning and memory were assessed by Morris water maze test. Blood pressure and peripheral insulin resistance were examined. Maternal HFD/obesity decreased adiponectin, phosphorylation alpha serine/threonine-protein kinase (pAKT), sirtuin 1 (SIRT1), and brain-derived neurotrophic factor (BDNF) in rat placenta, male fetal brain, and adult male offspring dorsal hippocampus. Maternal resveratrol treatment restored adiponectin, pAKT, and BDNF in fetal brain. It also reduced body weight, peripheral insulin resistance, increased blood pressure, and alleviated cognitive impairment in adult male offspring with combined maternal HFD and postnatal HFD. Maternal resveratrol treatment restored hippocampal pAKT and BDNF in rats with combined maternal HFD and postnatal HFD in adult male offspring dorsal hippocampus. Maternal resveratrol intake protects the fetal brain in the context of maternal HFD/obesity. It effectively reduced the synergistic effects of maternal HFD/obesity and postnatal HFD on metabolic disturbances and cognitive impairment in adult male offspring. Our data suggest that maternal resveratrol intake may serve as an effective therapeutic strategy in the context of maternal HFD/obesity.


Asunto(s)
Resistencia a la Insulina/fisiología , Fenómenos Fisiologicos Nutricionales Maternos/fisiología , Obesidad/fisiopatología , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Resveratrol/administración & dosificación , Adiponectina/metabolismo , Animales , Antioxidantes/administración & dosificación , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Disfunción Cognitiva/fisiopatología , Dieta Alta en Grasa/efectos adversos , Femenino , Humanos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Obesidad/metabolismo , Placenta/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal/etiología , Efectos Tardíos de la Exposición Prenatal/metabolismo , Ratas Sprague-Dawley , Destete
13.
Int J Mol Sci ; 21(12)2020 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-32604820

RESUMEN

Hypertension and chronic kidney disease (CKD) can originate during early-life. Tryptophan metabolites generated by different pathways have both detrimental and beneficial effects. In CKD, uremic toxins from the tryptophan-generating metabolites are endogenous ligands of the aryl hydrocarbon receptor (AHR). The interplay between AHR, nitric oxide (NO), the renin-angiotensin system (RAS), and gut microbiota is involved in the development of hypertension. We examined whether tryptophan supplementation in pregnancy can prevent hypertension and kidney disease programmed by maternal CKD in adult offspring via the aforementioned mechanisms. Sprague-Dawley (SD) female rats received regular chow or chow supplemented with 0.5% adenine for 3 weeks to induce CKD before pregnancy. Pregnant controls or CKD rats received vehicle or tryptophan 200 mg/kg per day via oral gavage during pregnancy. Male offspring were divided into four groups (n = 8/group): control, CKD, tryptophan supplementation (Trp), and CKD plus tryptophan supplementation (CKDTrp). All rats were sacrificed at the age of 12 weeks. We found maternal CKD induced hypertension in adult offspring, which tryptophan supplementation prevented. Maternal CKD-induced hypertension is related to impaired NO bioavailability and non-classical RAS axis. Maternal CKD and tryptophan supplementation differentially shaped distinct gut microbiota profile in adult offspring. The protective effect of tryptophan supplementation against maternal CKD-induced programmed hypertension is relevant to alterations to several tryptophan-metabolizing microbes and AHR signaling pathway. Our findings support interplay among tryptophan-metabolizing microbiome, AHR, NO, and the RAS in hypertension of developmental origins. Furthermore, tryptophan supplementation in pregnancy could be a potential approach to prevent hypertension programmed by maternal CKD.


Asunto(s)
Microbioma Gastrointestinal , Hipertensión/prevención & control , Efectos Tardíos de la Exposición Prenatal/prevención & control , Receptores de Hidrocarburo de Aril/metabolismo , Insuficiencia Renal Crónica/complicaciones , Triptófano/administración & dosificación , Triptófano/metabolismo , Animales , Antidepresivos de Segunda Generación/administración & dosificación , Antidepresivos de Segunda Generación/metabolismo , Suplementos Dietéticos , Femenino , Hipertensión/etiología , Hipertensión/metabolismo , Hipertensión/patología , Masculino , Exposición Materna , Embarazo , Efectos Tardíos de la Exposición Prenatal/etiología , Efectos Tardíos de la Exposición Prenatal/metabolismo , Efectos Tardíos de la Exposición Prenatal/patología , Sustancias Protectoras/administración & dosificación , Sustancias Protectoras/metabolismo , Ratas , Ratas Sprague-Dawley
14.
Int J Mol Sci ; 21(15)2020 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-32752013

RESUMEN

Gut microbiota-dependent metabolites, in particular trimethylamine (TMA), are linked to hypertension. Maternal 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exposure or consumption of food high in fructose (HFR) can induce hypertension in adult offspring. We examined whether 3,3-maternal dimethyl-1-butanol (DMB, an inhibitor of TMA formation) therapy can protect adult offspring against hypertension arising from combined HFR and TCDD exposure. Pregnant Sprague-Dawley rats received regular chow or chow supplemented with fructose (60% diet by weight) throughout pregnancy and lactation. Additionally, the pregnant dams received TCDD (200 ng/kg BW orally) or a corn oil vehicle on days 14 and 21 of gestation, and days 7 and 14 after birth. Some mother rats received 1% DMB in their drinking water throughout pregnancy and lactation. Six groups of male offspring were studied (n = 8 for each group): regular chow (CV), high-fructose diet (HFR), regular diet+TCDD exposure (CT), HFR+TCDD exposure (HRT), high-fructose diet+DMB treatment (HRD), and HFR+TCDD+DMB treatment (HRTD). Our data showed that TCDD exacerbates HFR-induced elevation of blood pressure in male adult offspring, which was prevented by maternal DMB administration. We observed that different maternal insults induced distinct enterotypes in adult offspring. The beneficial effects of DMB are related to alterations of gut microbiota, the increase in nitric oxide (NO) bioavailability, the balance of the renin-angiotensin system, and antagonization of aryl hydrocarbon receptor (AHR) signaling. Our findings cast new light on the role of early intervention targeting of the gut microbiota-dependent metabolite TMA, which may allow us to prevent the development of hypertension programmed by maternal excessive fructose intake and environmental dioxin exposure.


Asunto(s)
Dieta de Carga de Carbohidratos/efectos adversos , Microbioma Gastrointestinal , Hipertensión , Exposición Materna/efectos adversos , Efectos Tardíos de la Exposición Prenatal , Animales , Dioxinas/efectos adversos , Femenino , Fructosa/efectos adversos , Masculino , Metilaminas/farmacología , Embarazo , Sustancias Protectoras/farmacología , Ratas , Ratas Sprague-Dawley
15.
Lipids Health Dis ; 18(1): 19, 2019 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-30658634

RESUMEN

BACKGROUND: Prenatal dexamethasone treatment has been shown to enhance the susceptibility of offspring to postnatal high-fat (HF) diet-induced programmed obesity. We investigated the metabolic phenotypes, nutrient-sensing signal and circadian-clock genes in adipose tissue that are programmed by prenatal dexamethasone exposure and postnatal HF diet. METHODS: Male offspring of Sprague-Dawley rats were divided into four experimental groups: normal diet, prenatal dexamethasone exposure, postnatal HF diet, and prenatal dexamethasone plus postnatal HF diet. Postnatal HF diet was prescribed from weaning to 6 months of age. RESULTS: Prenatal dexamethasone and postnatal HF diet exerted synergistic effects on body weight and visceral adiposity, whereas prenatal dexamethasone and postnatal HF diet altered the metabolic profile and caused leptin dysregulation. Prenatal dexamethasone and postnatal HF diet distinctly influenced nutrient-sensing molecules and circadian-clock genes in adipose tissue. The mRNA expression of mTOR, AMPK-α2, PPAR-α, and PPAR-γ was suppressed by prenatal dexamethasone but enhanced by postnatal HF diet. CONCLUSION: Prenatal dexamethasone and postnatal HF treatment cause dysregulation of nutrient-sensing molecules and circadian-clock genes in visceral adipose tissue. Characterizing altered nutrient-sensing molecules and circadian-clock genes has potential therapeutic relevance with respect to the pathogenesis and treatment of prenatal stress and postnatal HF diet-related metabolic disorders.


Asunto(s)
Fenómenos Fisiológicos Nutricionales de los Animales/genética , Relojes Circadianos/genética , Dexametasona/efectos adversos , Dieta Alta en Grasa , Grasa Intraabdominal/patología , Obesidad/etiología , Obesidad/genética , Efectos Tardíos de la Exposición Prenatal/genética , Animales , Biomarcadores/metabolismo , Glucemia/metabolismo , Peso Corporal , Femenino , Inflamación/genética , Inflamación/patología , Grasa Intraabdominal/metabolismo , Leptina/metabolismo , Tamaño de los Órganos , Embarazo , Ratas Sprague-Dawley , Sirtuina 1/metabolismo
16.
Int J Mol Sci ; 20(3)2019 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-30695995

RESUMEN

Resveratrol, also known as 3,5,4'-trihydroxystilbene, is a natural polyphenol that occurs as a phytoalexin. It is produced by plant sources such as grapes, apples, blueberries, plums, peanuts, and other oilseeds. This compound has a variety of effects on human health and diseases. This review summarizes the mounting evidence that resveratrol is helpful in treating metabolic syndrome and related disorders. Resveratrol can be provided either early as a reprogramming agent or later as part of treatment. A few of the main mechanisms underlying the beneficial effects of resveratrol on metabolic syndrome are outlined. This review also discusses the potential of resveratrol derivatives as a complementary or alternative medicine. In conclusion, resveratrol could be a useful regimen for the prevention and treatment of metabolic syndrome and its related conditions.


Asunto(s)
Síndrome Metabólico/tratamiento farmacológico , Resveratrol/uso terapéutico , Animales , Modelos Animales de Enfermedad , Humanos , Resveratrol/síntesis química , Resveratrol/química , Resveratrol/farmacología
17.
Int J Mol Sci ; 20(16)2019 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-31416234

RESUMEN

Hypertension can originate from early-life exposure to oxidative stress. As reported, dimethyl fumarate (DMF) activates nuclear factor erythroid-derived 2-related factor 2 (Nrf2) and protects against oxidative stress damage. We examined whether maternal DMF therapy protects adult offspring against hypertension programmed by prenatal dexamethasone (DEX) and postnatal high-fat (HF) diet exposure. We examined male Sprague Dawley rat offspring at 4 months of age from five groups (n = 11-13/group): control, DEX (0.1mg/kg i.p. from gestational day 16 to 22), HF (D12331 diet from weaning to 16 weeks of age), DEX+HF, and DEX+HF+DMF (50mg/kg/day via gastric gavage for 3 weeks during pregnancy). Maternal DMF therapy prevented male offspring against hypertension programmed by combined DEX and HF exposures. The protective effects of maternal DMF include reduced oxidative stress, decreased plasma asymmetric dimethylarginine (ADMA) levels, downregulated the renin-angiotensin system (i.e. Ren, Agt, Ace, and Agtr1a), increased renal protein levels of certain nutrient-sensing signals, and promoted autophagy. In conclusion, maternal Nrf2 activation by DMF protects male adult offspring against hypertension programmed by combined DEX and HF exposures. Our results cast a new light on the therapeutic potential of targeting Nrf2 signaling pathway as reprogramming strategies to prevent programmed hypertension in children exposed to antenatal corticosteroids and postnatally excessive consumption of fat.


Asunto(s)
Dexametasona/administración & dosificación , Dieta Alta en Grasa , Dimetilfumarato/administración & dosificación , Hipertensión/etiología , Hipertensión/prevención & control , Exposición Materna , Factor 2 Relacionado con NF-E2/agonistas , Efectos Tardíos de la Exposición Prenatal , Animales , Biomarcadores , Femenino , Expresión Génica , Humanos , Hipertensión/diagnóstico , Hipertensión/metabolismo , Recién Nacido , Masculino , Factor 2 Relacionado con NF-E2/genética , Estrés Oxidativo , Fosforilación , Embarazo , Ratas
18.
Allergol Int ; 68(4): 486-493, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31248809

RESUMEN

BACKGROUND: Dengue fever (DF) is the most rapidly spreading mosquito-borne viral disease. Practical vaccines or specific therapeutics are still expected. Environmental factors and genetic factors affect the susceptibility of Dengue virus (DV) infection. Asthma is a common allergic disease, with house dust mites (HDMs) being the most important allergens. Asthmatic patients are susceptible to several microorganism infections. METHODS: A nationwide population-based cohort analysis was designed to assess whether to determine whether asthma can be a risk factor for DF. RESULTS: Unexpectedly, our data from a nationwide population-based cohort revealed asthmatic patients are at a decreased risk of DF. Compared to patients without asthma, the hazard ratio (HR) for DF in patients with asthma was 0.166 (95% CI: 0.118-0.233) after adjustment for possible confounding factors. In the age stratification, the adjusted HR for DF in young adult patients with asthma was 0.063. Dendritic cell-specific intercellular adhesion molecule 3-grabbing non-integrin (DC-SIGN) of dendritic cells (DCs) is an important entry for DV. Through another in vitro experiment, we found that HDM can diminish surface expression of DC-SIGN in monocyte-derived DCs and further decrease the cellular entry of DV. CONCLUSIONS: Decreased DC-SIGN expression in DCs of allergic asthmatic patient may be one of many factors for them to be protected against DF. This could implicate the potential for DC-SIGN modulation as a candidate target for designing therapeutic strategies for DF.


Asunto(s)
Asma/complicaciones , Asma/epidemiología , Dengue/epidemiología , Dengue/etiología , Susceptibilidad a Enfermedades , Vigilancia de la Población , Adolescente , Adulto , Alérgenos/inmunología , Asma/diagnóstico , Asma/etiología , Biomarcadores , Moléculas de Adhesión Celular/inmunología , Niño , Preescolar , Estudios de Cohortes , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Dengue/diagnóstico , Dengue/virología , Femenino , Humanos , Lactante , Recién Nacido , Lectinas Tipo C/inmunología , Recuento de Leucocitos , Masculino , Persona de Mediana Edad , Monocitos/inmunología , Monocitos/metabolismo , Receptores de Superficie Celular/inmunología , Adulto Joven
19.
Lipids Health Dis ; 17(1): 178, 2018 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-30055626

RESUMEN

BACKGROUND: Nonalcoholic fatty liver disease (NAFLD) can develop in prenatal stages and can be exacerbated by exposure to a postnatal high-fat (HF) diet. We investigated the protective effects of resveratrol on prenatal and postnatal HF diet-induced NAFLD. METHODS: Male Sprague-Dawley rat offspring were placed in five experimental groups (n = 10-12 per group): normal diet (VNF), maternal HF diet (ONF), postnatal HF diet (VHF), and maternal HF diet/postnatal HF diet (OHF). A therapeutic group with resveratrol for maternal HF diet/postnatal HF diet (OHFR) was used for comparison. Resveratrol (50 mg/kg/day) was dissolved in drinking water for offspring from post-weaning to postnatal day (PND) 120. RESULTS: We found that HF/HF-induced NAFLD was prevented in adult offspring by the administration of resveratrol. Resveratrol administration mediated a protective effect on rats on HF/HF by regulating lipid metabolism, reducing oxidative stress and apoptosis, restoring nutrient-sensing pathways by increasing Sirt1 and leptin expression, and mediating the renin-angiotensin system (RAS) to decrease angiotensinogen, renin, ACE1, and AT1R levels and increased ACE2, AT2R and MAS1 levels compared to those in the OHF group. CONCLUSION: Our results suggest that a maternal and post-weaning HF diet increases liver steatosis and apoptosis via the RAS. Resveratrol might serve as a therapeutic target by mediating protective actions against NAFLD in offspring exposed to a combination of maternal and postnatal HF diet.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Hipolipemiantes/farmacología , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Sistema Renina-Angiotensina/efectos de los fármacos , Estilbenos/farmacología , Enzima Convertidora de Angiotensina 2 , Angiotensinógeno/genética , Angiotensinógeno/metabolismo , Animales , Animales Recién Nacidos , Dieta Alta en Grasa/efectos adversos , Femenino , Leptina/genética , Leptina/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Fenómenos Fisiologicos Nutricionales Maternos , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Estrés Oxidativo/efectos de los fármacos , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 1/metabolismo , Receptor de Angiotensina Tipo 2/genética , Receptor de Angiotensina Tipo 2/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Renina/genética , Renina/metabolismo , Resveratrol , Sirtuina 1/genética , Sirtuina 1/metabolismo , Destete
20.
Lipids Health Dis ; 17(1): 50, 2018 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-29540174

RESUMEN

BACKGROUND: Hypertension may result from high-fat (HF) diet induced-obesity and overexposure to glucocorticoids in utero. Recent studies demonstrated the potent contribution of adipose tissue's renin-angiotensin system (RAS) to systemic RAS, which plays a key role in regulating blood pressure (BP). In this study, we investigated the effects of prenatal dexamethasone (DEX) exposure and postnatal HF diet on RAS of adipose tissue. METHODS: RAS and BP of 6-month old rats exposed to prenatal DEX and/or postnatal HF diet were examined. RESULTS: Prenatal DEX plus postnatal HF exerted a synergistic effect on systolic BP. Prenatal DEX exposure suppressed plasma angiotensin (ANG) I and ANG II, whereas postnatal HF suppressed plasma ANG-(1-7) level. Prenatal DEX increased prorenin receptor and renin levels, but suppressed angiotensinogen (AGT) and angiotensin-converting-enzyme 1 (ACE1) mRNA expressions in adipose tissue. Postnatal HF increased AGT mRNA expression, but suppressed prorenin receptor, renin, ACE2, ANG II type 2 receptor (AT2R), and Mas receptor (MasR) mRNA expression levels. CONCLUSIONS: Prenatal GC exposure altered the ACE1/ANG II/ANG II type 1 receptor (AT1R) axis, whereas postnatal HF negatively impacted the ACE2/ANG-(1-7)/MasR axis. Prenatal DEX exposure and postnatal HF synergistically elevated BP through a distinct programming mechanism of systemic and adipose RAS. Adipose RAS might be a target for precise hypertension treatment.


Asunto(s)
Dexametasona/efectos adversos , Dieta Alta en Grasa/efectos adversos , Hipertensión/genética , Sistema Renina-Angiotensina/efectos de los fármacos , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Angiotensinógeno/genética , Animales , Presión Sanguínea/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Hipertensión/inducido químicamente , Hipertensión/patología , Peptidil-Dipeptidasa A/genética , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/genética , Efectos Tardíos de la Exposición Prenatal/patología , Proto-Oncogenes Mas , Ratas , Receptores de Superficie Celular/genética , Renina/genética , Sistema Renina-Angiotensina/genética , ATPasas de Translocación de Protón Vacuolares/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA