Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Physiol Rep ; 11(23): e15881, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38031314

RESUMEN

At the end of gestation, fetal skeleton rapidly accumulates calcium, and bone development continues in offspring postnatally. To accommodate, maternal skeletal physiology is modulated in a serotonin-dependent manner. Selective serotonin reuptake inhibitors (SSRIs) are generally considered safe for treatment of major depressive disorder, postpartum depression, and other psychiatric illnesses during the peripartum period, but because serotonin affects bone remodeling, SSRIs are associated with decreased bone mass across all ages and sexes, and the impact of SSRIs during fetal and postnatal development has not been fully investigated. In the present study, our aim was to examine developmental fluoxetine exposure on offspring skeleton and to assess varying degrees of impact depending on dose and window of exposure in short-term and long-term contexts. We established that a low dose of lactational fluoxetine exposure caused a greater degree of insult to offspring bone than either a low dose during fetal and postpartum development or a high dose during lactation only in mice. We further discovered lasting impacts of developmental fluoxetine exposure, especially during lactation only, on adult bone and body composition. Herein, we provide evidence fluoxetine exposure during early development may have detrimental effects on the skeleton of offspring at weaning and into adulthood.


Asunto(s)
Trastorno Depresivo Mayor , Efectos Tardíos de la Exposición Prenatal , Femenino , Ratones , Animales , Humanos , Fluoxetina/toxicidad , Inhibidores Selectivos de la Recaptación de Serotonina/toxicidad , Trastorno Depresivo Mayor/tratamiento farmacológico , Serotonina , Huesos , Efectos Tardíos de la Exposición Prenatal/tratamiento farmacológico
2.
Molecules ; 26(13)2021 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-34209270

RESUMEN

Resveratrol butyrate esters (RBE) are derivatives of resveratrol (RSV) and butyric acid and exhibit biological activity similar to that of RSV but with higher bioavailability. The aim of this study was designed as an animal experiment to explore the effects of RBE on the serum biochemistry, and fat deposits in the offspring rats exposed to bisphenol A (BPA), along with the growth and decline of gut microbiota. We constructed an animal model of perinatal Bisphenol A (BPA) exposure to observe the effects of RBE supplementation on obesity, blood lipids, and intestinal microbiota in female offspring rats. Perinatal exposure to BPA led to weight gain, lipid accumulation, high levels of blood lipids, and deterioration of intestinal microbiota in female offspring rats. RBE supplementation reduced the weight gain and lipid accumulation caused by BPA, optimised the levels of blood lipids, significantly reduced the Firmicutes/Bacteroidetes (F/B) ratio, and increased and decreased the abundance of S24-7 and Lactobacillus, respectively. The analysis of faecal short-chain fatty acid (SCFA) levels revealed that BPA exposure increased the faecal concentration of acetate, which could be reduced via RBE supplementation. However, the faecal concentrations of propionate and butyrate were not only significantly lower than that of acetate, but also did not significantly change in response to BPA exposure or RBE supplementation. Hence, RBE can suppress BPA-induced obesity in female offspring rats, and it demonstrates excellent modulatory activity on intestinal microbiota, with potential applications in perinatological research.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Ácido Butírico/farmacología , Obesidad , Fenoles/toxicidad , Efectos Tardíos de la Exposición Prenatal , Resveratrol/farmacología , Animales , Ácidos Grasos Volátiles/metabolismo , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Obesidad/inducido químicamente , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/tratamiento farmacológico , Efectos Tardíos de la Exposición Prenatal/metabolismo , Ratas , Ratas Sprague-Dawley
3.
J Steroid Biochem Mol Biol ; 212: 105941, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34147644

RESUMEN

PURPOSE: The overexposure to synthetic glucocorticoids (GC) during pregnancy can predispose to metabolic diseases during adulthood. Vitamin D is not only crucial for fetal development, but also exerts direct effects on the GC sensitivity and down-regulates GC receptors. Given the vitamin D effects on glucocorticoid-related parameters, we aimed to investigate a possible protective role of maternal vitamin D administration on the glucose homeostasis of rats exposed to dexamethasone in utero. METHODS: Pregnant rats received dexamethasone (0.1 mg/kg, Dex) daily between the 14th and 19th days of pregnancy. A subgroup of dexamethasone-treated dams received oral administration of vitamin D (500UI, DexVD) during the whole gestation. The corresponding control groups of dams were included (CTL and VD groups, respectively). Male and female offspring were evaluated at 3, 6 and 12 months of age. RESULTS: Prenatal exposure to dexamethasone caused metabolic disruption in an age and sex-dependent manner being the older male offspring more susceptible to insulin resistance, fatty liver and beta-cell mass expansion than females. Furthermore, we demonstrated that prenatal GC led to glucose intolerance in male and female offspring in an age-dependent manner. Maternal vitamin D administration did not influence glucose intolerance but attenuated the insulin resistance, liver lipid accumulation and prevented the beta-cell mass expansion caused by prenatal dexamethasone in the male offspring. CONCLUSION: Maternal vitamin D administration mitigates metabolic disturbances that occur later in life in male rats exposed to GC in utero. Moreover, our data suggest vitamin D as an important nutritional supplement for pregnant overexposed to GC during gestation.


Asunto(s)
Dexametasona/efectos adversos , Glucocorticoides/efectos adversos , Enfermedades Metabólicas/tratamiento farmacológico , Efectos Tardíos de la Exposición Prenatal/tratamiento farmacológico , Vitamina D/uso terapéutico , Vitaminas/uso terapéutico , Animales , Femenino , Células Secretoras de Insulina/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Intercambio Materno-Fetal , Enfermedades Metabólicas/sangre , Enfermedades Metabólicas/inducido químicamente , Enfermedades Metabólicas/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal/sangre , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/metabolismo , Ratas Wistar , Caracteres Sexuales , Triglicéridos/sangre , Triglicéridos/metabolismo , Vitamina D/farmacología , Vitaminas/farmacología
4.
Neurotoxicology ; 85: 90-98, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33991534

RESUMEN

Motor dysfunctions are common comorbidities among autism spectrum disorder (ASD) patients. Abnormal cerebellar development throughout critical periods may have an effect on motor functions and result in motor impairments. Vitamin A (VA) plays a crucial role in the developing process of the nervous system. The correlation of VA deficiency (VAD) and ASD with motor dysfunctions, however, is not clear. Therefore, we built rat models with different VA levels based on the valproic acid (VPA)-treated autism model. ASD rats with VAD showed aggravated motor coordination abnormalities, Purkinje cell loss and impaired dendritic arborization of Purkinje cells compared to ASD rats with normal VA levels (VA normal, VAN). Additionally, the expression levels of retinoid-related orphan receptor α (RORα) and retinoic acid receptor α (RARα) were lower in the cerebellum of ASD rats with VAD than in those of ASD rats with VAN. VA supplementation (VAS) effectively improved motor coordination and cerebellar Purkinje cell abnormalities in ASD rats with VAD. Furthermore, the results of chromatin immunoprecipitation (ChIP) assays confirmed that the enrichment of RARα was detected on the RORα promoter in the cerebellum and that VAS could upregulate the binding capacity of RARα for RORα promoters. These results showed that VAD in autism might result in cerebellar impairments and be a factor aggravating a subtype of ASD with motor comorbidities. The therapeutic effect of VAS on motor deficits and Purkinje neuron impairments in autism might be due to the regulation of RORα by RARα.


Asunto(s)
Trastorno Autístico/metabolismo , Cerebelo/metabolismo , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Ácido Valproico/toxicidad , Deficiencia de Vitamina A/metabolismo , Vitamina A/administración & dosificación , Animales , Trastorno Autístico/inducido químicamente , Trastorno Autístico/tratamiento farmacológico , Cerebelo/efectos de los fármacos , Femenino , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/tratamiento farmacológico , Efectos Tardíos de la Exposición Prenatal/metabolismo , Desempeño Psicomotor/efectos de los fármacos , Desempeño Psicomotor/fisiología , Ratas , Ratas Sprague-Dawley , Deficiencia de Vitamina A/tratamiento farmacológico
5.
FASEB J ; 35(5): e21477, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33891326

RESUMEN

Chronic fetal hypoxia is one of the most common outcomes in complicated pregnancy in humans. Despite this, its effects on the long-term health of the brain in offspring are largely unknown. Here, we investigated in rats whether hypoxic pregnancy affects brain structure and function in the adult offspring and explored underlying mechanisms with maternal antioxidant intervention. Pregnant rats were randomly chosen for normoxic or hypoxic (13% oxygen) pregnancy with or without maternal supplementation with vitamin C in their drinking water. In one cohort, the placenta and fetal tissues were collected at the end of gestation. In another, dams were allowed to deliver naturally, and offspring were reared under normoxic conditions until 4 months of age (young adult). Between 3.5 and 4 months, the behavior, cognition and brains of the adult offspring were studied. We demonstrated that prenatal hypoxia reduced neuronal number, as well as vascular and synaptic density, in the hippocampus, significantly impairing memory function in the adult offspring. These adverse effects of prenatal hypoxia were independent of the hypoxic pregnancy inducing fetal growth restriction or elevations in maternal or fetal plasma glucocorticoid levels. Maternal vitamin C supplementation during hypoxic pregnancy protected against oxidative stress in the placenta and prevented the adverse effects of prenatal hypoxia on hippocampal atrophy and memory loss in the adult offspring. Therefore, these data provide a link between prenatal hypoxia, placental oxidative stress, and offspring brain health in later life, providing insight into mechanism and identifying a therapeutic strategy.


Asunto(s)
Ácido Ascórbico/uso terapéutico , Atrofia/tratamiento farmacológico , Hipoxia Fetal/complicaciones , Hipocampo/efectos de los fármacos , Trastornos de la Memoria/tratamiento farmacológico , Efectos Tardíos de la Exposición Prenatal/tratamiento farmacológico , Animales , Animales Recién Nacidos , Antioxidantes/uso terapéutico , Atrofia/etiología , Atrofia/metabolismo , Atrofia/patología , Suplementos Dietéticos , Modelos Animales de Enfermedad , Femenino , Retardo del Crecimiento Fetal/tratamiento farmacológico , Retardo del Crecimiento Fetal/etiología , Retardo del Crecimiento Fetal/metabolismo , Retardo del Crecimiento Fetal/patología , Masculino , Trastornos de la Memoria/etiología , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/patología , Embarazo , Complicaciones del Embarazo/tratamiento farmacológico , Complicaciones del Embarazo/etiología , Complicaciones del Embarazo/metabolismo , Complicaciones del Embarazo/patología , Efectos Tardíos de la Exposición Prenatal/etiología , Efectos Tardíos de la Exposición Prenatal/metabolismo , Efectos Tardíos de la Exposición Prenatal/patología , Ratas , Ratas Wistar
6.
Biochem Biophys Res Commun ; 554: 151-157, 2021 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-33798941

RESUMEN

It has been suggested that the intelligence quotient of children born to pregnant women taking 1000 mg or more of valproic acid per day is lower than that of children born to pregnant women taking other antiepileptic drugs. However, the mechanism whereby intelligence quotient is decreased in children exposed to valproic acid during the fetal period has not yet been elucidated. Therefore, we used the human neuroblastoma cell line SH-SY5Y to evaluate the effects of antiepileptic drugs containing valproic acid on nerve cells. We assessed the anti-proliferative effects of drugs in these cells via WST-8 colorimetric assay, using the Cell Counting Kit-8. We also quantified drug effects on axonal elongation from images using ImageJ software. We also evaluated drug effects on mRNA expression levels on molecules implicated in nervous system development and folic acid uptake using real-time PCR. We observed that carbamazepine and lamotrigen were toxic to SH-SY5Y cells at concentrations >500 µM. In contrast, phenytoin and valproic acid were not toxic to these cells. Carbamazepine, lamotrigen, phenytoin, and valproic acid did not affect axonal outgrowth in SH-SY5Y cells. Sodium channel neuronal type 1a (SCN1A) mRNA expression-level ratios increased when valproic acid was supplemented to cells. The overexpression of SCN1A mRNA due to high valproic acid concentrations during the fetal period may affect neurodevelopment. However, since detailed mechanisms have not yet been elucidated, it is necessary to evaluate it by comparing cell axon elongation and SCN1A protein expression due to high-concentration valproic acid exposure.


Asunto(s)
Anticonvulsivantes/farmacología , Epilepsia/tratamiento farmacológico , Canal de Sodio Activado por Voltaje NAV1.1/metabolismo , Neuroblastoma/tratamiento farmacológico , Proyección Neuronal/efectos de los fármacos , Neuronas/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal/tratamiento farmacológico , Carbamazepina/farmacología , Línea Celular Tumoral , Proliferación Celular , Niño , Epilepsia/complicaciones , Epilepsia/metabolismo , Femenino , Humanos , Lamotrigina/farmacología , Canal de Sodio Activado por Voltaje NAV1.1/genética , Neuroblastoma/complicaciones , Neuroblastoma/metabolismo , Fenitoína/farmacología , Embarazo , Ácido Valproico/farmacología
7.
J Gerontol A Biol Sci Med Sci ; 76(6): 991-995, 2021 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-33249457

RESUMEN

Folic acid (FA) supplementation is important during pregnancy to avoid malformations in the offspring. However, it is unknown if it can affect the offspring throughout their lives. To evaluate the offspring, female mother rats (dams) were separated into 5 groups: Four groups received the AIN-93 diet, divided into control and FA (5, 10, and 50 mg/kg), and an additional group received a FA-deficient diet, and the diet was performed during pregnancy and lactation. We evaluated the female offspring of these dams (at 2 and 18 months old). The aged offspring fed with FA-deficient diet presented habituation, spatial and aversive memory impairment and the FA maternal supplementation prevented this. The natural aging caused an increase in the TNF-α and IL-1ß levels in the hippocampus from 18-month-old offspring. FA maternal supplementation was able to prevent the increase of these cytokines. IL-4 levels decreased in the prefrontal cortex from aged control rats and FA prevented it. FA deficiency decreased the levels of IL-4 in the hippocampus of the young offspring. In addition, natural aging and FA deficiency decreased brain-derived neurotrophic factor levels in the hippocampus and nerve growth factor levels in the prefrontal cortex and FA supplementation prevented it. Thus, the present study shows for the first time the effect of FA maternal supplementation on memory, cytokines, and neurotrophins in the aged offspring.


Asunto(s)
Suplementos Dietéticos , Ácido Fólico/uso terapéutico , Inflamación/prevención & control , Trastornos de la Memoria/prevención & control , Efectos Tardíos de la Exposición Prenatal/tratamiento farmacológico , Envejecimiento/efectos de los fármacos , Animales , Femenino , Deficiencia de Ácido Fólico/complicaciones , Hipocampo/metabolismo , Inflamación/etiología , Trastornos de la Memoria/etiología , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Ratas , Ratas Wistar
8.
Nutrients ; 12(10)2020 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-33023237

RESUMEN

Attention deficit hyperactivity disorder (ADHD) is a neurodevelopmental disorder that occurs in children characterized by inattention and hyperactivity. Prenatal alcohol exposure (PAE) can disrupt fetal neuronal development and cause an ADHD-like hyperactive behavior in the offspring. In this study, we hypothesized that metabolic disturbance would involve in ADHD neuropathology and aimed to investigate the changes in metabolite profile in PAE-induced ADHD-like model and the effects of HX106, a nutraceutical, on ADHD-like pathophysiology and metabolite changes. To this end, we administered HX106 to the mouse offspring affected by PAE (OPAE) and assessed the hyperactivity using the open field test. We observed that HX106-treated OPAE showed less hyperactive behavior than vehicle-treated OPAE. The effects of HX106 were found to be related to the regulation of dopamine transporter and D2 dopamine receptor expression. Furthermore, using gas chromatography time-of-flight mass spectrometry-based metabolomics, we explored the metabolite changes among the experimental groups. The metabolite profile, particularly related with the amino acids, linoleic acid and amino sugar pathways, was altered by PAE and reversed by HX106 treatment partially similar to that observed in the control group. Overall, this study suggest that metabolite alteration would be involved in ADHD pathology and that HX106 can be an efficient supplement to overcome ADHD by regulating dopamine signaling-related protein expression and metabolite changes.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad/tratamiento farmacológico , Suplementos Dietéticos , Trastornos del Espectro Alcohólico Fetal/tratamiento farmacológico , Extractos Vegetales/farmacología , Efectos Tardíos de la Exposición Prenatal/tratamiento farmacológico , Animales , Trastorno por Déficit de Atención con Hiperactividad/inducido químicamente , Modelos Animales de Enfermedad , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/efectos de los fármacos , Femenino , Trastornos del Espectro Alcohólico Fetal/psicología , Cromatografía de Gases y Espectrometría de Masas/métodos , Metabolómica , Ratones , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/psicología
9.
J Assist Reprod Genet ; 37(12): 2963-2965, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33083862

RESUMEN

Paternally derived de novo mutations (DNMs) caused by oxidative stress (OS) have been implicated in the development of autism spectrum disorders (ASDs). Whether preconception antioxidant supplementation can reduce the incidence of ASDs by reducing OS is an area of uncertainty and potentially important future scientific investigation.


Asunto(s)
Antioxidantes/administración & dosificación , Trastorno del Espectro Autista/prevención & control , Suplementos Dietéticos , Retardo del Crecimiento Fetal/tratamiento farmacológico , Estrés Oxidativo , Atención Preconceptiva , Efectos Tardíos de la Exposición Prenatal/tratamiento farmacológico , Trastorno del Espectro Autista/epidemiología , Niño , Femenino , Humanos , Masculino , Mutación , Embarazo
10.
J Assist Reprod Genet ; 37(12): 2955-2962, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32949003

RESUMEN

Current research indicates that a sizable number of autism spectrum disorder (ASD) cases arise from de novo mutations (DNMs) occurring within the paternal germline, usually in an age-dependent manner. Andrologists have reported that somatic cells and gametes share the same pathologies that generate these DNMs-specifically, DNA hypomethylation caused by oxidative nucleoside base damage. Because many ASD researchers seek to identify genetic risk factors, teams are developing methods of assessing aberrant DNA patterns, such as parental gonadal mosaicism. Several studies propose antioxidant supplementation as a strategy to lower autism risk, and/or suggest connections between childhood neurodevelopmental disorders such as autism and paternally-derived DNMs. Actual data, however, are currently not available to determine whether male preconception antioxidant supplementation effectively lowers autism risk. The purpose of this paper is to (1) explore the mechanisms causing DNMs, specifically DNA hypomethylation; (2) explain how antioxidant supplementation may lower the risk of having a child with ASD; and, (3) advocate for the implementation of large prospective studies testing (2). These studies may very well find that male preconception supplementation with antioxidants prevents neurodevelopmental disorders in offspring, in much the same way that female prenatal consumption of folate was found to decrease the risk of birth defects. If this is indeed the case, the alarming rise in autism prevalence rates of the past few decades will slow-or even cease-upon the initiation of public awareness campaigns.


Asunto(s)
Antioxidantes/administración & dosificación , Trastorno del Espectro Autista/prevención & control , Suplementos Dietéticos , Retardo del Crecimiento Fetal/tratamiento farmacológico , Atención Preconceptiva , Efectos Tardíos de la Exposición Prenatal/tratamiento farmacológico , Niño , Metilación de ADN , Femenino , Humanos , Masculino , Embarazo
11.
J Tradit Chin Med ; 40(4): 646-653, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32744032

RESUMEN

OBJECTIVE: To assess the protective role of benazepril, an angiotensin-converting enzyme inhibitor, in renal damage caused by prenatal inflammation. METHODS: Saline or lipopolysaccharide were administered intraperitoneally to pregnant Sprague- Dawley rats on gestation days 8, 10, and 12. After birth, offspring received either tap water or benazepril in water between 7 and 68 weeks. Blood pressure, blood urea nitrogen, creatinine, and 24-h urine volume were measured as indices of renal function. Hematoxylin, eosin, periodic acid-Schiff, and Sirius Red staining were used to evaluate renal damage. RESULTS: Postnatal benazepril treatment ameliorated hypertension and restored normal 24-h urine volume and blood urea nitrogen and serum creatinine levels. Benazepril treatment also reduced glycoprotein accumulation and fibrosis in the glomerulus and in tubular epithelial cells and inhibited nuclear factor-kappa B activation. CONCLUSION: Together with our previous findings that postnatal inhibition of nuclear factor-kappa B activation blocks intra-renal renin-angiotensin system activation, our current data demonstrate that intra-renal activation of the renin-angiotensin system interacts with nuclear factor-kappa B activation to cause renal damage in adulthood following prenatal inflammation.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/administración & dosificación , Benzazepinas/administración & dosificación , Riñón/efectos de los fármacos , Exposición Materna/efectos adversos , Efectos Tardíos de la Exposición Prenatal/tratamiento farmacológico , Sistema Renina-Angiotensina/efectos de los fármacos , Animales , Presión Sanguínea/efectos de los fármacos , Femenino , Humanos , Riñón/inmunología , Riñón/lesiones , Lipopolisacáridos/efectos adversos , Masculino , FN-kappa B/genética , FN-kappa B/inmunología , Embarazo , Efectos Tardíos de la Exposición Prenatal/etiología , Efectos Tardíos de la Exposición Prenatal/genética , Efectos Tardíos de la Exposición Prenatal/inmunología , Ratas
12.
Int J Mol Sci ; 21(8)2020 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-32325788

RESUMEN

In previous studies, we produced changes in gene expression in the brain of mice by early postnatal administration of valproic acid (VPA), with distinct differences between genders. The addition of S-adenosine methionine (SAMe) normalized the expression of most genes in both genders, while SAMe alone induced no changes. We treated pregnant dams with a single injection of VPA on day 12.5 of gestation, or with SAMe during gestational days 12-14, or by a combination of VPA and SAMe. In the frontal half of the brain, we studied the expression of 770 genes of the pathways involved in neurophysiology and neuropathology using the NanoString nCounter method. SAMe, but not VPA, induced statistically significant changes in the expression of many genes, with differences between genders. The expression of 112 genes was changed in both sexes, and another 170 genes were changed only in females and 31 only in males. About 30% of the genes were changed by more than 50%. One of the most important pathways changed by SAMe in both sexes was the VEGF (vascular endothelial growth factor) pathway. Pretreatment with VPA prevented almost all the changes in gene expression induced by SAMe. We conclude that large doses of SAMe, if administered prenatally, may induce significant epigenetic changes in the offspring. Hence, SAMe and possibly other methyl donors may be epigenetic teratogens.


Asunto(s)
Trastorno del Espectro Autista/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal/metabolismo , S-Adenosilmetionina/administración & dosificación , Transducción de Señal/efectos de los fármacos , Ácido Valproico/administración & dosificación , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Animales Recién Nacidos , Trastorno del Espectro Autista/metabolismo , Encéfalo/metabolismo , Epigénesis Genética/efectos de los fármacos , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/genética , Ontología de Genes , Masculino , Ratones , Embarazo , Efectos Tardíos de la Exposición Prenatal/tratamiento farmacológico , S-Adenosilmetionina/metabolismo , Transducción de Señal/genética , Transcriptoma , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/genética
13.
J Neurodev Disord ; 12(1): 9, 2020 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-32164522

RESUMEN

BACKGROUND: Despite the high prevalence of fetal alcohol spectrum disorder (FASD), there are few interventions targeting its core neurocognitive and behavioral deficits. FASD is often conceptualized as static and permanent, but interventions that capitalize on brain plasticity and critical developmental windows are emerging. We present a long-term follow-up study evaluating the neurodevelopmental effects of choline supplementation in children with FASD 4 years after an initial efficacy trial. METHODS: The initial study was a randomized, double-blind, placebo-controlled trial of choline vs. placebo in 2-5-year-olds with FASD. Participants include 31 children (16 placebo; 15 choline) seen 4 years after trial completion. The mean age at follow-up was 8.6 years. Diagnoses were 12.9% fetal alcohol syndrome (FAS), 41.9% partial FAS, and 45.1% alcohol-related neurodevelopmental disorder. The follow-up included measures of intelligence, memory, executive functioning, and behavior. RESULTS: Children who received choline had higher non-verbal intelligence, higher visual-spatial skill, higher working memory ability, better verbal memory, and fewer behavioral symptoms of attention deficit hyperactivity disorder than the placebo group. No differences were seen for verbal intelligence, visual memory, or other executive functions. CONCLUSIONS: These data support choline as a potential neurodevelopmental intervention for FASD and highlight the need for long-term follow-up to capture treatment effects on neurodevelopmental trajectories. TRIAL REGISTRATION: ClinicalTrials.Gov #NCT01149538; Registered: June 23, 2010; first enrollment July 2, 2010.


Asunto(s)
Colina/uso terapéutico , Cognición/efectos de los fármacos , Trastornos del Espectro Alcohólico Fetal/tratamiento farmacológico , Nootrópicos/uso terapéutico , Preescolar , Método Doble Ciego , Femenino , Estudios de Seguimiento , Humanos , Inteligencia/efectos de los fármacos , Masculino , Memoria a Corto Plazo/efectos de los fármacos , Embarazo , Efectos Tardíos de la Exposición Prenatal/tratamiento farmacológico
14.
J Clin Endocrinol Metab ; 105(3)2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31665323

RESUMEN

CONTEXT & OBJECTIVES: The Controlled Antenatal Thyroid Screening (CATS) study was the first randomized controlled trial to investigate effects of treating suboptimal gestational thyroid function (SGTF) on child cognition. Since observational studies indicated that SGTF may also increase symptoms of autism and attention-deficit/hyperactivity disorder (ADHD), the CATS cohort was used to investigate whether treatment of mothers affected their children's behavior. DESIGN & PARTICIPANTS: Mothers (N = 475) completed 3 questionnaires: the Strengths and Difficulties Questionnaire (SDQ), the Child ADHD Questionnaire, and the Social Communication Questionnaire (SCQ, used as a screen for autism spectrum disorder [ASD]), about their children (mean age 9.5 years). Group comparisons of total scores, numbers of children above clinical thresholds, and association between high maternal free thyroxine (FT4) (> 97.5th percentile of the UK cohort, "overtreated") and child neurodevelopment were reported. RESULTS: There were no differences in total scores between normal gestational thyroid function (GTF) (n = 246), treated (n = 125), and untreated (n = 104) SGTF groups. More children of treated mothers scored above clinical thresholds, particularly the overtreated. Scores were above thresholds in SDQ conduct (22% vs 7%), SCQ total scores (7% vs 1%), and ADHD hyperactivity (17% vs 5%) when comparing overtreated (n = 40) and untreated (N = 100), respectively. We identified significantly higher mean scores for SDQ conduct (adjusted mean difference [AMD] 0.74; 95% confidence interval [CI], 0.021-1.431; P = 0.040, effect size 0.018) and ADHD hyperactivity (AMD 1.60, 95% CI, 0.361-2.633; P = 0.003, effect size 0.028) comparing overtreated with normal-GTF children. CONCLUSIONS: There was no overall association between SGTF and offspring ADHD, ASD, or behavior questionnaire scores. However, children of "overtreated" mothers displayed significantly more ADHD symptoms and behavioral difficulties than those of normal-GTF mothers. Thyroxine supplementation during pregnancy requires monitoring to avoid overtreatment.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad/tratamiento farmacológico , Conducta Infantil/efectos de los fármacos , Hipotiroidismo/fisiopatología , Madres , Diagnóstico Prenatal/métodos , Efectos Tardíos de la Exposición Prenatal/tratamiento farmacológico , Tiroxina/administración & dosificación , Adulto , Trastorno por Déficit de Atención con Hiperactividad/diagnóstico , Trastorno por Déficit de Atención con Hiperactividad/epidemiología , Biomarcadores/análisis , Estudios de Casos y Controles , Niño , Preescolar , Estudios de Cohortes , Femenino , Estudios de Seguimiento , Humanos , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal/diagnóstico , Efectos Tardíos de la Exposición Prenatal/epidemiología , Pronóstico , Encuestas y Cuestionarios , Pruebas de Función de la Tiroides , Reino Unido/epidemiología
15.
J Dev Orig Health Dis ; 11(4): 427-437, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-31525320

RESUMEN

Prenatal exposure to persistent organic pollutants (POPs) has been associated with the development of metabolic syndrome-related diseases in offspring. According to epidemiological studies, father's transmission of environmental effects in addition to mother's can influence offspring health. Moreover, maternal prenatal dietary folic acid (FA) may beneficially impact offspring health. The objective is to investigate whether prenatal FA supplementation can overcome the deleterious effects of prenatal exposure to POPs on lipid homeostasis and inflammation in three generations of male rat descendants through the paternal lineage. Female Sprague-Dawley rats (F0) were exposed to a POPs mixture (or corn oil) +/- FA supplementation for 9 weeks before and during gestation. F1 and F2 males were mated with untreated females. Plasma and hepatic lipids were measured in F1, F2, and F3 males after 12-h fast. Gene expression of inflammatory cytokines was determined by qPCR in epididymal adipose tissue. In F1 males, prenatal POPs exposure increased plasma lipids at 14 weeks old and hepatic lipids at 28 weeks old and prenatal FA supplementation decreased plasma total cholesterol at 14 weeks old. Prenatal POPs exposure decreased plasma triglycerides at 14 weeks old in F2 males. No change was observed in inflammatory markers. Our results show an impact of the paternal lineage on lipid homeostasis in rats up to the F2 male generation. FA supplementation of the F0 diet, regardless of POPs exposure, lowered plasma cholesterol in F1 males but failed to attenuate the deleterious effects of prenatal POPs exposure on plasma and hepatic lipids in F1 males.


Asunto(s)
Suplementos Dietéticos , Contaminantes Ambientales/toxicidad , Ácido Fólico/administración & dosificación , Inflamación/patología , Lípidos/análisis , Exposición Materna/efectos adversos , Efectos Tardíos de la Exposición Prenatal/patología , Animales , Animales Recién Nacidos , Femenino , Homeostasis , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/tratamiento farmacológico , Ratas , Ratas Sprague-Dawley
16.
J Dev Orig Health Dis ; 11(4): 419-426, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-31735181

RESUMEN

OBJECTIVE: Maternal obesity is associated with obesity and metabolic disorders in offspring. However, there remains a paucity of data on strategies to reverse the effects of maternal obesity on maternal and offspring health. With maternal undernutrition, taurine supplementation improves outcomes in offspring mediated in part via improved glucose-insulin homeostasis. The efficacy of taurine supplementation in the setting of maternal obesity on health and well-being of offspring is unknown. We examined the effects of taurine supplementation on outcomes related to growth and metabolism in offspring in a rat model of maternal obesity. DESIGN: Wistar rats were randomised to: 1) control diet during pregnancy and lactation (CON); 2) CON with 1.5% taurine in drinking water (CT); 3) maternal obesogenic diet (MO); or 4) MO with taurine (MOT). Offspring were weaned onto the control diet for the remainder of the study. RESULTS: At day 150, offspring body weights and adipose tissue weights were increased in MO groups compared to CON. Adipose tissue weights were reduced in MOT versus MO males but not females. Plasma fasting leptin and insulin were increased in MO offspring groups but were not altered by maternal taurine supplementation. Plasma homocysteine concentrations were reduced in all maternal taurine-supplemented offspring groups. There were significant interactions across maternal diet, taurine supplementation and sex for response to an oral glucose tolerance test , a high-fat dietary preference test and pubertal onset in offspring. CONCLUSIONS: These results demonstrate that maternal taurine supplementation can partially ameliorate adverse developmental programming effects in offspring in a sex-specific manner.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Suplementos Dietéticos , Fructosa/toxicidad , Enfermedades Metabólicas/tratamiento farmacológico , Obesidad Materna/fisiopatología , Efectos Tardíos de la Exposición Prenatal/tratamiento farmacológico , Taurina/administración & dosificación , Animales , Animales Recién Nacidos , Femenino , Masculino , Enfermedades Metabólicas/etiología , Enfermedades Metabólicas/patología , Embarazo , Efectos Tardíos de la Exposición Prenatal/etiología , Efectos Tardíos de la Exposición Prenatal/patología , Ratas , Ratas Wistar
17.
J Agric Food Chem ; 67(48): 13269-13281, 2019 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-31725275

RESUMEN

We studied the long-term influence of gestational diabetes mellitus (GDM) on the pancreas of offspring and the effect of omega-3 polyunsaturated fatty acids (n-3 PUFAs) on offspring's pancreas. GDM offspring were divided into three groups: GDM offspring, n-3 PUFA-adequate-GDM offspring, and n-3 PUFA-deficient GDM offspring. All healthy and GDM offspring were fed up to 11 months old. The pancreas of GDM offspring exhibited fatty infiltration at 11 months old, whereas n-3 PUFA improved the pancreatic fatty infiltration. n-3 PUFA lowered the pancreatic oxidative stress and inflammation. Surprisingly, n-3 PUFA postponed pancreatic telomere shortening of GDM offspring at old age. Nontargeted metabolomics showed that many metabolites were altered in the pancreas of GDM offspring at old age, including l-valine, ceramide, acylcarnitines, tocotrienol, cholesteryl acetate, and biotin. n-3 PUFA modulated some altered metabolites and metabolic pathways. Therefore, GDM caused the long-term effects on offspring's pancreas, whereas n-3 PUFA played a beneficial role.


Asunto(s)
Diabetes Gestacional/tratamiento farmacológico , Ácidos Grasos Omega-3/administración & dosificación , Páncreas/metabolismo , Efectos Tardíos de la Exposición Prenatal/tratamiento farmacológico , Animales , Diabetes Gestacional/metabolismo , Grasas/metabolismo , Femenino , Humanos , Masculino , Metabolómica , Páncreas/química , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Ratas , Ratas Wistar , Telómero/metabolismo
18.
Clin Exp Pharmacol Physiol ; 46(12): 1151-1165, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31410879

RESUMEN

Maternal salt overload programs cardiovascular and renal alterations in the offspring. However, beneficial and harmful effects of high dose vitamin E supplementation have been described in humans and animals. We investigated the hypothesis as to whether cardiac and renal alterations can be programmed by gestational salt overload, and can become further modified during lactation and after weaning. Male Wistar rats were used, being the offspring of mothers that drank either tap water or 0.3 mol/L NaCl for 20 days before and during pregnancy. α-Tocopherol (0.35 g/kg) was administered to mothers daily during lactation or to their offspring for 3 weeks post-weaning. Systolic blood pressure (tcSBP) was measured in juvenile rats aged 210 days. The response of mean arterial pressure (MAP) and heart rate (HR) to intravenous infusion of angiotensin II (Ang II) was also examined. Left ventricle plasma membrane (PMCA) and sarcoplasmic reticulum Ca2+ -ATPase (SERCA) activities, and certain parameters of renal function, were measured. Maternal saline programmed for increased body mass and kidney mass/body mass ratio, increased tcSBP, increased mean arterial pressure and heart rate with anomalous response to infused Ang II. In the heart, saline increased PMCA and α-Tocopherol per se increased PMCA/SERCA. In the kidney, the most remarkable result was the silent saline programming of CrCl , which was sensitized for a sharp decrease after α-Tocopherol. In conclusion, the combination of maternal saline overload and high α-Tocopherol immediately after birth leads to simultaneous cardiovascular and renal alterations in the young offspring, like those encountered in type V cardiorenal syndrome.


Asunto(s)
Desarrollo Embrionario/efectos de los fármacos , Corazón/efectos de los fármacos , Riñón/efectos de los fármacos , Lactancia/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal/tratamiento farmacológico , Cloruro de Sodio Dietético/efectos adversos , alfa-Tocoferol/administración & dosificación , Animales , Esquema de Medicación , Ingestión de Alimentos/fisiología , Femenino , Crecimiento y Desarrollo/efectos de los fármacos , Corazón/fisiología , Riñón/fisiología , Masculino , Fenómenos Fisiologicos Nutricionales Maternos , Embarazo , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Ratas , Ratas Wistar , Factores Sexuales , Cloruro de Sodio Dietético/administración & dosificación , Destete , alfa-Tocoferol/farmacología
19.
Amino Acids ; 51(5): 805-811, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30879150

RESUMEN

We recently reported that dietary supplementation with L-proline (proline) during gestation improved embryonic survival in C57BL/6J mice. The objective of the present study was to test the hypothesis that the effect of maternal proline supplementation on embryonic survival can be carried forward to the first generation female offspring. In the F0 generation, pregnant dams were fed a purified diet supplemented with 0 (control) or 5 g proline/kg diet. The F1 female adult offsprings were bred to fertile males. Fetal survival at embryonic day (E)12.5 and reproductive outcomes at term birth were recorded. The concentrations of amino acids, ammonia, and urea in plasma and amniotic fluid, as well as concentrations of polyamines in placental tissues and amniotic fluid at E12.5 were determined. Results showed that the F1 generation female offspring from proline-supplemented dams had higher (P < 0.05) concentrations of glutamate and taurine in plasma; of putrescine and spermidine in placental tissues; and of glycine, taurine, and spermidine in amniotic fluid at E12.5, as compared with F1 generation female offsprings from dams without proline supplementation. Concentration of proline in the plasma of offspring mice from proline-supplemented dams were lower (P < 0.05), as compared with the control group. No differences in fetal survival, reproductive outcomes, or concentrations of ammonia and urea in plasma and amniotic fluid were observed between the two groups of F1 female offspring. Collectively, our results indicate that the benefits of maternal proline supplementation during gestation on improving embryonic survival and fetal growth in F0 females are not transmitted to their F1 generation females.


Asunto(s)
Aminoácidos/metabolismo , Suplementos Dietéticos , Desarrollo Fetal/efectos de los fármacos , Placenta/metabolismo , Poliaminas/metabolismo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Prolina/administración & dosificación , Líquido Amniótico/efectos de los fármacos , Líquido Amniótico/metabolismo , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Placenta/efectos de los fármacos , Embarazo , Efectos Tardíos de la Exposición Prenatal/tratamiento farmacológico
20.
J Neonatal Perinatal Med ; 12(1): 109-112, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30149482

RESUMEN

BACKGROUND: Mitragyna speciosa, also known as kratom, is obtained from the coffee plant family 'Rubiaceae.' Kratom is available in the form of capsules, whole, processed and powdered leaves, and as liquids. Secondary to its 'natural herb' status and opioid effects, it is misconceived to be a safe alternative for the treatment of chronic pain. The use of kratom has increased by tenfold in the United States since 2010. METHODS AND RESULTS: We report a term neonate who was born to a chronic kratom user and required treatment with opiates for neonatal drug withdrawal. CONCLUSION: Physicians should be aware of these herbal supplements and its potential withdrawal effects in newborn which cannot be picked up by the standard toxicology screen.


Asunto(s)
Analgésicos Opioides/efectos adversos , Dolor de la Región Lumbar/tratamiento farmacológico , Mitragyna/efectos adversos , Síndrome de Abstinencia Neonatal/tratamiento farmacológico , Complicaciones del Embarazo/tratamiento farmacológico , Efectos Tardíos de la Exposición Prenatal/tratamiento farmacológico , Adulto , Femenino , Humanos , Recién Nacido , Mitragyna/química , Síndrome de Abstinencia Neonatal/fisiopatología , Manejo del Dolor/métodos , Fitoterapia , Embarazo , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Automedicación , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA