Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
1.
Mol Neurobiol ; 58(10): 5210-5223, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34272687

RESUMEN

Fetal alcohol syndrome (FAS) is characterized by disrupted fetal brain development and postnatal cognitive impairment. The targets of alcohol are diverse, and it is not clear whether there are common underlying molecular mechanisms producing these disruptions. Prior work established that acute ethanol exposure causes a transient increase in tyrosine phosphorylation of multiple proteins in cultured embryonic cortical cells. In this study, we show that a similar tyrosine phosphorylation transient occurs in the fetal brain after maternal dosing with ethanol. Using phospho-specific antibodies and immunohistochemistry, we mapped regions of highest tyrosine phosphorylation in the fetal cerebral cortex and found that areas of dendritic and axonal growth showed elevated tyrosine phosphorylation 10 min after maternal ethanol exposure. These were also areas of Src expression and Src family kinase (SFK) activation loop phosphorylation (pY416) expression. Importantly, maternal pretreatment with the SFK inhibitor dasatinib completely prevents both the pY416 increase and the tyrosine phosphorylation response. The phosphorylation response was observed in the perisomatic region and neurites of immature migrating and differentiating primary neurons. Importantly, the initial phosphotyrosine transient (~ 30 min) targets both Src and Dab1, two critical elements in Reelin signaling, a pathway required for normal cortical development. This initial phosphorylation response is followed by sustained reduction in Ser3 phosphorylation of n-cofilin, a critical actin severing protein and an identified downstream effector of Reelin signaling. This biochemical disruption is associated with sustained reduction of F-actin content and disrupted Golgi apparatus morphology in developing cortical neurons. The finding outlines a model in which the initial activation of SFKs by ethanol has the potential to disrupt multiple developmentally important signaling systems for several hours after maternal exposure.


Asunto(s)
Corteza Cerebral/embriología , Corteza Cerebral/enzimología , Desarrollo Embrionario/efectos de los fármacos , Etanol/toxicidad , Efectos Tardíos de la Exposición Prenatal/enzimología , Familia-src Quinasas/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Desarrollo Embrionario/fisiología , Femenino , Ratones , Ratones Endogámicos C57BL , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente
2.
Chem Biol Interact ; 333: 109314, 2021 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-33171135

RESUMEN

Arsenic is a pollutant widely found in the environment due to natural and anthropogenic sources. Exposure to arsenic forms in drinking water has been related with male reproductive dysfunctions in humans and experimental animals at adult age. However, the impact of this pollutant on postnatal reproductive development of male offspring exposed in utero to arsenic is still unknown. Therefore, this study aimed to investigate the effects of prenatal arsenic exposure on the postnatal development of the testes and epididymides of rats, during prepuberty. For this purpose, pregnant female Wistar rats were provided drinking water containing 0 or 10 mg/L sodium arsenite (AsNaO2) from gestational day 1 (GD 1) until GD 21 and the male offspring was evaluated in different periods of prepuberty. Our results showed that prenatal arsenic exposure affected the initial sexual development of male pups, reducing their body weight and relative anogenital distance at postnatal day 1. At different periods of prepuberty, male pups from arsenic exposed dams showed a reduction of body and reproductive organs weights, testosterone levels and testis morphometric parameters. Moreover, these pups presented changes in the expression of SOD1, SOD2, CAT and GSTK1 genes and in the activity of superoxide dismutase, catalase and glutathione s-transferase in the testes and epididymides during prepuberty. Taken together, our results show that prenatal arsenic exposure provoked reproductive disorders in prepubertal male rats, probably due to reproductive reprograming and oxidative stress induced by this pollutant.


Asunto(s)
Arsénico/toxicidad , Epidídimo/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal/metabolismo , Testículo/efectos de los fármacos , Animales , Antioxidantes/metabolismo , Peso Corporal/efectos de los fármacos , Epidídimo/metabolismo , Epidídimo/patología , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Masculino , Tamaño de los Órganos/efectos de los fármacos , Embarazo , Efectos Tardíos de la Exposición Prenatal/enzimología , Efectos Tardíos de la Exposición Prenatal/patología , Pubertad , Ratas , Ratas Wistar , Testículo/metabolismo , Testículo/patología
3.
BMC Anesthesiol ; 20(1): 228, 2020 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-32894054

RESUMEN

BACKGROUND: Bupivacaine, an amid-type local anesthetic, is widely used for clinical patients especially in pregnant women. In addition to neurotoxicity effect of bupivacaine, it can cross the placenta, accumulates in this tissue and retained in fetal tissues. Nevertheless, whether bupivacaine can cause neurotoxicity in fetus remains unclear. Hence, this study was design to investigate the effects of maternal bupivacaine use on fetus hippocampal cell apoptosis and the possible related mechanism. METHODS: On day 15 of pregnancy, sciatic nerve of pregnant wistar rat (180-200 g) were exposed by lateral incision of the right thigh and 0.2 ml of bupivacaine was injected. After their delivery, we randomly selected one male offspring of every mother. On day 30 after of their birth, the rat's hippocampi were isolated for molecular studies. Western blotting was used to examine the expression of cleaved caspase-3, caspase-8 and p-Akt in fetal hippocampus. RESULTS: Our results showed that maternal bupivacaine use caused a significant increment of cleaved caspase-3 and caspase-8 expression in fetal hippocampus compared with the sham group. In addition, maternally administered bupivacaine could significantly decrease hippocampal P.Akt/T.Akt ratio which was concurrent with an increment of cleaved caspase-3 and caspase-8 expression. CONCLUSION: Our data suggest that maternal bupivacaine use increases fetal hippocampal cell apoptosis markers such as caspase 8 and cleaved caspase 3, at least in part, via inhibiting the Akt activation.


Asunto(s)
Anestésicos Locales/toxicidad , Apoptosis/efectos de los fármacos , Bupivacaína/toxicidad , Hipocampo/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Nervio Ciático/efectos de los fármacos , Anestésicos Locales/administración & dosificación , Animales , Animales Recién Nacidos , Apoptosis/fisiología , Bupivacaína/administración & dosificación , Caspasa 3/biosíntesis , Caspasa 8/biosíntesis , Femenino , Hipocampo/enzimología , Hipocampo/patología , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal/enzimología , Efectos Tardíos de la Exposición Prenatal/patología , Ratas , Ratas Wistar , Nervio Ciático/enzimología , Nervio Ciático/patología
4.
Eur Neuropsychopharmacol ; 35: 100-110, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32439226

RESUMEN

In the central nervous system, activated microglia and astrocytes produce proinflammatory mediators such as inducible nitric oxide (iNOS) and cytokines. Uncontrolled release of these mediators induced by immune challenge can lead to increased vulnerability to complex brain disorders such as schizophrenia. In this study, BALB/c mice were injected intraperitoneally (i.p) with the viral mimetic polyriboinosinic-polyribocytidilic acid (poly(I:C)) or saline. At postnatal day 30 (PND0), the animals were sacrificed and the hippocampus, corpus callosum, striatum, cortex, fimbria and ventricle were immunostained for Iba-1, a microglial marker, glial fibrillary acidic protein (GFAP), an astrocyte marker, and iNOS, an activation marker for NO. Additionally, serum cytokine profiling (Interleukin-2 (IL-2), IL- 4, IL-6, interferon gamma (IFN-γ), tumour necrosis factor (TNF), IL-17A and IL-10) was determined using serum samples from poly(I:C)-treated and control mice. Our results demonstrated that poly(I:C) induced overactivation of differential proinflammatory responses in microglia and astrocytes, which could be strongly enhanced by a postnatal poly(I:C) administration before PND 30 in one part of the animals investigated. Specifically, there was significant iNOS upregulation in hippocampus, cortex and corpus callosum of poly(I:C)-affected off-springs. These inflammatory alterations were accompanied by increased circulating levels of the proinflammatory cytokines tumour necrosis factor alpha (TNF-α) and interleukin-6 (IL-6). This study provides insight into the role of microglia and astrocytes in an animal model of schizophrenia and an understanding of the regulation of iNOS expression in glial cells and cytokine networks. This knowledge could help identify novel targets for anti-oxidative and anti-inflammatory therapeutic schizophrenia intervention.


Asunto(s)
Astrocitos/enzimología , Microglía/enzimología , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Poli I-C/toxicidad , Efectos Tardíos de la Exposición Prenatal/enzimología , Esquizofrenia/enzimología , Animales , Animales Recién Nacidos , Astrocitos/efectos de los fármacos , Astrocitos/inmunología , Modelos Animales de Enfermedad , Inducción Enzimática/efectos de los fármacos , Inducción Enzimática/fisiología , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Microglía/efectos de los fármacos , Microglía/inmunología , Óxido Nítrico Sintasa de Tipo II/inmunología , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/inmunología , Esquizofrenia/inducido químicamente , Esquizofrenia/inmunología
5.
Neurochem Res ; 45(7): 1626-1635, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32285378

RESUMEN

Repeated or prolonged use of general anesthetics in pregnant women may disturb the neurodevelopment of infants. Compelling evidence indicates that maternal exercise during pregnancy has positive effects on the cognitive function of offspring. We previously confirmed the preventive potential of maternal treadmill training for cognitive deficits induced by in utero exposure to sevoflurane in rat pups. However, the underlying mechanism(s) needed further clarification. Thus, the aim of the present study was to investigate the effects of maternal exercise on the epigenetic regulation of genes linked to brain plasticity and function. Pregnant rats on gestational day 13 (GD 13) received 2 h of 3% sevoflurane or 30% oxygen daily on three consecutive days (GD 13-15). Pregnant rats in the exercise groups were forced to run on a treadmill for 60 min/day, 5 days/week, for a duration of 3 weeks. On postnatal day 0 (PND 0), the brains of rat pups were harvested for biochemical and histochemical studies. On PNDs 28-33, the learning and memory ability of rat pups was assessed using Morris water maze task. Maternal exercise ameliorated sevoflurane-induced decreases in p300 histone acetyltransferase (HAT) expression and inhibition of BDNF signaling. Maternal exercise improved performance in the Morris water maze task. However, these effects were reversed by p300 inhibitor. Our results indicated that maternal treadmill exercise during pregnancy can ameliorate cognitive dysfunction induced by prenatal sevoflurane exposure; p300 HAT-mediated BDNF signaling activation might contribute to the neuroprotective effects of maternal exercise.


Asunto(s)
Anestésicos por Inhalación/toxicidad , Proteína p300 Asociada a E1A/metabolismo , Prueba de Esfuerzo/tendencias , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/enzimología , Sevoflurano/toxicidad , Animales , Animales Recién Nacidos , Femenino , Histona Acetiltransferasas/metabolismo , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal/prevención & control , Ratas , Ratas Sprague-Dawley
6.
Biomolecules ; 10(3)2020 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-32182821

RESUMEN

Exposure of neural cells to harmful and toxic factors promotes oxidative stress, resulting in disorders of metabolism, cell differentiation, and maturation. The study examined the brains of rats pre- and postnatally exposed to sodium fluoride (NaF 50 mg/L) and activity of NADPH oxidase 4 (NOX4), catalase (CAT), superoxide dismutase (SOD), glutathione peroxidase (GPx), glutathione reductase (GR), concentration of glutathione (GSH), and total antioxidant capacity (TAC) in the cerebellum, prefrontal cortex, hippocampus, and striatum were measured. Additionally, NOX4 expression was determined by qRT-PCR. Rats exposed to fluorides (F-) showed an increase in NOX4 activity in the cerebellum and hippocampus, a decrease in its activity in the prefrontal cortex and hippocampus, and upregulation of NOX4 expression in hippocampus and its downregulation in other brain structures. Analysis also showed significant changes in the activity of all antioxidant enzymes and a decrease in TAC in brain structures. NOX4 induction and decreased antioxidant activity in central nervous system (CNS) cells may be central mechanisms of fluoride neurotoxicity. NOX4 contributes to blood-brain barrier damage, microglial activation, and neuronal loss, leading to impairment of brain function. Fluoride-induced oxidative stress involves increased reactive oxygen speciaes (ROS) production, which in turn increases the expression of genes encoding pro-inflammatory cytokines.


Asunto(s)
Encéfalo/enzimología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Glutatión/metabolismo , NADPH Oxidasa 4/biosíntesis , Síndromes de Neurotoxicidad/enzimología , Efectos Tardíos de la Exposición Prenatal/enzimología , Fluoruro de Sodio/toxicidad , Regulación hacia Arriba/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Femenino , Estrés Oxidativo/efectos de los fármacos , Embarazo , Efectos Tardíos de la Exposición Prenatal/patología , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo
7.
Horm Res Paediatr ; 91(4): 262-270, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31256164

RESUMEN

BACKGROUND: The HIV drugs lopinavir and ritonavir have recently been reported to cause transient adrenal insufficiency in preterm newborns. We, therefore, considered HIV drugs as a cause of transiently elevated 17-hydroxyprogesterone (17OHP) levels in a neonatal screening test for congenital adrenal hyperplasia in a preterm girl exposed to zidovudine, efavirenz, tenofovir, and emtricitabine. OBJECTIVE: So far, HIV drugs have not been tested for their effect on steroidogenesis and the steroidogenic enzyme activity of CYP21A2 specifically in an in vitro system. METHODS: We tested the effect of efavirenz, tenofovir, emtricitabine, and zidovudine on steroidogenesis of human adrenal H295R cells. Cells were treated with the drugs at different concentrations including concentrations in therapeutic use. The effect on CYP21A2 activity was assessed by testing the conversion of radiolabeled 17OHP to 11-deoxycortisol. Cell viability was tested by an MTT assay. In addition, recombinant human CYP21A2 protein was used to assess direct drug effects on CYP21A2 activity. RESULTS: We observed significantly decreased CYP21A2 activity in both in vitro testing systems after treatment with efavirenz at therapeutic concentrations. Moreover, efavirenz affected cell viability. By contrast, the other test drugs did not affect steroidogenesis. Follow-up of our patient revealed elevated 17OHP and androgen levels during the first weeks of life, but values normalized spontaneously. Genetic testing for CYP21A2 mutations was negative. Thus, it remains unsettled whether the transient 17OHP elevation in this baby was due to a drug effect. CONCLUSION: The HIV drug efavirenz inhibits CYP21A2 activity in vitro through direct interaction with enzyme catalysis at therapeutic concentrations. This may have clinical implications for HIV treatment in children and adults. However, so far, clinical data are scarce, and further studies are needed to be able to draw clinical conclusions.


Asunto(s)
Hiperplasia Suprarrenal Congénita , Benzoxazinas , Infecciones por VIH/tratamiento farmacológico , Complicaciones Infecciosas del Embarazo/tratamiento farmacológico , Nacimiento Prematuro , Efectos Tardíos de la Exposición Prenatal , Inhibidores de la Transcriptasa Inversa , Esteroide 21-Hidroxilasa/antagonistas & inhibidores , Hiperplasia Suprarrenal Congénita/inducido químicamente , Hiperplasia Suprarrenal Congénita/enzimología , Adulto , Alquinos , Benzoxazinas/administración & dosificación , Benzoxazinas/efectos adversos , Línea Celular , Ciclopropanos , Femenino , Humanos , Recién Nacido , Embarazo , Nacimiento Prematuro/inducido químicamente , Nacimiento Prematuro/enzimología , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/enzimología , Inhibidores de la Transcriptasa Inversa/administración & dosificación , Inhibidores de la Transcriptasa Inversa/efectos adversos , Esteroide 21-Hidroxilasa/metabolismo
8.
Neurotoxicology ; 74: 81-90, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31175943

RESUMEN

BACKGROUND: Sixty percent of the mammalian brain is composed of lipids including arachidonic acid (AA). AA released from cell membranes is metabolised in the cyclooxygenase (COX) pathway to prostanoids - biologically active substances involved in the regulation of many processes including inflammation. It has been shown that long-term exposure to fluoride in pre and neonatal period is dangerous because this element is able to penetrate through the placenta and to cross the blood-brain barrier. Exposure to fluoride during the development affects metabolism and physiology of neurons and glia which results in the impairment of cognitive functions but the exact mechanisms of fluoride neurotoxicity are not clearly defined. OBJECTIVE: The aim of this study was to determine whether exposure to fluoride during the development affects COXes activity and the synthesis of prostanoids. MATERIAL AND METHODS: Pre- and postnatal toxicity model in Wistar rats was used. Experimental animals received 50 mg/L of NaF in drinking water ad libitum, while control animals received tap water. In cerebral cortex, hippocampus, cerebellum and striatum were measured fluoride concentration, COX1 and COX2 genes expression, immunolocalization of the enzymatic proteins and concentration of PGE2 and TXB2. RESULTS: of this study showed statistically significant changes in the concentration of fluoride in brain structures between study group and control animals. Moreover, significant changes in the expression level of COX1 and COX2, and in the concentration of PGE2 and TXB2 were observed. CONCLUSION: Exposure to fluoride in the prenatal and neonatal period result in the increase in COX2 activity and increase in PGE2 concentration in rats brain, which may lead to disturbances in central nervous system homeostasis.‬‬.


Asunto(s)
Ciclooxigenasa 1/biosíntesis , Ciclooxigenasa 1/genética , Ciclooxigenasa 2/biosíntesis , Ciclooxigenasa 2/genética , Fluoruros/toxicidad , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Síndromes de Neurotoxicidad/enzimología , Síndromes de Neurotoxicidad/genética , Animales , Animales Recién Nacidos , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Encéfalo/metabolismo , Dinoprostona/biosíntesis , Femenino , Fluoruros/farmacocinética , Embarazo , Efectos Tardíos de la Exposición Prenatal/enzimología , Efectos Tardíos de la Exposición Prenatal/genética , Prostaglandinas/biosíntesis , Ratas , Ratas Wistar , Tromboxano B2/biosíntesis
9.
Neurosci Res ; 147: 9-16, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30452948

RESUMEN

Accumulating evidence from preclinical and clinical studies indicates prenatal exposure to stress or excess glucocorticoids can affect offspring brain. HDAC2 is an important target of glucocorticoid. Here we detected HDAC2 expression in male offspring hippocampus from gestational restraint stressed rat during development and the relationship between HDAC2 expression and behaviors and neurogenesis in male offspring. Pregnant rats received restrained stress during the last week of pregnancy. Expressions of HDAC2 in offspring hippocampus were detected on postnatal 0 day (P0) and 60 days (P60). Neurogenesis was evaluated by Doublecortin (DCX) staining on P60. Anxiety-like behavior and cognition were detected in open field, elevated plus maze, novel object recognition test, and Barnes maze. We found that HDAC2 expression in the hippocampus of male prenatally stressed offspring (MPSO) was similar to the male control offspring on P0, but significantly lower on P60. Corresponding to the decreased expression of HDAC2 in MPSO hippocampus at P60, neurogenesis in the dentate gyrus of MPSO was significantly lower than the control male offspring. And MPSO also showed greater anxiety and poorer learning and memories abilities than control male offspring. These showed that HDAC2 could partly explain the effects of gestational stress on male offspring behaviors.


Asunto(s)
Hipocampo/embriología , Hipocampo/enzimología , Histona Desacetilasa 2/metabolismo , Complicaciones del Embarazo/enzimología , Efectos Tardíos de la Exposición Prenatal/enzimología , Animales , Ansiedad/enzimología , Giro Dentado/embriología , Giro Dentado/enzimología , Proteína Doblecortina , Femenino , Aprendizaje/fisiología , Masculino , Memoria/fisiología , Actividad Motora/fisiología , Neurogénesis , Embarazo , Complicaciones del Embarazo/psicología , Ratas , Ratas Sprague-Dawley , Restricción Física , Estrés Psicológico/metabolismo
10.
Neurotoxicology ; 70: 180-186, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30385389

RESUMEN

Lead (Pb) is a developmental neurotoxicant. We have demonstrated that perinatally Pb-exposed rats consume more ethanol than their control counterparts, a response that seems to be mediated by catalase (CAT) and centrally-formed acetaldehyde, ethanol's first metabolite with attributed reinforcing effects in the brain. The present study sought to disrupt ethanol intake (2-10% ethanol v/v) in rats exposed to 220 ppm Pb or filtered water during gestation and lactation. Thus, to block brain CAT expression, a lentiviral vector coding for a shRNA against CAT (LV-antiCAT vector) was microinfused in the posterior ventral tegmental area (pVTA) either at the onset or towards the end of a chronic voluntary ethanol consumption test. At the end of the study, rats were euthanized and pVTA dissected to measure CAT expression by Western blot. The LV-antiCAT vector administration not only reversed, but also prevented the emergence of the elevated ethanol intake reported in the perinatally Pb-exposed animals, changes that were supported by a significant reduction in CAT expression in the pVTA. These results provide further evidence of the crucial role of this enzyme in the reinforcing properties of ethanol and in the impact of the perinatal Pb programming to challenging events later in life.


Asunto(s)
Consumo de Bebidas Alcohólicas/prevención & control , Encéfalo/enzimología , Catalasa/biosíntesis , Etanol/toxicidad , Plomo/toxicidad , Efectos Tardíos de la Exposición Prenatal/enzimología , Consumo de Bebidas Alcohólicas/efectos adversos , Animales , Encéfalo/efectos de los fármacos , Catalasa/antagonistas & inhibidores , Catalasa/genética , Etanol/administración & dosificación , Femenino , Regulación Enzimológica de la Expresión Génica , Plomo/administración & dosificación , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/prevención & control , Ratas , Ratas Wistar
11.
Hippocampus ; 29(2): 73-77, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30311334

RESUMEN

A combination of genetic and environmental factors contributes to schizophrenia (SZ), a catastrophic psychiatric disorder with a hypothesized neurodevelopmental origin. Increases in the brain levels of the tryptophan metabolite kynurenic acid (KYNA), an endogenous antagonist of α7 nicotinic acetylcholine and NMDA receptors, have been implicated specifically in the cognitive deficits seen in persons with SZ. Here we evaluated this role of KYNA by adding the KYNA precursor kynurenine (100 mg/day) to chow fed to pregnant rat dams from embryonic day (ED) 15 to ED 22 (control: ECon; kynurenine treated: EKyn). Upon termination of the treatment, all rats received normal rodent chow until the animals were evaluated in adulthood (postnatal days 56-85). EKyn treatment resulted in increased extracellular KYNA and reduced extracellular glutamate in the hippocampus, measured by in vivo microdialysis, and caused impairments in hippocampus-dependent learning in adult rats. Acute administration of BFF816, a systemically active inhibitor of kynurenine aminotransferase II (KAT II), the major KYNA-synthesizing enzyme in the brain, normalized neurochemistry and prevented contextual memory deficits in adult EKyn animals. Collectively, these results demonstrate that acute inhibition of KYNA neosynthesis can overcome cognitive impairments that arise as a consequence of elevated brain KYNA in early brain development.


Asunto(s)
Compuestos Heterocíclicos con 3 Anillos/uso terapéutico , Hipocampo/efectos de los fármacos , Quinurenina/toxicidad , Trastornos de la Memoria/enzimología , Efectos Tardíos de la Exposición Prenatal/enzimología , Tiazolidinedionas/uso terapéutico , Transaminasas/antagonistas & inhibidores , Factores de Edad , Animales , Femenino , Compuestos Heterocíclicos con 3 Anillos/farmacología , Hipocampo/enzimología , Masculino , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/tratamiento farmacológico , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/tratamiento farmacológico , Ratas , Ratas Wistar , Tiazolidinedionas/farmacología , Transaminasas/metabolismo
12.
J Appl Toxicol ; 39(4): 603-621, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30561030

RESUMEN

Polychlorinated biphenyls (PCBs) are toxic environmental pollutants. Humans are exposed to PCB mixtures via contaminated food or water. PCB exposure causes adverse effects in adults and after exposure in utero. PCB toxicity depends on the congener mixture and CYP1A2 gene activity. For coplanar PCBs, toxicity depends on ligand affinity for the aryl hydrocarbon receptor (AHR). Previously, we found that perinatal exposure of mice to a three-coplanar/five-noncoplanar PCB mixture induced deficits in novel object recognition and trial failures in the Morris water maze in Cyp1a2-/- ::Ahrb1 C57BL6/J mice compared with wild-type mice (Ahrb1  = high AHR affinity). Here we exposed gravid Cyp1a2-/- ::Ahrb1 mice to a PCB mixture on embryonic day 10.5 by gavage and examined the F1 and F3 offspring (not F2 ). PCB-exposed F1 mice exhibited increased open-field central time, reduced acoustic startle, greater conditioned contextual freezing and reduced CA1 hippocampal long-term potentiation with no change in spatial learning or memory. F1 mice also had inhibited growth, decreased heart rate and cardiac output, and impaired fertility. F3 mice showed few effects. Gene expression changes were primarily in F1 PCB males compared with wild-type males. There were minimal RNA and DNA methylation changes in the hippocampus from F1 to F3 with no clear relevance to the functional effects. F0 PCB exposure during a period of rapid DNA de-/remethylation in a susceptible genotype produced clear F1 effects with little evidence of transgenerational effects in the F3 generation. While PCBs show clear developmental neurotoxicity, their effects do not persist across generations for effects assessed herein.


Asunto(s)
Citocromo P-450 CYP1A2/metabolismo , Contaminantes Ambientales/toxicidad , Fertilidad/efectos de los fármacos , Potenciación a Largo Plazo/efectos de los fármacos , Bifenilos Policlorados/toxicidad , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Reflejo de Sobresalto/efectos de los fármacos , Memoria Espacial/efectos de los fármacos , Animales , Región CA1 Hipocampal/efectos de los fármacos , Región CA1 Hipocampal/fisiopatología , Condicionamiento Clásico , Citocromo P-450 CYP1A2/genética , Femenino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Embarazo , Efectos Tardíos de la Exposición Prenatal/enzimología , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Efectos Tardíos de la Exposición Prenatal/psicología
13.
Cell Death Dis ; 9(6): 659, 2018 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-29855476

RESUMEN

An imbalance of excitatory and inhibitory signals in the brain has been proposed to be one of the main pathological features of various diseases related to hypothalamic-pituitary-adrenal axis (HPAA) dysfunction. Excessive glutamate release induces neuronal excitotoxicity, while glutamic acid decarboxylase (GAD) 67 promotes the transformation of excessive glutamate to γ-aminobutyric acid (GABA). Our previous studies demonstrated that prenatal ethanol exposure (PEE) causes foetal over-exposure to maternal corticosterone and hypersensitivity of the HPAA after birth, but its intrauterine programming mechanism is unknown. In this study, PEE was shown to lead to an enhanced potential excitatory ability of the hypothalamus and hypersensitivity of the HPAA, as well as mild abnormal hippocampal morphology, demethylation of the -1019 to -691-bp region in the hippocampal GAD67 promoter and upregulation of GAD67 expression accompanied by a reduction in glutamatergic neurons and increase in GABAergic neurons in PEE male offspring. Similar changes were also found in PEE male foetal rats. Furthermore, corticosterone increased the expression of the glucocorticoid receptor (GR) and GAD67 in foetal hippocampal H19-7 cells in a concentration-dependent manner, accompanied by demethylation of the GAD67 promoter, a decrease in glutamatergic neurons and increase in GABAergic neurons. The GR inhibitor, mifepristone, reversed the effects of corticosterone on H19-7 cells. These results suggested that PEE-induced excessive corticosterone can lead to upregulation of GAD67 through epigenetic modification mediated by the GR in the male foetal hippocampus, thereby weakening the negative regulation of the HPAA by the hippocampus and increasing the potential excitatory ability of the hypothalamus. These changes persisted until after birth, resulting in hypersensitivity of the HPAA. However, gender differences were observed in the hippocampal development, morphology and GAD67 expression associated with PEE. Programming for the increased expression of hippocampal GAD67 is a potential mechanism responsible for the hypersensitivity of the HPAA in PEE male rats.


Asunto(s)
Etanol/toxicidad , Glutamato Descarboxilasa/metabolismo , Hipocampo/enzimología , Sistema Hipotálamo-Hipofisario/patología , Sistema Hipófiso-Suprarrenal/patología , Efectos Tardíos de la Exposición Prenatal/enzimología , Animales , Línea Celular , Femenino , Feto/efectos de los fármacos , Feto/patología , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Masculino , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Embarazo , Efectos Tardíos de la Exposición Prenatal/patología , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Ácido gamma-Aminobutírico/metabolismo
14.
Cell Death Dis ; 9(6): 638, 2018 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-29844424

RESUMEN

Prenatal dexamethasone exposure (PDE) induces developmental toxicities of multiple organs in offspring. Here, we verified the intergenerational effect of low peak bone mass induced by PDE and investigated its intrauterine programming mechanism. Pregnant rats were injected subcutaneously with 0.2 mg/kg/d dexamethasone from gestation day (GD) 9 to 20. Some pregnant rats were killed for the fetuses on GD20, and the rest went on to spontaneous labor to produce the first-generation (F1) offspring. The adult F1 male offspring were mated with normal females to produce the F2 offspring. In vivo, PDE leads to low peak bone mass in F1 male offspring rats at postnatal week (PW) 28. Furthermore, PDE reduced the bone mass in F1 male offspring from GD20 to PW12. Meanwhile, the osteogenic differentiation was suppressed and the local renin-angiotensin system (RAS) was activated continuously by PDE. Moreover, the histone 3 lysine 27 acetylation (H3K27ac) level in angiotensin-converting enzyme (ACE) promoter region was increased by PDE from GD20 to PW12. Likewise, PDE induced the low peak bone mass and the activated local RAS in F2 male offspring. Meaningfully, the H3K27ac level of ACE was increased by PDE in the F2 offspring. In vitro, dexamethasone inhibited bone marrow mesenchymal stem cells (BMSCs) osteogenic differentiation and promoted RAS activation. Furthermore, dexamethasone recruited CCAAT/enhancer-binding protein α and p300 into the BMSCs nucleus by activating glucocorticoid receptor, which cooperatively increased the H3K27ac level in the ACE promoter region. In conclusion, PDE induced the low peak bone mass and its intergenerational effect, which was mediated by sustained activation of RAS via increasing H3K27ac level of ACE.


Asunto(s)
Huesos/patología , Dexametasona/efectos adversos , Histonas/metabolismo , Lisina/metabolismo , Peptidil-Dipeptidasa A/metabolismo , Efectos Tardíos de la Exposición Prenatal/enzimología , Acetilación , Animales , Diferenciación Celular/efectos de los fármacos , Cruzamientos Genéticos , Femenino , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Osteogénesis/efectos de los fármacos , Linaje , Embarazo , Efectos Tardíos de la Exposición Prenatal/patología , Ratas Wistar , Sistema Renina-Angiotensina/efectos de los fármacos
15.
Mol Pain ; 14: 1744806918777406, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29712513

RESUMEN

Irritable bowel syndrome is a disorder of unknown etiology characterized by widespread, chronic abdominal pain associated with altered bowel movements. Increasing amounts of evidence indicate that stressors presented during gestational periods could have long-term effects on the offspring's tissue structure and function, which may predispose to gastrointestinal diseases. The aim of the present study is to determine whether prenatal maternal stressis a adverse factor affecting gastrointestinal sensitivity and to investigate possible mechanisms underlying prenatal maternal stress-induced visceral hypersensitivity in adult offspring. Prenatal maternal stress was induced in pregnant Sprague-Dawley rats by exposure to heterotypic intermitent stress from gestational day 7 to delivery. Prenatal maternal stress significantly increased visceromotor response to colorectal distention in adult offspring from the age of 6 weeks to 10 weeks. Prenatal maternal stress also enhanced neuronal excitability including depolarization of resting membrane potentials, reduction in rheobase, and an increase in the number of action potentials evoked by 2× and 3× rheobase current stimultion of colon-specific dorsal root ganglion neurons. Prenatal maternal stress remarkably enhanced expression of cystathionine-ß-synthase and Nav1.7 in T13-L2 thoracolumbar dorsal root ganglions both at protein and mRNA levels. Intraperitoneal injection of aminooxyacetic acid, an inhibitor of cystathionine-ß-synthase, attenuated prenatal maternal stress-induced visceral hypersensitivity in a dose-dependent manner. A consecutive seven-day administration of aminooxyacetic acid reversed the hyperexcitability of colon-specific dorsal root ganglion neurons and markedly reduced Nav1.7 expression. These results indicate that the presence of multiple psychophysical stressors during pregnancy is associated with visceral hypersensitivity in offspring, which is likely mediated by an upregualtion of cystathionine-ß-synthase and Nav1.7 expression. Prenatal maternal stress might be a significant contributor to irritable bowel syndrome, and cystathionine-ß-synthase might be a potential target for treatment for chronic visceral hypersensitivity in patients with irritable bowel syndrome.


Asunto(s)
Cistationina betasintasa/metabolismo , Efectos Tardíos de la Exposición Prenatal/enzimología , Células Receptoras Sensoriales/enzimología , Transducción de Señal , Estrés Psicológico/complicaciones , Dolor Visceral/enzimología , Dolor Visceral/etiología , Animales , Células Cultivadas , Colon/inervación , Colon/patología , Cistationina betasintasa/antagonistas & inhibidores , Cistationina betasintasa/genética , Electromiografía , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Femenino , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Ganglios Espinales/patología , Masculino , Canal de Sodio Activado por Voltaje NAV1.7/genética , Canal de Sodio Activado por Voltaje NAV1.7/metabolismo , Especificidad de Órganos , Embarazo , Efectos Tardíos de la Exposición Prenatal/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/patología , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética , Dolor Visceral/patología
16.
Toxicol Sci ; 164(1): 72-84, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29617909

RESUMEN

Globally, approximately 10%-25% of women smoke during pregnancy. Since nicotine is highly addictive, women may use nicotine-containing products like nicotine replacement therapies for smoking cessation, but the long-term consequences of early life exposure to nicotine remain poorly defined. Our laboratory has previously demonstrated that maternal nicotine exposed (MNE) rat offspring exhibit hypertriglyceridemia due to increased hepatic de novo lipogenesis. Hypertriglyceridemia may also be attributed to impaired white adipose tissue (WAT) lipid storage; however, the effects of MNE on WAT are not completely understood. We hypothesize that nicotine-induced alterations in adipose function (eg, lipid storage) underlie dyslipidemia in MNE adults. Female 6-month-old rats exposed to nicotine during gestation and lactation exhibited significantly decreased visceral adipocyte cell area by 40%, attributed, in part, to a 3-fold increase in adipose triglyceride lipase (ATGL) protein expression compared with vehicle. Given ATGL has antioxidant properties and in utero nicotine exposure promotes oxidative stress in various tissues, we next investigated if there was evidence of increased oxidative stress in MNE WAT. At both 3 weeks and 6 months, MNE offspring expressed 37%-48% higher protein levels of superoxide dismutase-1 and -2 in WAT. Since oxidative stress can induce inflammation, we examined the inflammatory profile of WAT and found increased expression of cytokines (interleukin-1ß, tumor necrosis factor α, and interleukin-6) by 44%-61% at 6 months. Collectively, this suggests that the expression of WAT ATGL may be induced to counter MNE-induced oxidative stress and inflammation. However, higher levels of ATGL would further promote lipolysis in WAT, culminating in impaired lipid storage and long-term dyslipidemia.


Asunto(s)
Tejido Adiposo Blanco/efectos de los fármacos , Antioxidantes/metabolismo , Lipasa/genética , Exposición Materna/efectos adversos , Nicotina/toxicidad , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Adipocitos Blancos/efectos de los fármacos , Adipocitos Blancos/enzimología , Tejido Adiposo Blanco/embriología , Tejido Adiposo Blanco/enzimología , Tejido Adiposo Blanco/crecimiento & desarrollo , Animales , Proteínas de Escherichia coli/efectos de los fármacos , Femenino , Lipogénesis/efectos de los fármacos , Embarazo , Efectos Tardíos de la Exposición Prenatal/enzimología , Efectos Tardíos de la Exposición Prenatal/genética , Ratas Wistar
17.
Endocrinology ; 159(2): 779-794, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29220483

RESUMEN

Exposure of mammalian fetuses to endocrine disruptors can increase the risk of adult-onset diseases. We previously showed that exposure of mouse fetuses to bisphenol A (BPA) caused adult-onset obesity. To examine roles of epigenetic changes in this delayed toxicity, we determined the effects of fetal mouse exposure to BPA on genome-wide DNA methylation and messenger RNA (mRNA) expression in gonadal white adipose tissues (WATs) by deep sequencing, bisulfite pyrosequencing, and real-time quantitative polymerase chain reaction. Pregnant CD-1 mice (F0) were dosed daily with 0, 5, or 500 µg/kg/d BPA during gestational days 9 to 18, and the weaned F1 animals were fed ad libitum with standard chow until they were euthanized at 19 weeks old. In the vehicle-exposed F1 animals, fggy promoter showed a clear bimodal pattern of very strong (55% to 95%) or very weak (5% to 30%) DNA methylation occurring at nearly equal incidence with no intermediate strength. Promoter hypermethylation completely suppressed mRNA expression. BPA exposure eliminated this naturally occurring dichotomy, shifting fggy promoter toward the hypomethylation state to release transcriptional suppression. The strength of Fggy mRNA expression significantly correlated with increased whole body weight and gonadal fat weight of males but not females. Bioinformatics studies showed that expression of Fggy mRNA is stronger in mouse WATs than in brown adipose tissues and enhanced in gonadal fat by diet-induced obesity. These observations suggest that prenatal exposure to BPA may disrupt the physiological bimodal nature of epigenetic regulation of fggy in mouse WATs, possibly contributing to the adult-onset obesity phenotype.


Asunto(s)
Compuestos de Bencidrilo/efectos adversos , Disruptores Endocrinos/efectos adversos , Epigénesis Genética/efectos de los fármacos , Obesidad/genética , Fenoles/efectos adversos , Fosfotransferasas/metabolismo , Efectos Tardíos de la Exposición Prenatal/genética , Regiones Promotoras Genéticas/efectos de los fármacos , Tejido Adiposo Blanco/efectos de los fármacos , Tejido Adiposo Blanco/metabolismo , Animales , Peso Corporal/efectos de los fármacos , Metabolismo de los Hidratos de Carbono , Metilación de ADN/efectos de los fármacos , Femenino , Humanos , Masculino , Ratones , Obesidad/enzimología , Obesidad/etiología , Obesidad/metabolismo , Fosfotransferasas/genética , Embarazo , Efectos Tardíos de la Exposición Prenatal/enzimología , Efectos Tardíos de la Exposición Prenatal/etiología , Efectos Tardíos de la Exposición Prenatal/metabolismo
18.
J Appl Toxicol ; 37(12): 1517-1526, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28681937

RESUMEN

Smoking throughout pregnancy can lead to complications during gestation, parturition and neonatal development. Thus, nicotine replacement therapies are a popular alternative thought to be safer than cigarettes. However, recent studies in rodents suggest that fetal and neonatal nicotine exposure alone results in cardiac dysfunction and high blood pressure. While it is well known that perinatal nicotine exposure causes increased congenital abnormalities, the mechanisms underlying longer-term deficits in cardiac function are not completely understood. Recently, our laboratory demonstrated that nicotine impairs placental protein disulfide isomerase (PDI) triggering an increase in endoplasmic reticulum stress, leading us to hypothesize that this may also occur in the heart. At 3 months of age, nicotine-exposed offspring had 45% decreased PDI levels in the absence of endoplasmic reticulum stress. Given the association of PDI and superoxide dismutase enzymes, we further observed that antioxidant superoxide dismutase-2 levels were reduced by 32% in these offspring concomitant with a 26-49% decrease in mitochondrial complex proteins (I, II, IV and V) and tissue inhibitor of metalloproteinase-4, a critical matrix metalloprotease for cardiac contractility and health. Collectively, this study suggests that perinatal nicotine exposure decreases PDI, which can promote oxidative damage and mitochondrial damage, associated with a premature decline in cardiac function.


Asunto(s)
Exposición Materna/efectos adversos , Mitocondrias Cardíacas/efectos de los fármacos , Miocardio/enzimología , Nicotina/toxicidad , Efectos Tardíos de la Exposición Prenatal/etiología , Proteína Disulfuro Isomerasas/metabolismo , Animales , Animales Recién Nacidos , Femenino , Proteínas Mitocondriales/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal/enzimología , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Ratas Wistar
19.
Biomed Res Int ; 2017: 8472312, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28567424

RESUMEN

The aim of the present study was to evaluate in vitro effects of dietary phytochemicals naringenin, quercetin, and sesamin on the activities of ethoxy- (EROD; CYP1A) and benzyloxy- (BROD; CYP3A) resorufin O-dealkylases after the exposure to the cocktail of persistent organic pollutants (POPs). CD-1 mice were exposed from weaning, through gestation and lactation to a defined mixture of POPs. Hepatic microsomes were prepared from their female offspring at postnatal day 42. Hepatic EROD and BROD activity were evaluated in the presence of quercetin, naringenin, and sesamin at nine concentrations from 5 to 100000 nM. EROD activity was strongly inhibited by quercetin with Ki values from 1.7 to 2.6 µM. BROD activity was inhibited by quercetin with Ki values from 64.9 to 75.3 µM and naringenin with Ki values from 39.3 to 45.8 µM. The IC50 and Ki values did not differ between the groups of mice with different levels of POPs exposure in any of the experimental sets. Sesamin did not inhibit either EROD or BROD. We concluded that the interactions of quercetin and naringenin with CYP1A and CYP3A in mice liver were not affected by the levels of POPs exposure.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Sistema Enzimático del Citocromo P-450/metabolismo , Familia 1 del Citocromo P450/metabolismo , Dioxoles/farmacología , Flavanonas/farmacología , Lignanos/farmacología , Exposición Materna/efectos adversos , Efectos Tardíos de la Exposición Prenatal/enzimología , Quercetina/farmacología , Animales , Citocromo P-450 CYP3A , Femenino , Ratones , Microsomas Hepáticos/enzimología , Microsomas Hepáticos/patología , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/patología
20.
J Matern Fetal Neonatal Med ; 30(3): 284-293, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27018008

RESUMEN

Glucococorticoids play a critical role in the developmental programing and fetal growth. Key molecules mediating and regulating tissue-specific glucocorticoid actions are 11beta-hydroxysteroid dehydrogenase (11beta-HSD) type 1 and 2 isozymes, both of which are expressed in the placenta and the fetal membranes. 11beta-HSD1 is implicated in the pathogenesis of metabolic syndrome and its dysregulation has been observed in pregnancy-related complications (pre-eclampsia, intrauterine growth restriction). Interestingly, preliminary clinical data have associated certain 11beta-HSD1 gene polymorphisms with hypertensive disorders in pregnancy, suggesting, if confirmed by further targeted studies, it's potential as a putative prognostic marker. Animal studies and observations in humans have confirmed that 11beta-HSD2 insufficiency is related with pregnancy adversity (pre-eclampsia, intrauterine growth restriction, preterm birth). Importantly, down-regulation or deficiency of placental 11beta-HSD2 is associated with significant restriction in fetal growth and low-birth weight, and unfavorable cardio-metabolic profile in adulthood. The potential association of 11beta-HSD1 tissue-specific dysregulation with gestational diabetes, as well as the plausible utility of 11beta-HSD2, as a biomarker of pregnancy adversity and later life morbidity, are emerging areas of intense scientific interest and future investigation.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/metabolismo , Enfermedades Fetales/enzimología , Complicaciones del Embarazo/enzimología , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/genética , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/genética , Regulación hacia Abajo , Epigénesis Genética , Femenino , Enfermedades Fetales/genética , Humanos , Placenta/enzimología , Polimorfismo Genético , Embarazo , Complicaciones del Embarazo/genética , Efectos Tardíos de la Exposición Prenatal/enzimología , Efectos Tardíos de la Exposición Prenatal/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...