Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 742
Filtrar
1.
Birth Defects Res ; 116(4): e2310, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38563145

RESUMEN

INTRODUCTION: In this study, we aimed to investigate the inflammatory factors, oxidative stress, and histopathological consequences of the brain-gut axis in male and female rats prenatally exposed to VPA. METHODS: Pregnant Wistar rats were randomly divided into two groups. The animals received saline, and valproic acid (VPA) (600 mg/kg, i.p.) on embryonic day 12.5 (E12.5). All offspring were weaned on postnatal day 21, and the experiments were done in male and female rats on day 60. The brain and intestine tissues were extracted to assess histopathology, inflammation, and oxidative stress. RESULTS: An increase of interleukin-1ß (IL-1ß) and interleukin-6 (IL-6) and a decrease of interleukin-10 (IL-10) were observed in the two sexes and two tissues of the autistic rats. In the VPA-exposed animals, malondialdehyde (MDA) and protein carbonyl (PC) increased in the brain of both sexes and the intestines of only the males. The total antioxidant capacity (TAC), superoxide dismutase (SOD), and catalase (CAT) significantly decreased in both tissues of male and female autistic groups. Histopathological evaluation showed that the %apoptosis of the cortex in the autistic male and female groups was more than in controls whereas this parameter in the CA1 and CA3 was significant only in the male rats. In the intestine, histopathologic changes were seen only in the male autistic animals. CONCLUSION: The inflammatory and antioxidant factors were in line in the brain-gut axis in male and female rats prenatally exposed to VPA. Histopathological consequences were more significant in the VPA-exposed male animals.


Asunto(s)
Trastorno Autístico , Ácido Valproico , Embarazo , Ratas , Masculino , Femenino , Animales , Ácido Valproico/toxicidad , Trastorno Autístico/inducido químicamente , Antioxidantes/metabolismo , Ratas Wistar , Eje Cerebro-Intestino , Estrés Oxidativo , Interleucina-6
2.
Congenit Anom (Kyoto) ; 64(2): 47-60, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38403785

RESUMEN

Cranial neural crest cells (NCCs) are critical for craniofacial development. The administration of valproic acid (VPA) to pregnant females causes craniofacial malformations in offspring. However, the in vivo influence of VPA on mammalian cranial NCCs remains unclear. In this study, we aimed to elucidate the developmental stage-specific effect of VPA on cranial NCCs through the administration of a single dose of VPA to pregnant rat females immediately prior to the formation of the cranial neural crest (NC). We performed whole-mount immunohistochemistry or in situ hybridization to examine localization changes of gene transcripts associated with the epithelial-mesenchymal transition of the cranial NC (i.e., cranial NCC formation) and cranial NCC migration. The results showed that Hoxa2 mRNA was abnormally detected and Sox9 mRNA expression was decreased in the midbrain-rhombomere (R) 1/2 NC, which forms cranial NCCs that migrate to the frontonasal mass (FNM) and branchial arch (BA) 1, through VPA administration, thus reducing the formation of SNAI2-positive NCCs. Hoxa2-positive NCCs were detected normally in BA2 and abnormally in FNM and BA1, which are normally Hox-free, implying VPA-induced abnormal cranial NCC migration. In vitro verification experiments using the whole embryo culture system revealed that midbrain-R4 NCC migration was abnormal. These results indicate that VPA reduces the formation/delamination of the midbrain-R1/2 NCCs in a developmental stage-specific manner and subsequently causes the abnormal migration of R4 NCCs, which suggests that the abnormal formation and migration of cranial NCCs contribute to the inhibition of axonal elongation in the trigeminal nerve and a reduction in head size.


Asunto(s)
Cresta Neural , Ácido Valproico , Animales , Ratas , Cresta Neural/metabolismo , Ácido Valproico/toxicidad , ARN Mensajero/metabolismo , ARN Mensajero/farmacología , Movimiento Celular , Mamíferos
3.
Reprod Toxicol ; 124: 108551, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38280688

RESUMEN

Gestational exposure to the anticonvulsant drug valproic acid (VPA) is associated with congenital malformations and neurodevelopmental disorders through its action as a histone deacetylase inhibitor. VPA can elicit placental toxicity and affect placental growth and development. The objective of this study was to evaluate the impact of maternal exposure to VPA on the mouse placenta following exposure on gestational day (GD) 13 since previous studies have shown that mice exposed at this time during gestation give birth to offspring with an autism spectrum disorder-like phenotype. We exposed CD-1 dams to a teratogenic dose (600 mg/kg) of VPA or saline on GD13 and assessed fetoplacental growth and development on GD18. We evaluated epigenetic modifications, including acetylated histone H4 (H4ac), methylated H3K4 (H3K4me2) using immunohistochemistry, and global DNA methylation in the placenta at 1, 3, and 24 h following maternal exposure on GD13. In utero exposure to VPA on GD13 significantly decreased placental weight and increased fetal resorptions. Moreover, VPA significantly increased the staining intensity of histone H4 acetylation and H3K4 di-methylation across the placenta at 1 and 3 h post maternal dose. Our results also demonstrate that VPA significantly decreased global DNA methylation levels in placental tissue. These results show that gestational exposure to VPA interferes with placental growth and elicits epigenetic modifications, which may play a vital role in VPA-induced developmental toxicity.


Asunto(s)
Trastorno del Espectro Autista , Ácido Valproico , Embarazo , Femenino , Ratones , Animales , Ácido Valproico/toxicidad , Histonas/metabolismo , Placenta/metabolismo , Epigénesis Genética
4.
J Toxicol Sci ; 49(2): 69-77, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38296531

RESUMEN

Placental dysfunction can disrupt pregnancy. However, few studies have assessed the effects of chemical-induced toxicity on placental function. Here, we examined the effects of valproic acid (VPA) as a model chemical on production of hormones and on glucose uptake in human choriocarcinoma cell line BeWo. Cells were treated with forskolin to differentiate into syncytiotrophoblasts, which were then treated with VPA for 72 hr. Real-time RT-PCR analysis showed that VPA significantly increased the mRNA expression of chorionic gonadotropin ß (CGB), a hormone that is produced by the placenta in the first trimester of pregnancy, relative to that in the forskolin-only group. It also suppressed the increase in intracellular glucose uptake and GLUT1 level observed in the forskolin-only group. RNA-seq analysis and pathway database analysis revealed that VPA consistently decreased the level of HIF-1α protein and expression of its downstream target genes HK2 and ADM in the hypoxia pathway. Cobalt chloride, a HIF-1α inducer, inhibited CGB upregulation in VPA-treated cells and rescued VPA-induced suppression of glucose uptake and GLUT1 level. Thus, HIF-1α-mediated elevation of CGB expression and suppression of glucose uptake by VPA is a novel mechanism of placental dysfunction.


Asunto(s)
Placenta , Ácido Valproico , Embarazo , Femenino , Humanos , Ácido Valproico/toxicidad , Placenta/metabolismo , Colforsina/metabolismo , Transportador de Glucosa de Tipo 1/genética , Transportador de Glucosa de Tipo 1/metabolismo , Glucosa/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Línea Celular Tumoral
5.
Neurotoxicology ; 101: 93-101, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38191030

RESUMEN

Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by social deficits and repetitive/stereotyped behaviors. Prenatal exposure to valproic acid (VPA) has been reported to induce ASD-like symptoms in human and rodents. However, the etiology and pathogenesis of ASD have not been well elucidated. This study aimed to explore the mechanisms underlying VPA-induced ASD-like behaviors using zebrafish model and investigated whether vitamin A could prevent VPA-induced neurotoxicity. Here, zebrafish embryos were exposed to 0, 25 and 50 µM VPA from 4 to 96 h post fertilization (hpf) and the neurotoxicity was assessed. Our results showed that VPA affected the normal development of zebrafish larvae and induced ASD-like behaviors, including reduced locomotor activity, decreased distance near conspecifics, impaired social interaction and repetitive swimming behaviors. Exposure to VPA decreased the GFP signal in transgenic HuC:egfp zebrafish according to the negative effect of VPA on the expression of neurodevelopmental genes. In addition, VPA enhanced oxidative stress by promoting the production of reactive oxygen species (ROS) and hydrogen peroxide (H2O2) and inhibiting the activity of superoxide dismutase, then triggered apoptosis by upregulation of apoptotic genes. These adverse outcomes were mitigated by vitamin A, suggesting that vitamin A rescued VPA-induced ASD-like symptoms by inhibiting oxidative stress and apoptosis. Overall, this study identified vitamin A as a promising strategy for future therapeutic regulator of VPA-induced ASD-like behaviors.


Asunto(s)
Trastorno del Espectro Autista , Trastorno Autístico , Efectos Tardíos de la Exposición Prenatal , Embarazo , Animales , Femenino , Humanos , Ácido Valproico/toxicidad , Trastorno Autístico/inducido químicamente , Trastorno Autístico/prevención & control , Trastorno Autístico/tratamiento farmacológico , Pez Cebra , Trastorno del Espectro Autista/inducido químicamente , Trastorno del Espectro Autista/prevención & control , Trastorno del Espectro Autista/tratamiento farmacológico , Vitamina A/uso terapéutico , Larva , Peróxido de Hidrógeno , Conducta Social , Conducta Animal , Estrés Oxidativo , Modelos Animales de Enfermedad , Efectos Tardíos de la Exposición Prenatal/inducido químicamente
6.
Artículo en Inglés | MEDLINE | ID: mdl-38244714

RESUMEN

Autism Spectrum Disorder (ASD) is a neurodevelopmental disorder characterized mainly by deficits in social communication and stereotyped and restricted behavior and interests with a male to female bias of 4.2/1. Social behavior in ASD animal models is commonly analyzed in males, and seldomly in females, using the widely implemented three-chambers test procedure. Here, we implemented a novel procedure, the Live Mouse Tracker (LMT), combining artificial intelligence, machine learning procedures and behavioral measures. We used it on mice that were prenatally exposed to valproic acid (VPA) (450 mg/kg) at embryonic day 12.5, a widely recognized and potent ASD model that we had previously extensively characterized. We focused on female mice offspring, in which social deficits have been rarely documented when using the 3-CT procedure. We recorded several parameters related to social behavior in these mice, continuously for three days in groups of four female mice. Comparisons were made on groups of 4 female mice with the same treatment (4 saline or 4 VPA) or with different treatments (3 saline and 1 VPA). We report that VPA females show several types of social deficits, which are different in nature and magnitude in relation with time. When VPA mice were placed in the LMT alongside saline mice, their social deficits showed significant improvement as early as 1 h from the start of the experiment, lasting up to 3 days throughout the duration of the experiment. Our findings suggest that ASD may be underdiagnosed in females. They also imply that ASD-related social deficits can be ameliorated by the presence of typical individuals.


Asunto(s)
Trastorno del Espectro Autista , Efectos Tardíos de la Exposición Prenatal , Femenino , Masculino , Animales , Ratones , Humanos , Ácido Valproico/toxicidad , Inteligencia Artificial , Trastorno del Espectro Autista/inducido químicamente , Conducta Social , Estereotipo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Modelos Animales de Enfermedad , Conducta Animal
7.
Mol Neurobiol ; 61(1): 167-174, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37592184

RESUMEN

Autism spectrum disorders (ASD) are a highly heterogeneous group of neurodevelopmental disorders caused by complex interaction between various genes and environmental factors. As the hippocampus and prefrontal cortex are involved in social recognition, they are the regions of the brain implicated in autism. The effects of prenatal exposure to valproic acid (VPA) can induce an ASD phenotype in both humans and rats; this tool is commonly used to model the complexity of ASD symptoms in the laboratory. However, researchers rarely undertake epigenetic regulation of the brain regions using this model. The present study has addressed this gap by examining gene expression abnormalities in the hippocampus and prefrontal cortex in the VPA rat model of ASD. mRNA and miRNA sequencing was performed on samples from the hippocampus and prefrontal cortex of the VPA model of autism. According to the analysis, 3000 mRNAs in the hippocampus and 2187 mRNAs in the prefrontal cortex showed a significant difference in expression between the VPA and saline groups. In addition, there were 115 DE miRNAs in the hippocampus and 14 DE miRNAs in the prefrontal cortex. Further, the predicted and validated target mRNA of DE miRNA enriched pathways involved neurotransmitter uptake, long-term synaptic depression, and AMPA receptor complex (anti-GluA2-b) in the hippocampus; as well as the neuroactive ligand-receptor interaction and regulation of postsynaptic membrane potential in the prefrontal cortex. This revealed the negative regulation network of miRNAs-mRNAs in the hippocampus and prefrontal cortex, while filtering out key genes (miR-10a-5p and Grm3). Finally, the significant variable miR-10a-5p and its negative regulated genes (Grm3) were verified in both brain regions by QPCR. Importantly, the fact that miR-10a-5p downregulated Grm3 in both the hippocampus and the prefrontal cortex may play a potentially significant role in the occurrence and development of autism. This study suggests that the VPA model has the potential to reproduce ASD-related hippocampus and prefrontal cortex abnormalities, at the epigenetic and transcriptional levels. Furthermore, the network of miRNAs-mRNAs was confirmed; this negative regulatory relationship may play a key role in determining the occurrence and development of autism. The study of this topic help better understand the pathogenesis of ASD.


Asunto(s)
Trastorno del Espectro Autista , MicroARNs , Efectos Tardíos de la Exposición Prenatal , Embarazo , Humanos , Femenino , Ratas , Animales , Ácido Valproico/toxicidad , Epigénesis Genética , Trastorno del Espectro Autista/genética , Corteza Prefrontal/metabolismo , Perfilación de la Expresión Génica , MicroARNs/metabolismo , Hipocampo/metabolismo , Modelos Animales de Enfermedad , Efectos Tardíos de la Exposición Prenatal/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
8.
Int J Dev Neurosci ; 84(1): 64-74, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37960995

RESUMEN

PURPOSE: Toxoplasmosis is a disease caused by infection with a type of coccidial protozoan parasite called Toxoplasma gondii. The relationship between toxoplasmosis and cognitive disorders in neurodegenerative diseases has been proven. There is also evidence that children born to Toxoplasma-infected mothers are more likely to develop autism. METHODS: In the present study, Toxoplasma-infected pregnant BALB/c mice were given valproic acid to induce autism in their male offspring, and their social behaviors, learning, and memory were examined. Chronic toxoplasmosis was established in BALB/c mice by intraperitoneal injection of cyst form of T. gondii. To induce autism, 600 mg/kg of valproic acid was injected intraperitoneally into mice on the 12.5th day of pregnancy. The behavioral experiments, such as social interaction, novel object recognition, and passive avoidance tasks, were performed on male offspring at 50 days. RESULTS: Toxoplasma and valproic acid during the embryonic period caused social communication deficits and disrupted recognition memory and avoidance memory in offspring. Our findings showed that administering valproic acid to Toxoplasma-infected mothers exacerbates cognitive disorders in their offspring.


Asunto(s)
Trastorno Autístico , Disfunción Cognitiva , Toxoplasma , Toxoplasmosis , Humanos , Embarazo , Femenino , Niño , Masculino , Animales , Ratones , Ácido Valproico/toxicidad , Trastorno Autístico/inducido químicamente , Trastorno Autístico/complicaciones , Ratones Endogámicos BALB C , Modelos Animales de Enfermedad , Toxoplasmosis/complicaciones , Toxoplasmosis/parasitología , Toxoplasmosis/psicología
9.
Environ Toxicol Pharmacol ; 105: 104343, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38122861

RESUMEN

Assessing the role of α-hexabromocyclododecane α-HBCDD as a factor of susceptibility for Autism Spectrum disorders by using valproic acid-exposed rat model (VPA) required characterizing VPA pharmacokinetic in the context of α-HBCDD-co-exposure in non-pregnant and pregnant rats. The animals were exposed to α-HBCDD by gavage (100 ng/kg/day) for 12 days. This was followed by a single intraperitoneal dose of VPA (500 mg/kg) or a daily oral dose of VPA (500 mg/kg) for 3 days. Exposure to α-HBCDD did not affect the pharmacokinetics of VPA in pregnant or non-pregnant rats. Surprisingly, VPA administration altered the pharmacokinetics of α-HBCDD. VPA also triggered higher foetal toxicity and lethality with the PO than IP route. α-HBCDD did not aggravate the embryotoxicity observed with VPA, regardless of the route of exposure. Based on this evidence, a single administration of 500 mg/kg IP is the most suitable VPA model to investigate α-HBCDD co-exposure.


Asunto(s)
Trastorno del Espectro Autista , Hidrocarburos Bromados , Efectos Tardíos de la Exposición Prenatal , Embarazo , Humanos , Femenino , Ratas , Animales , Ácido Valproico/toxicidad , Trastorno del Espectro Autista/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Hidrocarburos Bromados/toxicidad , Modelos Animales de Enfermedad
10.
Int. j. morphol ; 41(6): 1596-1602, dic. 2023. ilus
Artículo en Español | LILACS | ID: biblio-1528809

RESUMEN

El ácido valproico (VPA) es un fármaco antiepiléptico teratógenico que, al ser administrado durante etapas tempranas del embarazo, puede producir alteraciones en el desarrollo embriofetal, las que se manifiestan tanto a nivel del sistema nervioso como del testículo. No obstante, se ha reportado que la administración de vitamina E (VE) podría revertir dichas alteraciones. El objetivo del presente estudio fue determinar el efecto protector de la VE a nivel testicular en fetos y ratones púberes expuestos a VPA durante la fase embrionaria de su desarrollo. Se utilizó un total de 30 ratones hembra adultas gestantes (Mus musculus) cepa BALB/c, las cuales se dividieron en 6 grupos. El estudio contempló el análisis de fetos machos a los 17,5 días post-coital (dpc) y machos juveniles a las 6 semanas post-natal. A los grupos 1 y 4 se les administró 0,3 mL de solución fisiológica (grupos control para 17,5 dpc y 6 semanas postnatal, respectivamente). A los grupos 2 y 5 se les suministró la cantidad de 600 mg/kg de VPA (grupos VPA), en tanto que a los grupos 3 y 6 se les aplicó la misma dosis de VPA complementada con 200 UI de VE (grupos VPA+VE). Se describió la histología normal y patológica del compartimento peritubular del testículo. En los grupos VPA se evidenció una degeneración de la pared peritubular, y atrofia de túbulos seminíferos, así como exfoliación de las células germinales. Por el contrario, en los grupos VPA+VE tales signos no fueron observados y la morfología presentó aspecto normal solo con algunas alteraciones focales. Estos resultados corroboran el hecho que la administración de VE contrarresta en parte, los efectos deletéreos que ocasiona el VPA.


SUMMARY: Valproic acid (VPA) is a teratogenic antiepileptic drug that, when administered during the early stages of pregnancy, can produce alterations in embryo-fetal development, which manifest both at the level of the nervous system and the testicle. However, it has been reported that the administration of vitamin E (VE) could reverse these alterations. The study aimed to determine the protective effect of VE at the testicular level in fetuses and pubertal mice exposed to VPA during the embryonic phase of their development. 30 pregnant adult female mice (Mus musculus) BALB/c strain were used, which were divided into 6 groups. The study included the analysis of male fetuses at 17.5 days post-coital (dpc) and juvenile males at 6 weeks post-natal. Groups 1 and 4 were administered 0.3 mL of physiological solution. Groups 2 and 5 were given 600 mg/kg of VPA (VPA groups), while groups 3 and 6 were given the same dose of VPA supplemented with 200 IU of VE (VPA+VE). The normal and pathological histology of the peritubular compartment of the testis was described. In the VPA groups, degeneration of the peritubular wall, and atrophy of the seminiferous tubules, as well as exfoliation of the germ cells, were evident. On the contrary, in the VPA+VE groups such signs were not observed and the morphology presented a normal appearance with only some focal alterations. These results corroborate the fact that the administration of VE partially counteracts the deleterious effects caused by VPA.


Asunto(s)
Animales , Femenino , Embarazo , Ratones , Testículo/efectos de los fármacos , Vitamina E/administración & dosificación , Ácido Valproico/toxicidad , Efectos Tardíos de la Exposición Prenatal , Túbulos Seminíferos/citología , Túbulos Seminíferos/efectos de los fármacos , Testículo/citología , Vitamina E/farmacología , Ratones Endogámicos BALB C , Anticonvulsivantes/toxicidad
11.
Arh Hig Rada Toksikol ; 74(3): 218-223, 2023 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-37791674

RESUMEN

Valproate is known to disturb the kidney function, and high doses or prolonged intake may cause serum ion imbalance, kidney tubular acidosis, proteinuria, hyperuricosuria, polyuria, polydipsia, and dehydration. The aim of this in vivo study was to see whether naringin would counter the adverse effects of high-dose valproate in C57Bl/6 mice and to which extent. As expected, valproate (150 mg/kg bw a day for 10 days) caused serum hyperkalaemia, more in male than female mice. Naringin reversed (25 mg/kg bw a day for 10 days) the hyperkalaemia and activated antioxidative defence mechanisms (mainly catalase and glutathione), again more efficiently in females. In males naringin combined with valproate was not as effective and even showed some prooxidative effects.


Asunto(s)
Antioxidantes , Hiperpotasemia , Femenino , Masculino , Animales , Ratones , Antioxidantes/farmacología , Ácido Valproico/toxicidad , Peroxidación de Lípido , Ratones Endogámicos C57BL , Riñón , Catalasa/metabolismo , Catalasa/farmacología , Estrés Oxidativo , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa/farmacología
12.
Neurotoxicol Teratol ; 99: 107286, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37442398

RESUMEN

Valproic acid (VPA) is an anti-epileptic medication that increases the risk of neural tube defect (NTD) outcomes in infants exposed during gestation. Previous studies into VPA's mechanism of action have focused on alterations in gene expression and metabolism but have failed to consider how exposure changes the abundance of critical developmental proteins over time. This study evaluates the effects of VPA on protein abundance in the developmentally distinct tissues of the mouse visceral yolk sac (VYS) and embryo proper (EMB) using mouse whole embryo culture. Embryos were exposed to 600 µM VPA at 2 h intervals over 10 h during early organogenesis with the aim of identifying protein pathways relevant to VPA's mechanism of action in failed NTC. Protein abundance was measured through tandem mass tag (TMT) labeling followed by liquid chromatography and mass spectrometry. Overall, there were over 1500 proteins with altered abundance after VPA exposure in the EMB or VYS with 428 of these proteins showing previous gene expression associations with VPA exposure. Limited overlap of significant proteins between tissues supported the conclusion of independent roles for the VYS and EMB in response to VPA. Pathway analysis of proteins with increased or decreased abundance identified multiple pathways with mechanistic relevance to NTC and embryonic development including convergent extension, Wnt Signaling/planar cell polarity, cellular migration, cellular proliferation, cell death, and cytoskeletal organization processes as targets of VPA. Clustering of co-regulated proteins to identify shared patterns of protein abundance over time highlighted 4 h and 6/10 h as periods of divergent protein abundance between control and VPA-treated samples in the VYS and EMB, respectively. Overall, this study demonstrated that VPA temporally alters protein content in critical developmental pathways in the VYS and the EMB during early organogenesis in mice.


Asunto(s)
Defectos del Tubo Neural , Ácido Valproico , Humanos , Embarazo , Femenino , Ratones , Animales , Ácido Valproico/toxicidad , Proteínas/metabolismo , Embrión de Mamíferos , Defectos del Tubo Neural/metabolismo , Organogénesis
13.
Toxicol Lett ; 384: 96-104, 2023 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-37451652

RESUMEN

The evaluation of chemical and pharmaceutical safety for humans is moving from animal studies to New Approach Methodologies (NAM), reducing animal use and focusing on mechanism of action, whilst enhancing human relevance. In developmental toxicology, the mechanistic approach is facilitated by the assessment of predictive biomarkers, which allow mechanistic pathways perturbation monitoring at the basis of human hazard assessment. In our search for biomarkers of maldevelopment, we focused on chemically-induced perturbation of the retinoic acid signaling pathway (RA-SP), a major pathway implicated in a plethora of developmental processes. A genome-wide expression screening was performed on zebrafish embryos treated with two teratogens, all-trans retinoic acid (ATRA) and valproic acid (VPA), and a non-teratogen reference compound, folic acid (FA). Each compound was found to have a specific mRNA expression profile with 248 genes commonly dysregulated by both teratogenic compounds but not by FA. These genes were implicated in several developmental processes (e.g., the circulatory and nervous system). Given the prominent response of neurodevelopmental gene sets, and the crucial need to better understand developmental neurotoxicity, our study then focused on nervous system development. We found 62 genes that are potential early neurodevelopmental toxicity biomarker candidates. These results advance NAM-based safety assessment evaluation by highlighting the usefulness of the RA-SP in providing early toxicity biomarker candidates.


Asunto(s)
Tretinoina , Pez Cebra , Animales , Humanos , Tretinoina/toxicidad , Pez Cebra/genética , Pez Cebra/metabolismo , Ácido Valproico/toxicidad , Regulación de la Expresión Génica , Teratógenos/toxicidad , Biomarcadores , Sistema Nervioso/metabolismo , Regulación del Desarrollo de la Expresión Génica , Embrión no Mamífero
14.
Birth Defects Res ; 115(16): 1486-1499, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37522293

RESUMEN

BACKGROUND: Cardiovascular diseases are prevalent in autistic patients. As exercise is useful in the treatment of medical conditions, this study aimed to identify the effect of low-intensity endurance exercise (LIEE) and moderate-intensity endurance exercise (MIEE) on cardiovascular events in autistic rats. METHODS: Valproic acid (VPA) was administrated once on gestational day 12.5 to pregnant rats to produce autism-like symptoms in offspring. Thirty-day-old offspring were divided into 12 groups: Male-CTL, Male-VPA, Male-CTL + LIEE, Male-CTL + MIEE, Male-VPA + LIEE, Male-VPA + MIEE, Female-CTL, Female-VPA, Female-CTL + LIEE, Female-CTL + MIEE, Female-VPA + LIEE, and Female-VPA + MIEE. LIEE and MIEE were performed 5 days a week for 30 days. Twenty-four hours after the last exercise session, electrocardiogram and hemodynamic and cardiac function indices were recorded. RESULTS: The results indicated that +dp/dt max and contractility index (CI) decreased in the Female-VPA group compared to the Female-CTL group. LIEE increased these parameters in the Female-VPA + LIEE group. However, MIEE normalized CI in the Male-VPA + MIEE compared to the Male-VPA group. Tau increased in the Female-VPA group compared to the Female-CTL group and it decreased in the Female-VPA + MIEE group compared to the Female-VPA group. LIEE and MIEE recovered the reduction of heart rate and the increase in P, R, and T amplitudes in Male-VPA group. LIEE and MIEE increased heart rate variability in the Male-VPA and Female-VPA groups. CONCLUSIONS: The findings showed that LIEE and MIEE alleviated cardiac dysfunction and disturbances in heart rhythm in the autistic offspring. Exercise may be recommended as a routine program for autistic patients to prevent and treat the harmful cardiovascular consequences of autism.


Asunto(s)
Trastorno Autístico , Efectos Tardíos de la Exposición Prenatal , Humanos , Embarazo , Ratas , Masculino , Femenino , Animales , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Ácido Valproico/toxicidad
15.
Toxicol Sci ; 195(2): 169-183, 2023 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-37505509

RESUMEN

Valproic acid (VPA) has long been the most widely used antiepileptic drug (AED) for the treatment of epilepsy, bipolar psychiatric disorders, and migraine. However, long-term VPA treatment has several adverse effects on the male reproductive system notably on endocrine functions and/or spermatic parameters. In utero exposure of the fetus to VPA is well known to be associated with a higher risk of several congenital malformations including those of male reproductive organs. Subsequent generations of AEDs, such as carbamazepine (CARB) and lamotrigine (LAM), are considered safer and are currently recommended for women of child-bearing age with epilepsy. Because anomalies of the male genital tract mostly result from endocrine imbalance during fetal life, we hypothesized that AEDs could directly impair testis differentiation. We thus aimed at identifying and characterizing the effects of VPA, CARB, and LAM on the differentiation and function of the different testicular cell types, and at understanding the mechanisms underlying these effects. By using ex vivo culture of first-trimester human fetal testes, we show that VPA induces multiple endocrine disruptive effects, compared with the milder ones caused by CARB and LAM. AED also subtly altered the germ cell lineage in distinct manners. Transcriptomic analysis of VPA-induced alterations highlighted a very broad range of effects on the fetal testis. Overall, our results show that AEDs can behave as endocrine disruptors for the human fetal testis ex vivo. This is consistent with, and likely underlies, the VPA-induced male genital tract masculinization abnormalities observed in patients.


Asunto(s)
Disruptores Endocrinos , Epilepsia , Humanos , Masculino , Femenino , Anticonvulsivantes/toxicidad , Anticonvulsivantes/uso terapéutico , Testículo , Disruptores Endocrinos/metabolismo , Ácido Valproico/toxicidad , Epilepsia/inducido químicamente , Epilepsia/tratamiento farmacológico , Epilepsia/metabolismo , Feto
16.
Birth Defects Res ; 115(16): 1475-1485, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37507847

RESUMEN

Valproic acid (VPA) is a widely prescribed antiepileptic drug with various medicinal efficacies. Accumulated evidence implied that prenatal exposure to VPA is highly associated with autism spectrum disorder (ASD). In this study, the zebrafish were exposed to a set of VPA concentrations (0, 5, 10, 20, 40, 80, 160, 320, 640, 1280, and 2560 µM) at 5 h post fertilization (hpf) to 120 hpf. The adverse effects of VPA were extensively studied through the evaluations on the mortality, heartbeats, spontaneous tail coiling, and hatching rate. Morphological observations were conducted at 120 hpf, following the exposure termination. Basic locomotor responses and anxiety-like behavioral alterations evaluated for behavioral impairments are the hallmark feature of ASD. The exposure to VPA at teratogenic concentrations reduced the aforementioned parameters in a dose-dependent manner (p ≤ .05). At the selected non-teratogenic concentrations of VPA, the treated larvae demonstrated profound alterations of basic locomotor responses. No significant changes of anxiety and thigmotactic behaviors were observed on the VPA-treated fish compared to the control (p ≥ .005). This study depicted that embryonic zebrafish exposure to VPA produced significant toxicity and teratogenicity effects as well as the alterations of basic behavioral responses. Overall, this study provides a fundamental insight of the toxicity effects at morphological and behavioral levels to facilitate the understanding of ASD mechanisms at different molecular levels.


Asunto(s)
Trastorno del Espectro Autista , Teratogénesis , Animales , Ácido Valproico/toxicidad , Pez Cebra , Conducta Animal , Anticonvulsivantes/toxicidad , Teratógenos/toxicidad
17.
Physiol Behav ; 269: 114286, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37402416

RESUMEN

Social communication and interaction deficits, memory impairment, and anxiety-like behavior are characterized in many people identified with autism spectrum disorder (ASD). A thorough understanding of the specific aspects that contribute to the deficiencies associated with ASD can aid research into the etiology of the disorder while also providing targets for more effective intervention. As part of the ASD pathophysiology, alterations in synaptogenesis and abnormal network connections were seen in high-order brain areas, which control social behavior and communication. The early emergence of microglia during nervous system development may contribute to synaptic dysfunction and the pathobiology of ASD. Since aquaporin-4 (AQP4) appears to be required for the basic procedures of synapse activation, certain behavioral and cognitive impairments as well as disturbance in water homeostasis might likely arise from AQP4 deficiency. Here, through the measurement of the water content of the hippocampus and behavioral experiments we aim to explore the contribution of astrocytic AQP4 to the autism-like behavior induced by prenatal valproic acid (VPA) exposure and whether inhibition of AQP4 per se can induce autistic-like behavior in control rats. Microinjection of TGN-020 (10 µM, i.c.v), a specific AQP4 inhibitor, for 7 successive days before behavioral tasks from postnatal day 28 to 35 revealed that inhibition of AQP4 in the control offspring caused lower social interaction and locomotor activity, higher anxiety, and decreased ability to recognize novel objects, very similar to the behavioral changes observed in offspring prenatally exposed to VPA. However, VPA-exposed offspring treated with TGN-020, showed no further remarkable behavioral impairments than those detected in the autistic-like rats. Furthermore, both control offspring treated with TGN-020 and offspring exposed to VPA had a considerable accumulation of water in their hippocampi. But AQP4 inhibition did not affect the water status of the autistic-like rats. The findings of this study revealed that control offspring exhibited similar hippocampal water retention and behavioral impairments that were observed in maternal VPA-exposed offspring following inhibition of astrocytic AQP4, whereas, in autistic-like rats, it did not produce any significant change in water content and behaviors. Findings suggest that AQP4 deficiency could be associated with autistic disorder and may be a potential pharmaceutical target for treating autism in the future.


Asunto(s)
Acuaporinas , Trastorno del Espectro Autista , Trastorno Autístico , Efectos Tardíos de la Exposición Prenatal , Embarazo , Humanos , Femenino , Ratas , Animales , Ácido Valproico/toxicidad , Trastorno Autístico/inducido químicamente , Trastorno del Espectro Autista/inducido químicamente , Exposición Materna , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Conducta Social , Acuaporinas/farmacología , Modelos Animales de Enfermedad , Conducta Animal
18.
Hear Res ; 436: 108816, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37285705

RESUMEN

Auditory dysfunction is a common feature of autism spectrum disorder (ASD) and ranges from deafness to hypersensitivity. The auditory brainstem response (ABR) permits study of the amplitude and latency of synchronized electrical activity along the ascending auditory pathway in response to clicks and pure tone stimuli. Indeed, numerous studies have shown that subjects with ASD have ABR abnormalities. In utero exposure to the antiepileptic drug valproic acid (VPA) is associated with human cases of ASD and is used as an animal model of ASD. Previous studies have shown that VPA-exposed animals have significantly fewer neurons in the auditory brainstem and thalamus, reduced ascending projections to the auditory midbrain and thalamus and increased neuronal activation in response to pure tone stimuli. Accordingly, we hypothesized that VPA-exposed animals would have abnormal ABRs throughout their lifespans. We approached this hypothesis in two cohorts. First, we examined ABRs from both ears on postnatal day 22 (P22). Then, we examined monaural ABRs in animals at P28, 60, 120, 180, 240, 300 and 360. Our results suggest that at P22, VPA-exposed animals have elevated thresholds and increased peak latencies. However, by P60 these differences largely normalize with differences appearing only near hearing threshold. Additionally, our analysis revealed that maturation of ABR waves occurred at different trajectories in control and VPA-exposed animals. These results, together with our previous work, suggest that VPA exposure not only impacts total neuron number and connectivity, but also auditory evoked responses. Finally, our longitudinal analysis suggests that delayed maturation of auditory brainstem circuits may impact ABRs throughout the lifespan of the animal.


Asunto(s)
Trastorno del Espectro Autista , Potenciales Evocados Auditivos del Tronco Encefálico , Animales , Humanos , Potenciales Evocados Auditivos del Tronco Encefálico/fisiología , Trastorno del Espectro Autista/inducido químicamente , Trastorno del Espectro Autista/metabolismo , Vías Auditivas , Tronco Encefálico , Ácido Valproico/toxicidad , Ácido Valproico/metabolismo , Modelos Animales de Enfermedad , Umbral Auditivo/fisiología
19.
Toxicol Appl Pharmacol ; 474: 116611, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37385477

RESUMEN

The placenta is a critical organ for fetal development and a healthy pregnancy, and has multifaceted functions (e.g., substance exchange and hormone secretion). Syncytialization of trophoblasts is important for maintaining placental functions. Epilepsy is one of the most common neurological conditions worldwide. Therefore, this study aimed to reveal the influence of antiepileptic drugs, including valproic acid (VPA), carbamazepine, lamotrigine, gabapentin, levetiracetam, topiramate, lacosamide, and clobazam, at clinically relevant concentrations on syncytialization using in vitro models of trophoblasts. To induce differentiation into syncytiotrophoblast-like cells, BeWo cells were treated with forskolin. Exposure to VPA was found to dose-dependently influence syncytialization-associated genes (ERVW-1, ERVFRD-1, GJA1, CGB, CSH, SLC1A5, and ABCC4) in differentiated BeWo cells. Herein, the biomarkers between differentiated BeWo cells and the human trophoblast stem model (TSCT) were compared. In particular, MFSD2A levels were low in BeWo cells but abundant in TSCT cells. VPA exposure affected the expression of ERVW-1, ERVFRD-1, GJA1, CSH, MFSD2A, and ABCC4 in differentiated cells (ST-TSCT). Furthermore, VPA exposure attenuated BeWo and TSCT cell fusion. Finally, the relationships between neonatal/placental parameters and the expression of syncytialization markers in human term placentas were analyzed. MFSD2A expression was positively correlated with neonatal body weight, head circumference, chest circumference, and placental weight. Our findings have important implications for better understanding the mechanisms of toxicity of antiepileptic drugs and predicting the risks to placental and fetal development.


Asunto(s)
Placenta , Trofoblastos , Recién Nacido , Humanos , Embarazo , Femenino , Placenta/metabolismo , Ácido Valproico/toxicidad , Anticonvulsivantes/farmacología , Línea Celular , Transportadoras de Casetes de Unión a ATP/metabolismo , Antígenos de Histocompatibilidad Menor/metabolismo , Antígenos de Histocompatibilidad Menor/farmacología , Sistema de Transporte de Aminoácidos ASC/metabolismo
20.
Environ Toxicol ; 38(10): 2400-2415, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37357844

RESUMEN

Autism spectrum disorder (ASD) is a multifactorial neurodevelopmental disorder characterized by restrictive and repetitive behavior followed by impairment in social, verbal, and non-verbal interaction and communication. Valproic acid (VPA) is a well-known anti-epileptic drug, but its prenatal exposure to animals causes social impairment, neurotransmitters imbalance, and neuroinflammation with ASD-like phenotypes. Syringic acid (SA) is a polyphenolic compound with anti-inflammatory, anti-apoptotic, antioxidant, and neuromodulator activity. The purpose of study was to investigate the protective effect of Syringic acid (SA) in prenatal VPA-treated rats through behavioral, neuroinflammation, oxidative stress, neurotransmitters, neuronal integrity, and apoptotic marker. Single dose of VPA was administered 600 mg/kg, i.p. on a gestational day (GD) 12th and SA was administrated from PnD 26th to 54th at the dose of 25, 50, and 100 mg/kg, p.o. On PnD 56th behavioral parameters (Pain sensitivity, open field test, narrow beam walks test and social impairment test) were performed and all animals were sacrificed, and brain tissue was isolated for oxidative stress (GSH, CAT, and LPO), neuroinflammation (TNF-α and IL-6) and neurotransmitters (GABA and Glutamate), histopathology (H&E, Nissl), immunohistochemistry (p38 MAPK) analysis. Rat treated with SA dose-dependently prevented behavioral alteration, restored antioxidant enzymes, neurotransmitters level, decreased neuroinflammatory markers, and improved neuronal integrity. Furthermore, immunohistochemistry confirmed the reduced p38 MAPK marker expression by SA in VPA induced autistic behavior.


Asunto(s)
Trastorno del Espectro Autista , Trastorno Autístico , Efectos Tardíos de la Exposición Prenatal , Embarazo , Femenino , Ratas , Animales , Humanos , Ácido Valproico/toxicidad , Trastorno Autístico/inducido químicamente , Trastorno Autístico/tratamiento farmacológico , Trastorno del Espectro Autista/inducido químicamente , Trastorno del Espectro Autista/tratamiento farmacológico , Antioxidantes/uso terapéutico , Proteínas Quinasas p38 Activadas por Mitógenos , Enfermedades Neuroinflamatorias , Ratas Wistar , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Modelos Animales de Enfermedad , Conducta Animal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA