Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 15.164
Filter
1.
Neurosci Lett ; 839: 137957, 2024 Sep 14.
Article in English | MEDLINE | ID: mdl-39218294

ABSTRACT

Post-traumatic stress disorder (PTSD) presents distinct sex-specific differences in both symptom expression and treatment outcomes, with the underlying biological mechanisms still remain unclear. Epigenetic modifications, particularly histone acetylation, have been increasingly recognized as critical factors in the pathophysiology of PTSD. Valproic acid (VPA), a potent histone deacetylase (HDAC) inhibitor, has shown promise in modulating epigenetic responses and improving therapeutic outcomes is PTSD, though its effect may differ between sexes. This study aimed to explore the sex-specific epigenetic changes in response to trauma and the impact of VPA treatment in a rat model of PTSD induced by predator scent stress. Sprague-Dawley rats of both sexes were randomly assigned to stressed and non-stressed groups and treated with either VPA (100 mg/kg) or vehicle. Anxiety levels were assessed using the elevated plus maze, followed by analysis of histone H3 and H4 acetylation, HDAC activity, and c-fos expression in the hippocampus. Our findings revealed that traumatic stress led to increased freezing time and anxiety levels, with more pronounced effects observed in females. Additionally, we have identified sex-specific differences in hippocampal epigenetic modifications; stressed females exhibited higher H3 acetylation, and VPA-treated stressed males showed increased H4 acetylation. These results highlight the importance of considering sex differences in the epigenetic mechanism underlying PTSD and suggest that personalized therapeutic approaches may be necessary to address these complexities.


Subject(s)
Epigenesis, Genetic , Histone Deacetylase Inhibitors , Rats, Sprague-Dawley , Stress Disorders, Post-Traumatic , Valproic Acid , Animals , Valproic Acid/pharmacology , Stress Disorders, Post-Traumatic/drug therapy , Stress Disorders, Post-Traumatic/genetics , Stress Disorders, Post-Traumatic/metabolism , Male , Female , Epigenesis, Genetic/drug effects , Rats , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Disease Models, Animal , Histones/metabolism , Sex Characteristics , Hippocampus/metabolism , Hippocampus/drug effects , Acetylation/drug effects , Anxiety/drug therapy
2.
Epilepsy Behav ; 159: 110028, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39217758

ABSTRACT

BACKGROUND: Aprepitant (APR), a neurokinin 1 receptor antagonist, is an approved drug for treating chemotherapy-induced nausea and vomiting. OBJECTIVES: Investigate the beneficial roles of APR alone or in combination with sodium valproate (VPA) against lithium pilocarpine [li-pilo]-induced seizures, behavioral changes, and cognitive deficits. METHODS: Thirty male mice were divided into five groups, each containing 6. "Vehicle Group I," "Control Group II "li-pilo, " Valproate (VPA) group III (400 mg/kg/i.p.), "APR group IV, " and "Combination Group V." Videos of mice were recorded, and they were watched for episodes of spontaneous recurring seizures (SRS). Behavioral Tests were performed. At the end of the study, animal brains were taken for biochemical assays and gene expression studies. RESULTS: APR partially protected against SRS with partial restoration of average behavioral and standard cognitive skills associated with a significant increase in brain SOD activity and a significant decrease in MDA, IL-1ß, NF-КB, and SP-3 levels in relation to the control group. Interestingly, a combination of APR with VPA in epileptic mice showed complete protection against li-pilo-induced behavioral changes and cognitive deficits, a significant increase in brain SOD activity, and a considerable decrease in MDA, IL-1ß, NF-ΚB, and SP levels to normal. CONCLUSION: Using APR as an adjuvant to VPA is more effective in protecting against li-pilo-induced seizures, behavioral changes, and cognitive deficits due to its antioxidant, anti-inflammatory, and NK1 antagonist effects than using APR alone as drug therapy.


Subject(s)
Anticonvulsants , Aprepitant , Disease Models, Animal , Epilepsy , Pilocarpine , Seizures , Valproic Acid , Animals , Male , Aprepitant/pharmacology , Mice , Valproic Acid/pharmacology , Anticonvulsants/pharmacology , Seizures/chemically induced , Seizures/drug therapy , Epilepsy/drug therapy , Epilepsy/chemically induced , Pilocarpine/toxicity , Morpholines/pharmacology , Brain/drug effects , Brain/metabolism , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Maze Learning/drug effects , Superoxide Dismutase/metabolism
3.
Epilepsy Behav ; 159: 110024, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39217754

ABSTRACT

PURPOSE: Prenatal exposure to antiseizure medications (ASMs) has been associated with an increased risk of major malformations and neurodevelopmental disorders, with the latter being mainly associated with valproate (VPA). Our aim was to compare neurocognitive outcome at age 6-7 years in children exposed prenatally to lamotrigine (LTG), carbamazepine (CBZ), valproate (VPA) or levetiracetam (LEV) monotherapy. METHODS: Eligible mother-child pairs were identified from the observational prospective multinational EURAP cohort study. Assessor-blinded testing was conducted at age 6-7 years using WISC-III and NEPSY-II. Verbal IQ (VIQ), performance IQ (PIQ), full scale IQ (FSIQ) and performance in neuropsychological tasks were compared across ASM groups by ANOVA. Scores were adjusted for maternal IQ, paternal education, maternal epilepsy type and child sex. RESULTS: Of 169 children enrolled in the study, 162 (LTG n = 80, CBZ n = 37, VPA n = 27, LEV n = 18) had sufficient data from WISC-III, NEPSY-II or both, and were included in the analyses. Observed (unadjusted) PIQ and FSIQ did not differ across exposure groups, but a difference was identified for VIQ (P<0.05), with children exposed to VPA having lower scores than children exposed to LEV (P<0.05) and children from all groups combined (P<0.01). Adjusted VIQ, PIQ and FSIQ scores did not differ significantly across groups, but VPA-exposed children had borderline significantly lower adjusted VIQ scores than children from all groups combined (P=0.051). VPA-exposed children had lower scores in comprehension of instructions before and after adjustment for confounding variables than children exposed to LTG (P<0.001), LEV (P<0.01) or children from all groups combined (p < 0.001). The VPA-exposed group also had lower scores in immediate and delayed memory for faces compared to children exposed to CBZ (P<0.05 and P<0.001, respectively) and LTG (P<0.05 and P<0.02, respectively), and children from all groups combined (P<0.02 and P<0.001, respectively). LEV-exposed children had lower scores in delayed memory for names than children exposed to LTG (P<0.001), CBZ (P<0.001), VPA (P<0.05) and children from all groups combined (P<0.001). CONCLUSIONS: Consistent with previous reports, our results provide evidence for an adverse effect of prenatal exposure to valproate on verbal development. Our finding of relatively weaker performance of VPA-exposed children compared to other ASM exposures in both comprehension of instructions and face memory also suggest that children of mothers treated with VPA are at increased risk for compromised memory functions or altered processing of socially relevant information.


Subject(s)
Anticonvulsants , Carbamazepine , Epilepsy , Lamotrigine , Levetiracetam , Prenatal Exposure Delayed Effects , Valproic Acid , Humans , Female , Prenatal Exposure Delayed Effects/chemically induced , Anticonvulsants/adverse effects , Child , Pregnancy , Male , Levetiracetam/adverse effects , Valproic Acid/adverse effects , Lamotrigine/adverse effects , Lamotrigine/therapeutic use , Carbamazepine/adverse effects , Epilepsy/drug therapy , Neuropsychological Tests , Triazines/adverse effects , Cohort Studies , Piracetam/analogs & derivatives , Piracetam/adverse effects , Adult , Cognition/drug effects , Prospective Studies , Intelligence/drug effects
4.
CNS Neurosci Ther ; 30(9): e70015, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39218796

ABSTRACT

INTRODUCTION: Neuroinflammation and microglial activation-related dendritic injury contribute to the pathogenesis of Autism Spectrum Disorder (ASD). Previous studies show that Progranulin (PGRN) is a growth factor associated with inflammation and synaptic development, but the role of PGRN in autism and the mechanisms underlying changes in PGRN expression remain unclear. AIMS: To investigate the impact of PGRN in autism, we stereotactically injected recombinant PGRN into the hippocampus of ASD model rats. Additionally, we explored the possibility that sortilin may be the factor behind the alterations in PGRN by utilizing SORT1 knockdown. Ultimately, we aimed to identify potential targets for the treatment of autism. RESULTS: PGRN could alleviate inflammatory responses, protect neuronal dendritic spines, and ameliorate autism-like behaviors. Meanwhile, elevated expression of sortilin and decreased levels of PGRN were observed in both ASD patients and rats. Enhanced sortilin levels facilitated PGRN internalization into lysosomes. Notably, suppressing SORT1 expression amplified PGRN levels, lessened microglial activation, and mitigated inflammation, thereby alleviating autism-like behaviors. CONCLUSION: Collectively, our findings highlight elevated sortilin levels in ASD rat brains, exacerbating dendrite impairment by affecting PGRN expression. PGRN supplementation and SORT1 knockdown hold potential as therapeutic strategies for ASD.


Subject(s)
Adaptor Proteins, Vesicular Transport , Autistic Disorder , Progranulins , Valproic Acid , Animals , Female , Humans , Male , Rats , Adaptor Proteins, Vesicular Transport/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Autistic Disorder/metabolism , Autistic Disorder/chemically induced , Autistic Disorder/drug therapy , Dendritic Spines/drug effects , Dendritic Spines/pathology , Dendritic Spines/metabolism , Hippocampus/metabolism , Hippocampus/drug effects , Microglia/metabolism , Microglia/drug effects , Progranulins/genetics , Rats, Sprague-Dawley , Valproic Acid/pharmacology
6.
Molecules ; 29(17)2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39275128

ABSTRACT

PURPOSE: Chronic wounds that are difficult to heal pose a major challenge for clinicians and researchers. Currently, common treatment methods focus on isolating the wound from the outside world, relying on the tissue at the wound site to grow and heal unaided. Umbilical cord mesenchymal stem cell (MSC) exosomes can promote wound healing by enhancing new blood vessel growth at the wound site. Valproic acid (VPA) reduces the inflammatory response and acts on macrophages to accelerate wound closure. In this study, VPA was loaded into umbilical cord MSC exosomes to form a drug carrier exosome (VPA-EXO) with the aim of investigating the effect of VPA-EXO on wound healing. METHODS: This study first isolated and obtained umbilical cord MSC exosomes, then added VPA to the exosomes and explored the ability of VPA-EXO to promote the proliferation and migration of human skin fibroblasts (HSFs) and human umbilical vein endothelial cells (HUVECs), as well as the ability to promote the angiogenesis of HUVECs, by using scratch, Transwell, and angiogenesis assays. An in vitro cell model was established and treated with VPA-EXO, and the expression levels of inflammation and pro-angiogenesis-related proteins and genes were examined using Western blot and qRT-PCR. The therapeutic effect of VPA-EXO on promoting wound healing in a whole skin wound model was investigated using image analysis of the wound site, H&E staining, and immunohistochemical staining experiments in a mouse wound model. RESULTS: The in vitro model showed that VPA-EXO effectively promoted the proliferation and migration of human skin fibroblast cells and human umbilical vein endothelial cells; significantly inhibited the expression of MMP-9, IL-1ß, IL-8, TNF-α, and PG-E2; and promoted the expression of vascular endothelial growth factors. In the mouse wound model, VPA-EXO reduced inflammation at the wound site, accelerated wound healing, and significantly increased the collagen content of tissue at the wound site. CONCLUSIONS: As a complex with dual efficacy in simultaneously promoting tissue regeneration and inhibiting inflammation, VPA-EXO has potential applications in tissue wound healing and vascular regeneration. In future studies, we will further investigate the mechanism of action and application scenarios of drug-loaded exosome complexes in different types of wound healing and vascular regeneration.


Subject(s)
Exosomes , Human Umbilical Vein Endothelial Cells , Inflammation , Mesenchymal Stem Cells , Neovascularization, Physiologic , Valproic Acid , Wound Healing , Valproic Acid/pharmacology , Wound Healing/drug effects , Exosomes/metabolism , Humans , Animals , Inflammation/drug therapy , Inflammation/metabolism , Mice , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/cytology , Neovascularization, Physiologic/drug effects , Cell Proliferation/drug effects , Cell Movement/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Angiogenesis
7.
Balkan Med J ; 41(5): 369-376, 2024 09 06.
Article in English | MEDLINE | ID: mdl-39239940

ABSTRACT

Background: Mesenchymal stem cells (MSCs) play a key role in regenerative medicine due to their capacity to differentiate into multiple cell lines, regulate the immune system, and exert paracrine effects. The therapeutic impact of MSCs is primarily mediated through their secretome. The secretory and therapeutic potential of MSCs can be improved through preconditioning, which entails the application of hypoxic environments, 3-dimensional cell cultures, and pharmacological agents. Valproic acid (VPA) is a histone deacetylase inhibitor that is employed in medical practice for treating epilepsy and bipolar disorder. Hence, preconditioning MSCs with VPA is expected to induce histone acetylation, enhance gene expression, and beneficially modify the cells' secretomes. Aims: To assess the effectiveness of VPA in enhancing and regulating the therapeutic potential of cells as well as its impact on MSC secretome profiles and ultrastructural morphologies. Study Design: Expiremental study. Methods: Human umbilical cord MSCs were preconditioned with 2 mM VPA for 24 and 48 hours; untreated MSCs served as controls. The secretome secreted by the cells was assessed for its total protein content. Subsequently, interferon-gamma (IFN-γ), interleukin-17 (IL-17), IL-10, vascular endothelial growth factor, nerve growth factor (NGF), glial cell line-derived neurotrophic factor, and brain-derived neurotrophic factor (BDNF) levels in the secretome were analyzed using the ELISA method. The ultrastructural properties of the cells were studied under transmission electron microscopy. Results: Ultrastructural examinations revealed that the chromatin content of VPA-treated cells was reduced. VPA-preconditioned cells exhibited a higher density of rough endoplasmic reticulum, autophagic vesicles, and myelin figures on cytoplasmic structure analysis, which was indicative of increased secretion. Protein secretion was elevated in those cells, with notable increases in NGF and BDNF levels. Furthermore, the cytoskeletal rearrangement and elevated autophagic activity observed in the 48-hour preconditioned cells could indicate the initiation of neuronal differentiation. IL-10, IL-17, and IFN-γ were not detected in the secretome. Conclusion: This study indicate that preconditioning with VPA enhances MSC activity and subsequently modifies the secretome content.


Subject(s)
Histone Deacetylase Inhibitors , Mesenchymal Stem Cells , Umbilical Cord , Valproic Acid , Humans , Valproic Acid/pharmacology , Mesenchymal Stem Cells/drug effects , Histone Deacetylase Inhibitors/pharmacology , Umbilical Cord/cytology , Umbilical Cord/drug effects , Cells, Cultured/drug effects
9.
BMC Neurol ; 24(1): 331, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39251922

ABSTRACT

BACKGROUND: This study aimed to compare the impact of olanzapine, magnesium valproate, and lamotrigine as adjunctive treatments for anti-N-methyl-D-aspartate receptor (NMDAR) encephalitis. And it is expected to add supporting points related to the rebalance of neurotransmitters in the brain through adjuvant therapy in the clinical management of anti-NMDAR encephalitis. METHODS: This retrospective study included patients diagnosed with anti-NMDAR encephalitis who received standardized immunotherapy at Hunan Brain Hospital between January 2018 and December 2020. RESULTS: Compared to the olanzapine group, both the magnesium valproate and lamotrigine groups showed lower scores on the positive and negative symptom scale (PANSS) total score after 3 weeks of treatment (all P < 0.05). The Montreal Cognitive Assessment Scale (MoCA) scores in the magnesium valproate and lamotrigine groups were significantly higher than in the olanzapine group after 3 weeks and 3 months of treatment (all P < 0.05). After 3 months of treatment, the proportions of patients with a modified Rankin scale score (mRS) of 0-1 in the magnesium valproate and lamotrigine groups were significantly higher than in the olanzapine group (all P < 0.05). The electroencephalogram (EEG) abnormality ranks at 3 months were significantly lower in the magnesium valproate and lamotrigine groups compared with the olanzapine group (all P < 0.05). Furthermore, the Glx/Cr ratio significantly decreased after 3 months of treatment (all P < 0.05) in the magnesium valproate and lamotrigine groups, while the Glx/Cr ratio in the olanzapine group showed no significant change (P > 0.05). CONCLUSION: Compared with olanzapine, the addition of magnesium valproate or lamotrigine to immunotherapy might be associated with a lower PANSS score, higher MoCA score, and lower mRS score. The improvement of neurological functions and cognitive function may be related to the decreased Glx/Cr ratio.


Subject(s)
Anti-N-Methyl-D-Aspartate Receptor Encephalitis , Lamotrigine , Olanzapine , Valproic Acid , Humans , Lamotrigine/therapeutic use , Retrospective Studies , Olanzapine/therapeutic use , Male , Female , Valproic Acid/therapeutic use , Adult , Anti-N-Methyl-D-Aspartate Receptor Encephalitis/drug therapy , Anti-N-Methyl-D-Aspartate Receptor Encephalitis/diagnosis , Young Adult , Middle Aged , Adolescent , Treatment Outcome , Anticonvulsants/therapeutic use
10.
Psychopharmacol Bull ; 54(4): 131-133, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39263199

ABSTRACT

Valproate and Autism complexity is manifold. From an established environmental risk factor for autism, to a translational animal model, valproate's composite mode of action might unfold to address core autistic domains transcending mere aggressive behavioural control.


Subject(s)
Autistic Disorder , Valproic Acid , Valproic Acid/adverse effects , Valproic Acid/administration & dosage , Humans , Animals , Autistic Disorder/chemically induced , Autistic Disorder/drug therapy , Anticonvulsants/adverse effects , Anticonvulsants/administration & dosage , Aggression/drug effects , Disease Models, Animal
11.
Neurologia (Engl Ed) ; 39(7): 549-554, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39232592

ABSTRACT

OBJECTIVE: There is early evidence about Valproic acid (VPA) antiviral effect. Our aim was to investigate the incidence and severity of SARS-CoV-2 infection in VPA users as compared with the general population. MATERIAL AND METHODS: A case-control study nested within a cohort, carried out between March 1 and December 17, 2020. Retrospectively, we identified confirmed SARS-CoV-2 infection patients exposed to VPA in our health department (defined as case). We ascertained VPA regimen (all the time (AT) (292 days) or at least 20% of the study period (notAT) (≥58 days) and if VPA levels were in therapeutic range (ATR) (50-100mcg/mL) in the last 24 months. We calculated the cumulative incidence of SARS-CoV-2 infection and hospital admission in the cases, comparing it with the general unexposed VPA population (controls). RESULTS: During the study period, 6183 PCR+ were detected among 281,035 inhabitants, of these, 746 were hospitalized. 691 patients were on VPA notAT and 628 (90.1%) AT. The indication for VPA use was epilepsy in 54.9%. The incidence of PCR+ was 1.736% (OR 0.785 (95%CI 0.443-1.390) and 1.910% (OR 0.865 (95%CI 0.488-1.533), on VPA notAT and VPA AT patients, respectively vs. 2.201% in people without VPA regimen. Those patients with VPA ATR had a lower risk of PCR + (OR 0.233 (95%CI 0.057-0.951) notAT; OR 0.218 (95%CI 0.053-0.890) AT). Hospital admission incidence was lower in patient on VPA (OR was 0.543 (95% CI 0.076-3.871). CONCLUSION: Patients with VPA within the therapeutic range had a reduction of SARS-Cov-2 infection incidence greater than 75%. There is a downward trend in the risk of COVID-19 admission by SARS-CoV-2 in patients on VPA therapy. These findings warrant further investigation.


Subject(s)
COVID-19 , Epilepsy , Valproic Acid , Humans , Valproic Acid/therapeutic use , Case-Control Studies , Male , Female , Middle Aged , COVID-19/epidemiology , Retrospective Studies , Adult , Aged , Epilepsy/drug therapy , COVID-19 Drug Treatment , Incidence , Antiviral Agents/therapeutic use , Anticonvulsants/therapeutic use , Hospitalization/statistics & numerical data , SARS-CoV-2
12.
Int. j. morphol ; 42(4): 1111-1118, ago. 2024. ilus, tab
Article in English | LILACS | ID: biblio-1569249

ABSTRACT

Epilepsy is the chronic non-communicable disease of the nervous system most prevalent in the world. Valproic acid (VPA) is one of the most used drugs in the treatment of epilepsy but with various side effects. One of the organs that can be affected is the testis, where it has been seen that men treated with VPA reduce their fertility rates, in addition to causing endocrine disorders by decreasing androgens and gonadotropins. In animal models, it has been shown to reduce the weights of the glands attached to the male reproductive tract, as well as at the testicular level, decreasing sperm concentration and increasing apoptotic cell count. These effects are because VPA increases reactive oxygen species (ROS), causing damage to macromolecules and affecting all cellular processes sensitive to oxide reduction. Throughout testicular development, in utero, it has been seen that the expression of antioxidant enzymes such as superoxide dismutase, catalase and glutathione peroxidase, are lower during early embryonic development, as well as vitamin E (VE) is decreased. Therefore, they are not sufficient to reverse the toxic effects of ROS. The objective of this study was to review the use of VPA during pregnancy, its effect on testicular development, and to explore the potential protective role of vitamin E.


La epilepsia es una enfermedad crónica no transmisible que afecta al sistema nervioso más prevalente en el mundo. Dentro de los tratamientos, uno de los fármacos más utilizados es el ácido valproico (AVP), el que ocasiona diversos efectos secundarios. Entre los órganos que se pueden ver afectados se encuentra la gónada masculina, en donde se ha visto que hombres en tratamiento con AVP reducen sus tasas de fecundidad, además de causar trastornos endocrinos disminuyendo andrógenos y gonadotrofinas. En modelos animales, se ha visto que disminuye los pesos de las glándulas anexas al tracto reproductor masculino, como también a nivel testicular, disminuyendo la concentración espermática y aumentando el recuento de células apoptóticas. Estos efectos se deberían a que el AVP aumenta las especies reactivas de oxígeno (ROS), ocasionando daño en macromoléculas, afectando todos los procesos celulares sensibles a óxido reducción. A lo largo del desarrollo testicular, in utero se ha visto que la expresión de enzimas antioxidantes como superóxido dismutasa, catalasa y glutatión peroxidasa, son más bajos durante el desarrollo embrionario temprano, como también la vitamina E (VE) se encuentra disminuida. Por tanto, no resultan suficientes para revertir los efectos tóxicos de las ROS. El objetivo de esta revisión fue asociar el uso de AVP durante la gestación y sus efectos a nivel del desarrollo testicular y describir el potencial rol protector de la VE.


Subject(s)
Humans , Animals , Male , Female , Pregnancy , Testis/drug effects , Vitamin E/pharmacology , Valproic Acid/adverse effects , Teratogens , Testis/growth & development , Valproic Acid/toxicity , Reactive Oxygen Species , Epilepsy/drug therapy , Embryonic and Fetal Development/drug effects
13.
Cells ; 13(16)2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39195239

ABSTRACT

Autism spectrum disorder (ASD) is a multifactorial neurodevelopmental condition with several identified risk factors, both genetic and non-genetic. Among these, prenatal exposure to valproic acid (VPA) has been extensively associated with the development of the disorder. The zebrafish, a cost- and time-effective model, is useful for studying ASD features. Using validated VPA-induced ASD zebrafish models, we aimed to provide new insights into VPA exposure effects during embryonic development and to identify new potential biomarkers associated with ASD-like features. Dose-response analyses were performed in vivo to study larval phenotypes and mechanisms underlying neuroinflammation, mitochondrial dysfunction, oxidative stress, microglial cell status, and motor behaviour. Wild-type and transgenic Tg(mpeg1:EGFP) zebrafish were water-exposed to VPA doses (5 to 500 µM) from 6 to 120 h post-fertilisation (hpf). Embryos and larvae were monitored daily to assess survival and hatching rates, and numerous analyses and tests were conducted from 24 to 120 hpf. VPA doses higher than 50 µM worsened survival and hatching rates, while doses of 25 µM or more altered morphology, microglial status, and larval behaviours. VPA 50 µM also affected mRNA expression of inflammatory cytokines and neurogenesis-related genes, mitochondrial respiration, and reactive oxygen species accumulation. The study confirmed that VPA alters brain homeostasis, synaptic interconnections, and neurogenesis-related signalling pathways, contributing to ASD aetiopathogenesis. Further studies are essential to identify novel ASD biomarkers for developing new drug targets and tailored therapeutic interventions for ASD.


Subject(s)
Autism Spectrum Disorder , Disease Models, Animal , Valproic Acid , Zebrafish , Animals , Valproic Acid/pharmacology , Valproic Acid/adverse effects , Autism Spectrum Disorder/chemically induced , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/pathology , Larva/drug effects , Animals, Genetically Modified , Oxidative Stress/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Microglia/drug effects , Microglia/pathology , Microglia/metabolism , Brain/drug effects , Brain/pathology , Brain/metabolism , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Neurogenesis/drug effects
14.
Autism Res ; 17(8): 1534-1544, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39169698

ABSTRACT

Autism spectrum disorders (ASDs) are characterized by core behavioral symptoms in the domains of sociability, language/communication, and repetitive or stereotyped behaviors. Deficits in the prefrontal and hippocampal excitatory/inhibitory balance due to a functional loss of GABAergic interneurons are proposed to underlie these symptoms. Increasing the postsynaptic effects of GABA with compounds that selectively modulate GABAergic receptors could be a potential target for treating ASD symptoms. In addition, deficits in GABAergic interneurons have been linked to dopamine (DA) system dysregulation, and, despite conflicting evidence, abnormalities in the DA system activity may underly some ASD symptoms. Here, we investigated whether the positive allosteric modulator of α5-containing GABAA receptors (α5-GABAARs) SH-053-2'F-R-CH3 (10 mg/kg) attenuates behavioral abnormalities in rats exposed to valproic acid (VPA) in utero, an established risk factor for autism. We also evaluated if animals exposed to VPA in utero present changes in the ventral tegmental area (VTA) DA system activity using in vivo electrophysiology and if SH-053-2'F-R-CH3 could attenuate these changes. SH-053-2'F-R-CH3 was administered intraperitoneally 30 min before each behavioral test and electrophysiology. In utero VPA exposure caused male and female rats to present increased repetitive behavior (self-grooming) in early adolescence and deficits in social interaction in adulthood. Male, but not female VPA rats, also presented deficits in recognition memory as adults. SH-053-2'F-R-CH3 attenuated the impairments in sociability and cognitive function in male VPA-exposed rats without attenuating the decreased social interaction in females. Adult male and female VPA-exposed rats also showed an increased VTA DA neuron population activity, which was not changed by SH-053-2'F-R-CH3. Despite sex differences, our findings indicate that α5-GABAARs positive allosteric modulators may effectively attenuate some core ASD symptoms.


Subject(s)
Prenatal Exposure Delayed Effects , Receptors, GABA-A , Social Behavior , Valproic Acid , Animals , Female , Valproic Acid/pharmacology , Rats , Male , Pregnancy , Receptors, GABA-A/drug effects , Dopamine/metabolism , Autism Spectrum Disorder/chemically induced , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/physiopathology , Rats, Sprague-Dawley , Allosteric Regulation/drug effects , Disease Models, Animal , Behavior, Animal/drug effects , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/physiopathology
15.
Elife ; 122024 Aug 27.
Article in English | MEDLINE | ID: mdl-39190448

ABSTRACT

Dravet syndrome (DS) is a devastating early-onset refractory epilepsy syndrome caused by variants in the SCN1A gene. A disturbed GABAergic interneuron function is implicated in the progression to DS but the underlying developmental and pathophysiological mechanisms remain elusive, in particularly at the chromatin level. Induced pluripotent stem cells (iPSCs) derived from DS cases and healthy donors were used to model disease-associated epigenetic abnormalities of GABAergic development. Chromatin accessibility was assessed at multiple time points (Day 0, Day 19, Day 35, and Day 65) of GABAergic differentiation. Additionally, the effects of the commonly used anti-seizure drug valproic acid (VPA) on chromatin accessibility were elucidated in GABAergic cells. The distinct dynamics in the chromatin profile of DS iPSC predicted accelerated early GABAergic development, evident at D19, and diverged further from the pattern in control iPSC with continued differentiation, indicating a disrupted GABAergic maturation. Exposure to VPA at D65 reshaped the chromatin landscape at a variable extent in different iPSC-lines and rescued the observed dysfunctional development of some DS iPSC-GABA. The comprehensive investigation on the chromatin landscape of GABAergic differentiation in DS-patient iPSC offers valuable insights into the epigenetic dysregulations associated with interneuronal dysfunction in DS. Moreover, the detailed analysis of the chromatin changes induced by VPA in iPSC-GABA holds the potential to improve the development of personalized and targeted anti-epileptic therapies.


Subject(s)
Cell Differentiation , Epigenesis, Genetic , Epilepsies, Myoclonic , GABAergic Neurons , Induced Pluripotent Stem Cells , Valproic Acid , Induced Pluripotent Stem Cells/metabolism , Humans , Epilepsies, Myoclonic/genetics , Epilepsies, Myoclonic/drug therapy , Epilepsies, Myoclonic/metabolism , Valproic Acid/pharmacology , Cell Differentiation/drug effects , GABAergic Neurons/metabolism , GABAergic Neurons/drug effects , Chromatin/metabolism , NAV1.1 Voltage-Gated Sodium Channel/genetics , NAV1.1 Voltage-Gated Sodium Channel/metabolism , Anticonvulsants/pharmacology
16.
Birth Defects Res ; 116(8): e2392, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39189597

ABSTRACT

On January 12, 2024 the safety committee of the European Medicines Agency (EMA) recommended precautionary measures over a potential risk of neurodevelopmental disorders in children born to men treated with valproate. These new measures recommend patient supervision by a specialist in the management of epilepsy, bipolar disorder, or migraine. In the United Kingdom, the Medicines and Healthcare products Regulatory Agency (MHRA) issued a far more stringent precaution, warning against prescribing valproate to anyone under 55 years of age. We, members of the European Network of Teratology Information Services (ENTIS) and the Organization of Teratology Information Specialists (OTIS), believe that the EMA and MHRA warnings were premature. We are of the opinion that the underlying scientific data do not convincingly substantiate the inference of a paternally mediated risk from valproate to children, much less to an extent that justifies these far-reaching recommendations.


Subject(s)
Neurodevelopmental Disorders , Valproic Acid , Humans , Valproic Acid/adverse effects , Neurodevelopmental Disorders/prevention & control , Neurodevelopmental Disorders/chemically induced , Male , Child , Epilepsy/drug therapy , United Kingdom , Anticonvulsants/adverse effects , Anticonvulsants/therapeutic use , Female
17.
CNS Drugs ; 38(9): 733-742, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39096467

ABSTRACT

BACKGROUND AND OBJECTIVES: Cenobamate (CNB) is a new antiseizure medication (ASM) to treat drug-resistant, focal-onset seizures. Data on its use in early therapy lines are not yet available, and clinicians frequently consider CNB to be a later ASM drug choice. We investigated the efficacy and safety of CNB as an early adjunctive treatment in drug-resistant, focal-onset seizures. METHODS: The study population were patients with drug-resistant, focal-onset seizures who were initiated with CNB after they did not respond to two or three lifetime ASMs, including all prior and concomitant ASMs. These patients were matched (1:2) by sex, age, and seizure frequency to controls who were initiated with any ASM other than CNB. All participants participated in the Mainz Epilepsy Registry. We evaluated the retention rate after 12 months of CNB and after each new adjunctive ASM in the control group. In addition, seizure freedom and the response rate (reduction of seizure frequency by ≥ 50% from baseline) after 12 months were estimated. RESULTS: We included 231 patients aged 44.4 ± 15.8 years. Of these, 33.3% (n = 77) were on CNB, 19.0% (n = 44) on valproate (VPA), 17.3% (n = 40) on lacosamide (LCS), 16.4% (n = 38) on levetiracetam (LEV), and 13.9% (n = 32) on topiramate (TPM). The highest retention rate after 12 months since the beginning of the early adjunctive therapy was observed on CNB (92.0%), compared with LCS (80.0%), LEV (73.3%), VPA (68.2%), or TPM (62.5%) (p < 0.05). Seizure freedom and response rate were also the best on CNB (19.5% and 71.4%, respectively) compared with other ASMs (8.3% and 52.5%, respectively; p < 0.05). No significant differences in adverse events between CNB and other ASMs were observed. CONCLUSIONS: Our study provides evidence that CNB is an effective ASM with a good safety profile in the early therapy lines of drug-resistant, focal-onset seizures. This data should support medical decision making in the management of patients with refractory epilepsy. CLINICAL TRIAL ID: NCT05267405.


Subject(s)
Anticonvulsants , Carbamates , Chlorophenols , Drug Resistant Epilepsy , Drug Therapy, Combination , Seizures , Humans , Male , Anticonvulsants/administration & dosage , Anticonvulsants/therapeutic use , Anticonvulsants/adverse effects , Female , Adult , Carbamates/therapeutic use , Carbamates/adverse effects , Carbamates/administration & dosage , Middle Aged , Cohort Studies , Drug Resistant Epilepsy/drug therapy , Seizures/drug therapy , Chlorophenols/administration & dosage , Chlorophenols/adverse effects , Chlorophenols/therapeutic use , Treatment Outcome , Tetrazoles/administration & dosage , Tetrazoles/therapeutic use , Tetrazoles/adverse effects , Valproic Acid/therapeutic use , Valproic Acid/administration & dosage , Valproic Acid/adverse effects
18.
Proc Natl Acad Sci U S A ; 121(35): e2406928121, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39178233

ABSTRACT

Autism spectrum disorders (ASD) can be caused by environmental factors. These factors act early in the development of the nervous system and induce stereotyped repetitive behaviors and diminished social interactions, among other outcomes. Little is known about how these behaviors are produced. In pregnant women, delivery of valproic acid (VPA) (to control seizure activity or stabilize mood) or immune activation by a virus increases the incidence of ASD in offspring. We found that either VPA or Poly Inosine:Cytosine (which mimics a viral infection), administered at mouse embryonic day 12.5, induced a neurotransmitter switch from GABA to glutamate in PV- and CCK-expressing interneurons in the medial prefrontal cortex by postnatal day 10. The switch was present for only a brief period during early postnatal development, observed in male and female mice at postnatal day 21 and reversed in both males and females by postnatal day 30. At postnatal day 90, male mice exhibited stereotyped repetitive behaviors and diminished social interaction while female mice exhibited only stereotyped repetitive behavior. Transfecting GAD1 in PV- and CCK-expressing interneurons at postnatal day 10, to reintroduce GABA expression, overrode the switch and prevented expression of autistic-like behavior. These findings point to an important role of neurotransmitter switching in mediating the environmental causes of autism.


Subject(s)
Valproic Acid , gamma-Aminobutyric Acid , Animals , Female , Mice , Male , Pregnancy , Valproic Acid/toxicity , gamma-Aminobutyric Acid/metabolism , Interneurons/metabolism , Animals, Newborn , Behavior, Animal , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/pathology , Glutamate Decarboxylase/metabolism , Glutamate Decarboxylase/genetics , Autistic Disorder/etiology , Autistic Disorder/metabolism , Glutamic Acid/metabolism , Neurotransmitter Agents/metabolism , Poly I-C , Prefrontal Cortex/metabolism , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/etiology , Autism Spectrum Disorder/pathology , Cholecystokinin/metabolism , Parvalbumins/metabolism , Mice, Inbred C57BL , Stereotyped Behavior/drug effects
19.
PLoS One ; 19(8): e0307154, 2024.
Article in English | MEDLINE | ID: mdl-39093886

ABSTRACT

The SARS-CoV-2 pandemic has caused unprecedented worldwide infections from persistent mutant variants with various degrees of infectivity and virulence. The elusiveness of a highly penetrant, worldwide vaccination strategy suggests that the complete eradication of SARS-CoV-2 is unlikely. Even with the advent of new antiviral agents, the disease burden worldwide continues to exceed current preventative and therapeutic strategies. Greater interest has been placed towards the development of affordable,broadly effective antiviral therapeutics. Here, we report that the small branched-chain fatty acid Valproic acid (VPA), approved for maintenance of seizure and bipolar disorder, has a novel anti- coronavirus activity that can be augmented with the addition of a long-chain, polyunsaturated omega-3 fatty acid, Docosahexaenoic acid (DHA). An EMR-based epidemiological study of patients tested for COVID-19 demonstrated a correlation exists between a reduced infection rate in patients treated withVPA of up to 25%, as well as a decreased risk of emergency room visits, hospitalization, ICU admission,and use of mechanical ventilation. In vitro studies have demonstrated that VPA modifies gene expression in MRC5 cells. Interestingly, VPA correlates with the inhibition of several SARS-CoV2 interacting genes and the greater inhibition of alpha-coronavirus HCoV-229E (a "common cold" virus) and SARS-CoV2. The VPA-DHA combination activates pre-existing intracellular antiviral mechanisms normally repressed by coronaviruses. Gene expression profiles demonstrate subtle differences in overall gene expression between VPA-treated and VPA-DHA-treated cells. HCoV-229E infection caused an intensely different response with a marked induction of multiple intracellular inflammatory genes. Changes in gene expression took at least 24 hours to manifest and most likely why prior drug screens failed to identify any antiviral VPA activity despite in silico predictions. This report demonstrates an interaction between HDAC inhibition and the potent activation of cellular antiviral responses. A foundation now exists for a low-cost, highly effective antiviral strategy when supplemented with DHA.


Subject(s)
Antiviral Agents , COVID-19 , SARS-CoV-2 , Valproic Acid , Valproic Acid/pharmacology , Valproic Acid/therapeutic use , Humans , COVID-19/epidemiology , COVID-19/virology , SARS-CoV-2/drug effects , Antiviral Agents/therapeutic use , Antiviral Agents/pharmacology , COVID-19 Drug Treatment , Docosahexaenoic Acids/pharmacology , Docosahexaenoic Acids/therapeutic use , Severity of Illness Index , Male , Female , Middle Aged
20.
Eur J Pharm Sci ; 201: 106876, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39128815

ABSTRACT

BACKGROUND: Valproic acid (VPA) is a commonly used broad-spectrum antiepileptic drug. For elderly epileptic patients, VPA plasma concentrations have a considerable variation. We aim to establish a prediction model via a combination of machine learning and population pharmacokinetics (PPK) for VPA plasma concentration. METHODS: A retrospective study was performed incorporating 43 variables, including PPK parameters. Recursive Feature Elimination with Cross-Validation was used for feature selection. Multiple algorithms were employed for ensemble model, and the model was interpreted by Shapley Additive exPlanations. RESULTS: The inclusion of PPK parameters significantly enhances the performance of individual algorithm model. The composition of categorical boosting, light gradient boosting machine, and random forest (7:2:1) with the highest R2 (0.74) was determined as the ensemble model. The model included 11 variables after feature selection, of which the predictive performance was comparable to the model that incorporated all variables. CONCLUSIONS: Our model was specifically tailored for elderly epileptic patients, providing an efficient and cost-effective approach to predict VPA plasma concentration. The model combined classical PPK with machine learning, and underwent optimization through feature selection and algorithm integration. Our model can serve as a fundamental tool for clinicians in determining VPA plasma concentration and individualized dosing regimens accordingly.


Subject(s)
Anticonvulsants , Epilepsy , Machine Learning , Valproic Acid , Humans , Valproic Acid/pharmacokinetics , Valproic Acid/blood , Epilepsy/drug therapy , Epilepsy/blood , Anticonvulsants/pharmacokinetics , Anticonvulsants/blood , Anticonvulsants/administration & dosage , Aged , Retrospective Studies , Male , Female , Models, Biological , Aged, 80 and over , Algorithms , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL