Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.122
Filter
1.
Bull Exp Biol Med ; 177(1): 47-50, 2024 May.
Article in English | MEDLINE | ID: mdl-38955852

ABSTRACT

Ectonucleotidases play an important role in regulating the level of extracellular nucleotides and nucleosides and are an important part of the regulation of the effects of adenosine and ATP on adenosine and P2 receptors, respectively. We have previously established the ambiguous effect of P2 receptor agonists on the contractile activity of smooth muscle tissue in rats with the valproate model of autism. In this work, HPLC was used to evaluate the activity of ectonucleotidases in the smooth muscle tissues of the internal organs of rats with a valproate model of autism. The activity of ectonucleotidases was significantly higher in the smooth muscle tissues of the duodenum, vas deferens, and bladder, but lower in the ileum and uterus. The results obtained make it possible to compare the activity of ectonucleotidases identified here with changes in P2 receptor-mediated contractility of smooth muscle tissues revealed in our previous experiments.


Subject(s)
Autistic Disorder , Muscle Contraction , Muscle, Smooth , Urinary Bladder , Valproic Acid , Vas Deferens , Animals , Rats , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Valproic Acid/pharmacology , Autistic Disorder/metabolism , Autistic Disorder/chemically induced , Autistic Disorder/drug therapy , Male , Female , Vas Deferens/drug effects , Vas Deferens/metabolism , Urinary Bladder/drug effects , Urinary Bladder/metabolism , Urinary Bladder/enzymology , Muscle Contraction/drug effects , Uterus/drug effects , Uterus/metabolism , Ileum/drug effects , Ileum/metabolism , Ileum/enzymology , Disease Models, Animal , Rats, Wistar , Receptors, Purinergic P2/metabolism , Adenosine Triphosphatases/metabolism
2.
Arch Biochem Biophys ; 758: 110084, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38971420

ABSTRACT

Nuclear factor erythroid factor 2 (Nrf2) is the key regulatory of the antioxidant response elements. Also, Nrf2 interacts with nuclear factor kappa B (NF-ĸB) to inhibit subsequent inflammatory cascade. Activation of Nrf2 signaling ameliorates drug-induced liver injury. Sodium valproate (SVP) is an anti-epilepsy drug with a hepatotoxic adverse effect that restricts its clinical use. In this study, coadministration of Dihydromyricetin (DHM), a natural flavonoid, with SVP to rats upregulated gene expression of Nrf2 and its downstream gene, heme oxygenase 1 (HO-1), while suppressed the Nrf2 repressor, Keap-1. Additionally, DHM led to downregulation of proinflammatory factors in liver tissues, including NF-ĸB, interleukin 1 beta (IL-1ß), and tumor necrosis factor alpha (TNF-α). This was accompanied by a decrease in the proapoptotic protein (cleaved caspase-3) expression level. Furthermore, biochemical and histopathological studies showed that DHM treatment improved liver function and lipid profile while decreased inflammatory cell infiltration, congestion, and hepatocellular damage. According to our knowledge, prior research has not examined the protective effect of DHM on the liver injury induced by SVP. Consequently, this study provides DHM as a promising herbal medication that, when used with SVP, can prevent its induced hepatotoxicity owing to its potential anti-oxidative, anti-inflammatory, and anti-apoptotic properties.


Subject(s)
Caspase 3 , Chemical and Drug Induced Liver Injury , Flavonols , Kelch-Like ECH-Associated Protein 1 , NF-E2-Related Factor 2 , NF-kappa B , Signal Transduction , Valproic Acid , Animals , NF-E2-Related Factor 2/metabolism , Male , Signal Transduction/drug effects , Flavonols/pharmacology , NF-kappa B/metabolism , Valproic Acid/pharmacology , Rats , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/prevention & control , Chemical and Drug Induced Liver Injury/pathology , Kelch-Like ECH-Associated Protein 1/metabolism , Caspase 3/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Rats, Sprague-Dawley , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase-1/metabolism
3.
Birth Defects Res ; 116(7): e2379, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38958465

ABSTRACT

BACKGROUND: Autism spectrum disorder (ASD) is the fastest-growing child neuropsychiatric condition. Cognitive dysfunctions such as memory impairments are experienced by patients along with social disturbances and repetitive/stereotypic movements. We have used the radial arm maze (RAM), for measurement of working and reference memory errors in an animal model of autism. In addition, the potential effects of agmatine, an endogenous NMDA antagonist, on RAM performance and autistic-like behaviors were assessed. METHODS: Autism was modeled by valproic acid (VPA) administration at gestational Day 12.5. Autism-associated behaviors in male offspring were examined in an open field test (OFT) and three-chambered test (TCT) on postnatal days 50-51. Thereafter, the animals were trained in the RAM (PND 55) until they attained the criteria of 80% correct choices during five consecutive trials. Forty-eight hours after the acquisition of criteria, agmatine was injected 30 min before subsequent behavioral testing, which included the retention phase of the RAM, OFT, and TCT. RESULTS: VPA-treated and intact rats showed the same performance in RAM, and acute injection of agmatine rescued social and anxiety-like behavior induced by VPA without the effect on RAM. CONCLUSION: In a rat model of autism, spatial learning, and memory did not change. Agmatine rescued social and anxiety-like behavior in autistic animals.


Subject(s)
Agmatine , Autistic Disorder , Behavior, Animal , Disease Models, Animal , Maze Learning , Animals , Agmatine/pharmacology , Male , Rats , Maze Learning/drug effects , Autistic Disorder/drug therapy , Autistic Disorder/psychology , Behavior, Animal/drug effects , Memory/drug effects , Valproic Acid/pharmacology , Female , Pregnancy
4.
Commun Biol ; 7(1): 851, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38992101

ABSTRACT

In autism spectrum disorder (ASD), atypical sensory experiences are often associated with irregularities in predictive coding, which proposes that the brain creates hierarchical sensory models via a bidirectional process of predictions and prediction errors. However, it remains unclear how these irregularities manifest across different functional hierarchies in the brain. To address this, we study a marmoset model of ASD induced by valproic acid (VPA) treatment. We record high-density electrocorticography (ECoG) during an auditory task with two layers of temporal control, and applied a quantitative model to quantify the integrity of predictive coding across two distinct hierarchies. Our results demonstrate a persistent pattern of sensory hypersensitivity and unstable predictions across two brain hierarchies in VPA-treated animals, and reveal the associated spatio-spectro-temporal neural signatures. Despite the regular occurrence of imprecise predictions in VPA-treated animals, we observe diverse configurations of underestimation or overestimation of sensory regularities within the hierarchies. Our results demonstrate the coexistence of the two primary Bayesian accounts of ASD: overly-precise sensory observations and weak prior beliefs, and offer a potential multi-layered biomarker for ASD, which could enhance our understanding of its diverse symptoms.


Subject(s)
Autism Spectrum Disorder , Brain , Callithrix , Disease Models, Animal , Animals , Autism Spectrum Disorder/physiopathology , Autism Spectrum Disorder/chemically induced , Brain/physiopathology , Brain/drug effects , Male , Valproic Acid/pharmacology , Electrocorticography
5.
Transl Psychiatry ; 14(1): 272, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38961057

ABSTRACT

Valproic acid (VPA) is one of the most effective antiepileptic drugs, and exposing animals to VPA during gestation has been used as a model for autism spectrum disorder (ASD). Numerous studies have shown that impaired synaptic transmission in the cerebellar cortical circuits is one of the reasons for the social deficits and repetitive behavior seen in ASD. In this study, we investigated the effect of VPA exposure during pregnancy on tactile stimulation-evoked cerebellar mossy fiber-granule cell (MF-GC) synaptic transmission in mice anesthetized with urethane. Three-chamber testing showed that mice exposed to VPA mice exhibited a significant reduction in social interaction compared with the control group. In vivo electrophysiological recordings revealed that a pair of air-puff stimulation on ipsilateral whisker pad evoked MF-GC synaptic transmission, N1, and N2. The evoked MF-GC synaptic responses in VPA-exposed mice exhibited a significant increase in the area under the curve (AUC) of N1 and the amplitude and AUC of N2 compared with untreated mice. Cerebellar surface application of the selective N-methyl-D-aspartate (NMDA) receptor blocker D-APV significantly inhibited facial stimulation-evoked MF-GC synaptic transmission. In the presence of D-APV, there were no significant differences between the AUC of N1 and the amplitude and AUC of N2 in the VPA-exposed mice and those of the untreated mice. Notably, blockade of the GluN2A subunit-containing, but not the GluN2B subunit-containing, NMDA receptor, significantly inhibited MF-GC synaptic transmission and decreased the AUC of N1 and the amplitude and AUC of N2 in VPA-exposed mice to levels similar to those seen in untreated mice. In addition, the GluN2A subunit-containing NMDA receptor was expressed at higher levels in the GC layer of VPA-treated mice than in control mice. These results indicate that gestational VPA exposure in mice produces ASD-like behaviors, accompanied by increased cerebellar MF-GC synaptic transmission and an increase in GluN2A subunit-containing NMDA receptor expression in the offspring.


Subject(s)
Autism Spectrum Disorder , Disease Models, Animal , Prenatal Exposure Delayed Effects , Receptors, N-Methyl-D-Aspartate , Synaptic Transmission , Valproic Acid , Animals , Receptors, N-Methyl-D-Aspartate/metabolism , Valproic Acid/pharmacology , Pregnancy , Female , Mice , Prenatal Exposure Delayed Effects/physiopathology , Synaptic Transmission/drug effects , Autism Spectrum Disorder/chemically induced , Male , Cerebellum/drug effects , Cerebellum/metabolism , Anticonvulsants/pharmacology
6.
Sci Rep ; 14(1): 13168, 2024 06 07.
Article in English | MEDLINE | ID: mdl-38849397

ABSTRACT

Autism spectrum disorder (ASD) is a pervasive neurodevelopmental condition characterized by social interaction deficits, communication impairments, repetitive behaviors, and sensory sensitivities. While the etiology of ASD is multifaceted, abnormalities in glutamatergic neurotransmission and synaptic plasticity have been implicated. This study investigated the role of metabotropic glutamate receptor 8 (mGlu8) in modulating long-term potentiation (LTP) in a rat model of ASD induced by prenatal valproic acid (VPA) exposure. To induce an animal model with autism-like characteristics, pregnant rats received an intraperitoneal injection of 500 mg/kg of sodium valproate (NaVPA) on embryonic day 12.5. High-frequency stimulation was applied to the perforant path-dentate gyrus (PP-DG) synapse to induce LTP, while the mGlu8 receptor agonist (S)-3,4-dicarboxyphenylglycine (DCPG) was administered into the DG. The results revealed that VPA-exposed rats exhibited reduced LTP compared to controls. DCPG had contrasting effects, inhibiting LTP in controls and enhancing it in VPA-exposed rats. Moreover, reduced social novelty preference index (SNPI) in VPA-exposed rats was reversed by intra-DG administration of S-3,4-DCPG. In conclusion, our study advances our understanding of the complex relationship between glutamatergic neurotransmission, synaptic plasticity, and VPA-induced autism model. The findings suggest that mGlu8 receptor dysfunction plays a role in the impaired synaptic plasticity seen in ASD.


Subject(s)
Dentate Gyrus , Disease Models, Animal , Long-Term Potentiation , Prenatal Exposure Delayed Effects , Receptors, Metabotropic Glutamate , Synapses , Valproic Acid , Animals , Valproic Acid/pharmacology , Valproic Acid/adverse effects , Long-Term Potentiation/drug effects , Female , Pregnancy , Rats , Dentate Gyrus/drug effects , Synapses/drug effects , Synapses/metabolism , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/metabolism , Prenatal Exposure Delayed Effects/chemically induced , Perforant Pathway/drug effects , Autistic Disorder/chemically induced , Glycine/analogs & derivatives , Glycine/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Rats, Sprague-Dawley , Autism Spectrum Disorder/chemically induced , Male
7.
Viruses ; 16(6)2024 May 30.
Article in English | MEDLINE | ID: mdl-38932169

ABSTRACT

Repression of human cytomegalovirus (HCMV) immediate-early (IE) gene expression is a key regulatory step in the establishment and maintenance of latent reservoirs. Viral IE transcription and protein accumulation can be elevated during latency by treatment with histone deacetylase inhibitors such as valproic acid (VPA), rendering infected cells visible to adaptive immune responses. However, the latency-associated viral protein UL138 inhibits the ability of VPA to enhance IE gene expression during infection of incompletely differentiated myeloid cells that support latency. UL138 also limits the accumulation of IFNß transcripts by inhibiting the cGAS-STING-TBK1 DNA-sensing pathway. Here, we show that, in the absence of UL138, the cGAS-STING-TBK1 pathway promotes both IFNß accumulation and VPA-responsive IE gene expression in incompletely differentiated myeloid cells. Inactivation of this pathway by either genetic or pharmacological inhibition phenocopied UL138 expression and reduced VPA-responsive IE transcript and protein accumulation. This work reveals a link between cytoplasmic pathogen sensing and epigenetic control of viral lytic phase transcription and suggests that manipulation of pattern recognition receptor signaling pathways could aid in the refinement of MIEP regulatory strategies to target latent viral reservoirs.


Subject(s)
Cytomegalovirus , Membrane Proteins , Myeloid Cells , Nucleotidyltransferases , Protein Serine-Threonine Kinases , Signal Transduction , Valproic Acid , Humans , Valproic Acid/pharmacology , Myeloid Cells/virology , Myeloid Cells/metabolism , Myeloid Cells/drug effects , Signal Transduction/drug effects , Membrane Proteins/metabolism , Membrane Proteins/genetics , Cytomegalovirus/physiology , Cytomegalovirus/drug effects , Cytomegalovirus/genetics , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Cytomegalovirus Infections/virology , Cytomegalovirus Infections/metabolism , Cytomegalovirus Infections/genetics , Virus Latency/drug effects , Transcription, Genetic/drug effects , Cell Differentiation/drug effects , Gene Expression Regulation, Viral/drug effects , Genes, Immediate-Early , Interferon-beta/metabolism , Interferon-beta/genetics
8.
Rev Col Bras Cir ; 51: e20243676, 2024.
Article in English, Portuguese | MEDLINE | ID: mdl-38896636

ABSTRACT

INTRODUCTION: valproic acid (VPA), an epigenetic drug, has potential for the treatment of neoplasms. Its effects on the healing of the peritoneal-musculo-aponeurotic plane (PMA) of the abdominal wall are studied. METHOD: sixty Wistar rats were allocated into two groups: experimental (VPA) and control (0.9% sodium chloride), treated daily, starting three days before the intervention and until euthanasia. Under anesthesia, a median laparotomy was performed and repaired with two synthetic layers. Assessments took place 3, 7 and 14 days after surgery. The integrity of the wounds, the quality of the inflammatory reaction, the intensity of the leukocyte infiltrate, collagen synthesis, the intensity of angiogenesis and the presence of myofibroblasts were studied. RESULTS: there was dehiscence of the PMA plane in 11 of the 30 animals (p=0.001) in the experimental group. There was no difference in the quality and intensity of the inflammatory reaction. Immunohistochemistry revealed, in the experimental group, less collagen I (p3=0.003, p7=0.013 and p14=0.001) and more collagen III (p3=0.003, p7=0.013 and p14= 0.001). Collagen evaluated by Sirus Supra Red F3BA showed, in the experimental group, less collagen at all three times (p<0.001) with less collagen I and collagen III (p<0.001). A lower number of vessels was found on the 3rd day (p<0.001) and on the 7th day (p=0.001) and did not affect the number of myofibroblasts. CONCLUSION: VPA showed dehiscence of the PMA plane, with less deposition of total collagen and collagen I, less angiogenic activity, without interfering with the number of myofibroblasts.


Subject(s)
Abdominal Wall , Rats, Wistar , Valproic Acid , Wound Healing , Animals , Wound Healing/drug effects , Rats , Abdominal Wall/surgery , Valproic Acid/pharmacology , Male , Abdominal Muscles/drug effects
9.
Pak J Pharm Sci ; 37(2(Special)): 423-428, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38822545

ABSTRACT

This study assessed the inhibitory effect of sodium valproate (VPA) on apoptosis of cardiomyocytes in lethally scalded rats. The model of a 50% total body surface area (TBSA) third-degree full-thickness scald was produced, 48 male SD rats were randomly divided into three groups (n = 16), the sham group and the scald group were given an intraperitoneal injection of 0.25ml of saline, the scald +VPA group was given an intraperitoneal injection of VPA (300 mg/kg) after scalded, Each group was subdivided into two subgroups (n=8) according to the two observation time points of 3h and 6h after scald. Apoptotic cardiomyocytes were observed, and myocardial tissue levels of nitric oxide (NO), cysteine protease-3 (caspase-3) activity, hypoxia-inducible factor-1α (HIF-1α), inducible nitric oxide synthase (iNOS), BCL2/adenovirus E1B interacting protein 3 (BNIP3) and caspase-3 protein were measured. Compared with sham scald group, severe scald elevated CK-MB, cardiomyocyte apoptosis rate, caspase-3 activity and protein levels, NO content, and HIF-1α signalling pathway proteins; whereas VPA decreased CK-MB, cardiomyocyte apoptosis rate and inhibited HIF-1α signalling pathway protein expression. In conclusion, these results suggested that VPA inhibited early cardiomyocyte apoptosis and attenuated myocardial injury in lethally scalded rats, which may be related to the regulation of the HIF-1α signalling pathway.


Subject(s)
Apoptosis , Burns , Hypoxia-Inducible Factor 1, alpha Subunit , Myocytes, Cardiac , Valproic Acid , Animals , Male , Rats , Apoptosis/drug effects , Burns/drug therapy , Burns/metabolism , Burns/pathology , Caspase 3/metabolism , Disease Models, Animal , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Membrane Proteins/metabolism , Mitochondrial Proteins , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Rats, Sprague-Dawley , Valproic Acid/pharmacology
10.
Molecules ; 29(11)2024 May 30.
Article in English | MEDLINE | ID: mdl-38893448

ABSTRACT

Epilepsy is a prevalent neurological disorder characterized by recurrent seizures. Validamycin A (VA) is an antibiotic fungicide that inhibits trehalase activity and is widely used for crop protection in agriculture. In this study, we identified a novel function of VA as a potential anti-seizure medication in a zebrafish epilepsy model. Electroencephalogram (EEG) analysis demonstrated that VA reduced pentylenetetrazol (PTZ)-induced seizures in the brains of larval and adult zebrafish. Moreover, VA reduced PTZ-induced irregular movement in a behavioral assessment of adult zebrafish. The developmental toxicity test showed no observable anatomical alteration when the zebrafish larvae were treated with VA up to 10 µM within the effective range. The median lethal dose of VA in adult zebrafish was > 14,000 mg/kg. These results imply that VA does not demonstrate observable toxicity in zebrafish at concentrations effective for generating anti-seizure activity in the EEG and alleviating abnormal behavior in the PTZ-induced epileptic model. Furthermore, the effectiveness of VA was comparable to that of valproic acid. These results indicate that VA may have a potentially safer anti-seizure profile than valproic acid, thus offering promising prospects for its application in agriculture and medicine.


Subject(s)
Anticonvulsants , Disease Models, Animal , Epilepsy , Pentylenetetrazole , Zebrafish , Animals , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Pentylenetetrazole/adverse effects , Epilepsy/drug therapy , Epilepsy/chemically induced , Seizures/drug therapy , Seizures/chemically induced , Electroencephalography , Valproic Acid/pharmacology , Larva/drug effects , Brain/drug effects , Brain/pathology , Inositol/analogs & derivatives
11.
Brain Res ; 1838: 148963, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38705555

ABSTRACT

BACKGROUND AND AIM: Autism spectrum disorder (ASD) is a neurodevelopmental disorder with two core behavioral symptoms restricted/repetitive behavior and social-communication deficit. The unknown etiology of ASD makes it difficult to identify potential treatments. Valproic acid (VPA) is an anticonvulsant drug with teratogenic effects during pregnancy in humans and rodents. Prenatal exposure to VPA induces autism-like behavior in both humans and rodents. This study aimed to investigate the protective effects of Diosgenin in prenatal Valproic acid-induced autism in rats. METHOD: pregnant Wister female rats were given a single intraperitoneal injection of VPA (600 mg/kg, i.p.) on gestational day 12.5. The male offspring were given oral Dios (40 mg/kg, p.o.) or Carboxymethyl cellulose (5 mg/kg, p.o.) for 30 days starting from postnatal day 23. On postnatal day 52, behavioral tests were done. Additionally, biochemical assessments for oxidative stress markers were carried out on postnatal day 60. Further, histological evaluations were performed on the prefrontal tissue by Nissl staining and Immunohistofluorescence. RESULTS: The VPA-exposed rats showed increased anxiety-like behavior in the elevated plus maze (EPM). They also demonstrated repetitive and grooming behaviors in the marble burying test (MBT) and self-grooming test. Social interaction was reduced, and they had difficulty detecting the novel object in the novel object recognition (NOR) test. Also, VPA-treated rats have shown higher levels of oxidative stress malondialdehyde (MDA) and lower GPX, TAC, and superoxide dismutase (SOD) levels. Furthermore, the number of neurons decreased and the ERK signaling pathway upregulated in the prefrontal cortex (PFC). On the other hand, treatment with Dios restored the behavioral consequences, lowered oxidative stress, and death of neurons, and rescued the overly activated ERK1/2 signaling in the prefrontal cortex. CONCLUSION: Chronic treatment with Dios restored the behavioral, biochemical, and histological abnormalities caused by prenatal VPA exposure.


Subject(s)
Autistic Disorder , Diosgenin , Disease Models, Animal , Neuroprotective Agents , Oxidative Stress , Prenatal Exposure Delayed Effects , Rats, Wistar , Valproic Acid , Animals , Valproic Acid/pharmacology , Female , Pregnancy , Neuroprotective Agents/pharmacology , Rats , Oxidative Stress/drug effects , Male , Diosgenin/pharmacology , Autistic Disorder/chemically induced , Autistic Disorder/metabolism , Autistic Disorder/drug therapy , Prenatal Exposure Delayed Effects/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Behavior, Animal/drug effects , Anticonvulsants/pharmacology , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/chemically induced , Autism Spectrum Disorder/drug therapy , Anxiety/drug therapy , Anxiety/chemically induced , Social Behavior
12.
Commun Biol ; 7(1): 642, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38802535

ABSTRACT

Alterations in the experience-dependent and autonomous elaboration of neural circuits are assumed to underlie autism spectrum disorder (ASD), though it is unclear what synaptic traits are responsible. Here, utilizing a valproic acid-induced ASD marmoset model, which shares common molecular features with idiopathic ASD, we investigate changes in the structural dynamics of tuft dendrites of upper-layer pyramidal neurons and adjacent axons in the dorsomedial prefrontal cortex through two-photon microscopy. In model marmosets, dendritic spine turnover is upregulated, and spines are generated in clusters and survived more often than in control marmosets. Presynaptic boutons in local axons, but not in commissural long-range axons, demonstrate hyperdynamic turnover in model marmosets, suggesting alterations in projection-specific plasticity. Intriguingly, nasal oxytocin administration attenuates clustered spine emergence in model marmosets. Enhanced clustered spine generation, possibly unique to certain presynaptic partners, may be associated with ASD and be a potential therapeutic target.


Subject(s)
Callithrix , Disease Models, Animal , Neuronal Plasticity , Oxytocin , Animals , Oxytocin/metabolism , Male , Synapses/metabolism , Dendritic Spines/metabolism , Dendritic Spines/pathology , Dendritic Spines/drug effects , Autism Spectrum Disorder/metabolism , Autistic Disorder/metabolism , Autistic Disorder/pathology , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Prefrontal Cortex/drug effects , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , Valproic Acid/pharmacology , Presynaptic Terminals/metabolism , Female , Axons/metabolism
13.
Behav Brain Res ; 470: 115069, 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-38797494

ABSTRACT

Recent clinical studies have highlighted mutations in the voltage-gated potassium channel Kv10.2 encoded by the KCNH5 gene among individuals with autism spectrum disorder (ASD). Our preliminary study found that Kv10.2 was decreased in the hippocampus of valproic acid (VPA) - induced ASD rats. Nevertheless, it is currently unclear how KCNH5 regulates autism-like features, or becomes a new target for autism treatment. We employed KCNH5 knockout (KCNH5-/-) rats and VPA - induced ASD rats in this study. Then, we used behavioral assessments, combined with electrophysiological recordings and hippocampal brain slice, to elucidate the impact of KCNH5 deletion and environmental factors on neural development and function in rats. We found that KCNH5-/- rats showed early developmental delay, neuronal overdevelopment, and abnormal electroencephalogram (EEG) signals, but did not exhibit autism-like behavior. KCNH5-/- rats exposed to VPA (KCNH5-/--VPA) exhibit even more severe autism-like behaviors and abnormal neuronal development. The absence of KCNH5 excessively enhances the activity of the Protein Kinase B (Akt)/Mechanistic Target of Rapamycin (mTOR) signaling pathway in the hippocampus of rats after exposure to VPA. Overall, our findings underscore the deficiency of KCNH5 increases the susceptibility to autism under environmental exposures, suggesting its potential utility as a target for screening and diagnosis in ASD.


Subject(s)
Autism Spectrum Disorder , Hippocampus , Proto-Oncogene Proteins c-akt , TOR Serine-Threonine Kinases , Animals , Male , Rats , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/genetics , Autistic Disorder/metabolism , Autistic Disorder/genetics , Disease Models, Animal , ERG1 Potassium Channel/metabolism , ERG1 Potassium Channel/genetics , Ether-A-Go-Go Potassium Channels/metabolism , Ether-A-Go-Go Potassium Channels/genetics , Hippocampus/metabolism , Hippocampus/drug effects , Neurons/metabolism , Neurons/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Signal Transduction/physiology , TOR Serine-Threonine Kinases/metabolism , Valproic Acid/pharmacology
14.
Cell Mol Biol (Noisy-le-grand) ; 70(5): 40-47, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38814236

ABSTRACT

Periodontal ligament stem cells (PDLSCs) show plasticity towards the adipogenic lineage; however, little has been done on the participation of epigenetic mechanisms. Histone acetylation is a dynamic process, though balanced by histone acetyltransferases (HATs) and histone deacetylases (HDACs) activities. This process can be halted by HDACs inhibitors, such as trichostatin A (TSA) and valproic acid (VPA). This study aimed to determine the role of HDACs class I in adipogenic differentiation of PDL cells. PDLSCs were treated with TSA at concentrations of 100, 200, and 250 nM, or VPA at 1, 4 and 8 mM. Cell viability was assessed using MTT assays. Gene expression of pluripotency markers (NANOG, OCT4, SOX2), HAT genes (p300, GCN5), and HDACs genes (HDAC1-3) was analyzed by RT-qPCR. Adipogenic differentiation was evaluated via oil red O staining, and acetylation of histone H3 lysine 9 (H3K9ac) was examined by Western blot. VPA treatment resulted in a 60% reduction in cell proliferation, compared to a 50% when using TSA. Cell viability was not affected by either inhibitor. Furthermore, both TSA and VPA induced adipogenic differentiation, through an increase in the deposition of lipid droplets and in GCN5 and p300 expression were observed. Western blot analysis showed that TSA increased H3K9ac levels on adipogenic differentiation of PDLSCs. These findings highlight the potential of HDAC inhibitors as a tool for modulating H3K9 acetylation status and thus influencing adipogenic differentiation of PDLCs.


Subject(s)
Adipogenesis , Cell Differentiation , Cell Survival , Histone Deacetylase Inhibitors , Periodontal Ligament , Valproic Acid , Humans , Periodontal Ligament/cytology , Periodontal Ligament/drug effects , Histone Deacetylase Inhibitors/pharmacology , Adipogenesis/drug effects , Adipogenesis/genetics , Valproic Acid/pharmacology , Cell Differentiation/drug effects , Cell Survival/drug effects , Acetylation/drug effects , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Hydroxamic Acids/pharmacology , Cells, Cultured , Histones/metabolism , Cell Proliferation/drug effects , Stem Cells/drug effects , Stem Cells/cytology , Stem Cells/metabolism
15.
Behav Brain Res ; 469: 115052, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38782096

ABSTRACT

Autism spectrum disorder (ASD) is a pervasive developmental disorder with gender differences. Oxytocin (OXT) is currently an important candidate drug for autism, but the lack of data on female autism is a big issue. It has been reported that the effect of OXT is likely to be different between male and female ASD patients. In the study, we specifically explored the role of the OXT signaling pathway in a VPA-induced female rat's model of autism. The data showed that there was an increase of either oxytocin or its receptor expressions in both the hippocampus and the prefrontal cortex of VPA-induced female offspring. To determine if the excess of OXT signaling contributed to autism symptoms in female rats, exogenous oxytocin and oxytocin receptor antagonists Atosiban were used in the experiment. It was found that exogenous oxytocin triggered autism-like behaviors in wild-type female rats by intranasal administration. More interestingly, several autism-like deficits including social interaction, anxiety, and repeat stereotypical sexual behavior in the VPA female offspring were significantly attenuated by oxytocin receptor antagonists Atosiban. Moreover, Atosiban also effectively improved the synaptic plasticity impairment induced by VPA in female offspring. Our results suggest that oxytocin receptor antagonists significantly improve autistic-like behaviors in a female rat model of valproic acid-induced autism.


Subject(s)
Autistic Disorder , Disease Models, Animal , Oxytocin , Receptors, Oxytocin , Valproic Acid , Vasotocin , Animals , Valproic Acid/pharmacology , Female , Receptors, Oxytocin/antagonists & inhibitors , Receptors, Oxytocin/metabolism , Oxytocin/pharmacology , Oxytocin/metabolism , Oxytocin/administration & dosage , Rats , Vasotocin/analogs & derivatives , Vasotocin/pharmacology , Autistic Disorder/chemically induced , Autistic Disorder/drug therapy , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Autism Spectrum Disorder/chemically induced , Autism Spectrum Disorder/drug therapy , Hippocampus/drug effects , Hippocampus/metabolism , Behavior, Animal/drug effects , Rats, Sprague-Dawley , Neuronal Plasticity/drug effects , Social Interaction/drug effects , Sexual Behavior, Animal/drug effects , Anxiety/drug therapy , Anxiety/chemically induced , Pregnancy
16.
Pharmacol Biochem Behav ; 240: 173788, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38734150

ABSTRACT

Autism is a complex neurodevelopmental disorder with no effective treatment available currently. Repetitive transcranial magnetic stimulation (rTMS) is emerging as a promising neuromodulation technique to treat autism. However, the mechanism how rTMS works remains unclear, which restrict the clinical application of magnetic stimulation in the autism treatment. In this study, we investigated the effect of low-frequency rTMS on the autistic-like symptoms and explored if this neuroprotective effect was associated with synaptic plasticity and neuroinflammation in the hippocampus. A rat model of autism was established by intraperitoneal injection of valproic acid (VPA) in pregnant rats and male offspring were treated with 1 Hz rTMS daily for two weeks continuously. Behavior tests were performed to identify behavioral abnormality. Synaptic plasticity was measured by in vivo electrophysiological recording and Golgi-Cox staining. Synapse and inflammation associated proteins were detected by immunofluorescence and Western blot analyses. Results showed prenatal VPA-exposed rats exhibited autistic-like and anxiety-like behaviors, and cognitive impairment. Synaptic plasticity deficits and the abnormality expression of synapse-associated proteins were found in the hippocampus of prenatal VPA-exposed rats. Prenatal VPA exposure increased the level of inflammation cytokines and promoted the excessive activation of microglia. rTMS significantly alleviated the prenatal VPA-induced abnormalities including behavioral and synaptic plasticity deficits, and excessive neuroinflammation. TMS maybe a potential strategy for autism therapy via rescuing synaptic plasticity and inhibiting neuroinflammation.


Subject(s)
Autistic Disorder , Disease Models, Animal , Hippocampus , Neuronal Plasticity , Prenatal Exposure Delayed Effects , Transcranial Magnetic Stimulation , Valproic Acid , Animals , Valproic Acid/pharmacology , Neuronal Plasticity/drug effects , Rats , Autistic Disorder/therapy , Autistic Disorder/chemically induced , Female , Male , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Hippocampus/metabolism , Hippocampus/drug effects , Rats, Sprague-Dawley , Neuroinflammatory Diseases/therapy , Behavior, Animal/drug effects
17.
Int J Mol Sci ; 25(10)2024 May 13.
Article in English | MEDLINE | ID: mdl-38791325

ABSTRACT

Perinatal exposure to valproic acid is commonly used for autism spectrum disorder (ASD) animal model development. The inhibition of histone deacetylases by VPA has been proposed to induce epigenetic changes during neurodevelopment, but the specific alterations in genetic expression underlying ASD-like behavioral changes remain unclear. We used qPCR-based gene expression and epigenetics tools and Western blotting in the hippocampi of neonatal valproic acid-exposed animals at 4 weeks of age and conducted the social interaction test to detect behavioral changes. Significant alterations in gene expression were observed in males, particularly concerning mRNA expression of Foxo3, which was significantly associated with behavioral changes. Moreover, notable differences were observed in H3K27ac chromatin immunoprecipitation, quantitative PCR (ChIP-qPCR), and methylation-sensitive restriction enzyme-based qPCR targeting the Foxo3 gene promoter region. These findings provide evidence that epigenetically regulated hippocampal Foxo3 expression may influence social interaction-related behavioral changes. Furthermore, identifying sex-specific gene expression and epigenetic changes in this model may elucidate the sex disparity observed in autism spectrum disorder prevalence.


Subject(s)
Animals, Newborn , Autism Spectrum Disorder , Epigenesis, Genetic , Forkhead Box Protein O3 , Hippocampus , Valproic Acid , Animals , Valproic Acid/pharmacology , Valproic Acid/adverse effects , Forkhead Box Protein O3/metabolism , Forkhead Box Protein O3/genetics , Hippocampus/metabolism , Hippocampus/drug effects , Epigenesis, Genetic/drug effects , Male , Female , Rats , Autism Spectrum Disorder/chemically induced , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , DNA Methylation/drug effects , Sex Characteristics , Disease Models, Animal , Pregnancy , Behavior, Animal/drug effects , Sex Factors , Rats, Sprague-Dawley , Promoter Regions, Genetic
18.
Cells ; 13(9)2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38727269

ABSTRACT

The histone deacetylase inhibitor (HDACi) valproic acid (VPA) has neuroprotective and anti-inflammatory effects in experimental traumatic brain injury (TBI), which have been partially attributed to the epigenetic disinhibition of the transcription repressor RE1-Silencing Transcription Factor/Neuron-Restrictive Silencer Factor (REST/NRSF). Additionally, VPA changes post-traumatic brain injury (TBI) brain metabolism to create a neuroprotective environment. To address the interconnection of neuroprotection, metabolism, inflammation and REST/NRSF after TBI, we subjected C57BL/6N mice to experimental TBI and intraperitoneal VPA administration or vehicle solution at 15 min, 1, 2, and 3 days post-injury (dpi). At 7 dpi, TBI-induced an up-regulation of REST/NRSF gene expression and HDACi function of VPA on histone H3 acetylation were confirmed. Neurological deficits, brain lesion size, blood-brain barrier permeability, or astrogliosis were not affected, and REST/NRSF target genes were only marginally influenced by VPA. However, VPA attenuated structural damage in the hippocampus, microgliosis and expression of the pro-inflammatory marker genes. Analyses of plasma lipidomic and polar metabolomic patterns revealed that VPA treatment increased lysophosphatidylcholines (LPCs), which were inversely associated with interleukin 1 beta (Il1b) and tumor necrosis factor (Tnf) gene expression in the brain. The results show that VPA has mild neuroprotective and anti-inflammatory effects likely originating from favorable systemic metabolic changes resulting in increased plasma LPCs that are known to be actively taken up by the brain and function as carriers for neuroprotective polyunsaturated fatty acids.


Subject(s)
Brain Injuries, Traumatic , Inflammation , Lysophosphatidylcholines , Mice, Inbred C57BL , Neurons , Valproic Acid , Animals , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/blood , Brain Injuries, Traumatic/complications , Valproic Acid/pharmacology , Valproic Acid/therapeutic use , Mice , Male , Neurons/drug effects , Neurons/pathology , Neurons/metabolism , Inflammation/pathology , Inflammation/drug therapy , Lysophosphatidylcholines/blood , Cell Death/drug effects , Disease Models, Animal , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Repressor Proteins/metabolism , Repressor Proteins/genetics
19.
Neurochem Int ; 177: 105767, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38750962

ABSTRACT

AIM AND OBJECTIVE: The purpose of the study is to determine the neuroprotective effect of capric acid on sodium valproate-induced model of autism. METHODS: In this study, the effect of CA was observed in animals with single dose of valproic acid (600 mg/kg, i. p.) where the disease condition was confirmed by developmental impairment in pups. Behavioral tests that assess anxiety, depression, stereotypical and repetitive behavior, social interaction, learning and memory, and other confounding variables were performed. Subsequently, oxidative stress parameters, pro-inflammatory cytokine levels and mitochondrial complex activities in the selected brain regions were analyzed. RESULTS: Valproic acid successfully produced autism-like symptoms from post-natal day 7 and also demonstrated impairment in social behavior, learning and memory, and anxiety and depression. Valproic acid was found to produce oxidative stress and neuro-inflammation in the hippocampus, prefrontal cortex, and cerebellum. Treatment with capric acid produced a positive effect on the alterations with maximum effects evident at 400 mg/kg, p. o. through amelioration of behavioral as well as biochemical changes. CONCLUSION: The current study concluded that capric acid could act as a likely candidate for the treatment and management of autism via significant modulation of neurobehavioral parameters, oxidative stress, mitochondrial dysfunction and inflammatory markers.


Subject(s)
Autistic Disorder , Disease Models, Animal , Neuroprotective Agents , Oxidative Stress , Valproic Acid , Valproic Acid/pharmacology , Animals , Autistic Disorder/chemically induced , Autistic Disorder/drug therapy , Autistic Disorder/metabolism , Male , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Behavior, Animal/drug effects , Decanoic Acids/pharmacology , Female , Rats , Rats, Wistar , Brain/drug effects , Brain/metabolism
20.
J Clin Psychopharmacol ; 44(4): 345-352, 2024.
Article in English | MEDLINE | ID: mdl-38639433

ABSTRACT

PURPOSE: The aim of this study was to compare the efficacy and safety profile of lurasidone combined with either lithium or valproate, in the short-term treatment of patients with bipolar depression. METHODS: Data were pooled from two 6-week, double-blind, placebo-controlled trials of patients with bipolar depression on stable doses of lithium or valproate randomized to lurasidone (20-120 mg/d) or placebo. Efficacy measures included the Montgomery-Åsberg Depression Rating Scale, Clinical Global Impressions Bipolar Scale, and the Quick Inventory of Depressive Symptomatology via self-assessment and were analyzed using a mixed model for repeated measures approach. RESULTS: Notably larger week 6 effect sizes were observed when lurasidone was added to lithium, compared with when lurasidone was added to valproate, on 2 of the 3 depression outcome measures, Montgomery-Åsberg Depression Rating Scale total score (d = 0.45 vs 0.22) and Quick Inventory of Depressive Symptomatology via self-assessment (d = 0.63 vs 0.29); the efficacy advantage was smaller on the Clinical Global Impressions Bipolar Scale depression score (d = 0.34 vs 0.29). Similar adverse event profiles were observed for lurasidone treatment in combination with either lithium or valproate. The most frequently reported events (≥5%) in both groups were nausea, parkinsonism, somnolence, akathisia, and insomnia. Minimal changes in weight, lipids, and measures of glycemic control were observed during treatment with lurasidone combined with either lithium or valproate. CONCLUSIONS: Lurasidone added to either lithium or valproate was found to be an effective treatment for bipolar depression, with a larger antidepressant effect observed when lurasidone was combined with lithium. There were no clinically meaningful differences in the safety or tolerability of lurasidone when used adjunctively with lithium or valproate.


Subject(s)
Antimanic Agents , Bipolar Disorder , Drug Therapy, Combination , Lurasidone Hydrochloride , Valproic Acid , Humans , Lurasidone Hydrochloride/administration & dosage , Lurasidone Hydrochloride/adverse effects , Lurasidone Hydrochloride/pharmacology , Lurasidone Hydrochloride/therapeutic use , Bipolar Disorder/drug therapy , Valproic Acid/administration & dosage , Valproic Acid/adverse effects , Valproic Acid/pharmacology , Valproic Acid/therapeutic use , Female , Male , Adult , Double-Blind Method , Antimanic Agents/administration & dosage , Antimanic Agents/adverse effects , Antimanic Agents/pharmacology , Middle Aged , Treatment Outcome , Antipsychotic Agents/administration & dosage , Antipsychotic Agents/adverse effects , Antipsychotic Agents/pharmacology , Lithium Compounds/administration & dosage , Lithium Compounds/adverse effects , Lithium Compounds/pharmacology , Psychiatric Status Rating Scales
SELECTION OF CITATIONS
SEARCH DETAIL
...