Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 975
Filtrer
1.
Article de Russe | MEDLINE | ID: mdl-39113443

RÉSUMÉ

Mutations in the human PCDH19 gene lead to epileptic encephalopathy of early childhood. It is characterized by the early onset of serial seizures, cognitive impairment and behavioral disorders (including autistic personality traits). In most cases, difficulties arise in selecting therapy due to pharmacoresistance. The pathogenesis of the disease is complex. The data available to us at the moment from numerous studies present the pathogenesis of «PCDH19 syndrome¼ as multi-level, affecting both the epigenetic support of cell life, and development of stem cells and progenitor cells in the process of neuroontogenesis, and the influence on the neurotransmitter mechanisms of the brain, and disruption of the formation of neural networks with an inevitable increase in the excitability of the cerebral cortex as a whole, and local changes in the highly labile regulatory structures of the hippocampal region. And it is not surprising that all these changes entail not only (and perhaps not so much) epileptization, but a profound disruption of the regulation of brain activity, accompanied by autism spectrum disorders, more profound disorders in the form of schizophrenia or cyclothymia, and the formation of delayed psychomotor development. A «side branch¼ of these pathogenetic processes can also be considered the participation of PCDH19 dysfunctions in certain variants of oncogenesis. The need for polypharmacy (in most cases) confirms the diversity of mechanisms involved in the pathogenesis of the disease and makes the prospects for the development of effective and rational treatment regimens very vague. Cautious optimism is caused only by attempts at relatively specific treatment with ganaxolone.


Sujet(s)
Épilepsie , Polypharmacie , Humains , Trouble du spectre autistique/génétique , Trouble du spectre autistique/traitement médicamenteux , Encéphale , Cadhérines/génétique , Épilepsie/traitement médicamenteux , Épilepsie/génétique , Mutation , Protocadhérines
2.
Theranostics ; 14(11): 4375-4392, 2024.
Article de Anglais | MEDLINE | ID: mdl-39113803

RÉSUMÉ

Rationale: Autism spectrum disorder (ASD) represents a complex neurodevelopmental condition lacking specific pharmacological interventions. Given the multifaced etiology of ASD, there exist no effective treatment for ASD. Rapamycin (RAPA) can activate autophagy by inhibiting the mTOR pathway and has exhibited promising effects in treating central nervous system disorders; however, its limited ability to cross the blood-brain barrier (BBB) has hindered its clinical efficacy, leading to substantial side effects. Methods: To address this challenge, we designed a drug delivery system utilizing red blood cell membrane (CM) vesicles modified with SS31 peptides to enhance the brain penetration of RAPA for the treatment of autism. Results: The fabricated SCM@RAPA nanoparticles, with an average diameter of 110 nm, exhibit rapid release of RAPA in a pathological environment characterized by oxidative stress. In vitro results demonstrate that SCM@RAPA effectively activate cellular autophagy, reduce intracellular ROS levels, improve mitochondrial function, thereby ameliorating neuronal damage. SS31 peptide modification significantly enhances the BBB penetration and rapid brain accumulation of SCM@RAPA. Notably, SCM@RAPA nanoparticles demonstrate the potential to ameliorate social deficits, improve cognitive function, and reverse neuronal impairments in valproic acid (VPA)-induced ASD models. Conclusions: The therapeutic potential of SCM@RAPA in managing ASD signifies a paradigm shift in autism drug treatment, holding promise for clinical interventions in diverse neurological conditions.


Sujet(s)
Trouble du spectre autistique , Autophagie , Barrière hémato-encéphalique , Nanoparticules , Stress oxydatif , Sirolimus , Sirolimus/administration et posologie , Sirolimus/pharmacologie , Stress oxydatif/effets des médicaments et des substances chimiques , Trouble du spectre autistique/traitement médicamenteux , Trouble du spectre autistique/métabolisme , Animaux , Autophagie/effets des médicaments et des substances chimiques , Nanoparticules/composition chimique , Barrière hémato-encéphalique/métabolisme , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Souris , Humains , Systèmes de délivrance de médicaments/méthodes , Modèles animaux de maladie humaine , Mâle , Matériaux biomimétiques/administration et posologie , Matériaux biomimétiques/composition chimique , Matériaux biomimétiques/pharmacologie , Biomimétique/méthodes , Encéphale/métabolisme , Encéphale/effets des médicaments et des substances chimiques , Peptides/administration et posologie , Espèces réactives de l'oxygène/métabolisme , Acide valproïque/administration et posologie , Acide valproïque/pharmacologie
3.
J Clin Psychopharmacol ; 44(5): 462-467, 2024.
Article de Anglais | MEDLINE | ID: mdl-39174017

RÉSUMÉ

BACKGROUND: Despite the use of behavioral interventions and psychotropic medications, many individuals with autism spectrum disorder (ASD) who engage in severe aggression remain refractory to conventional treatment. Propranolol, a beta-blocker, has accumulated much anecdotal evidence as a promising option. However, well-designed studies are rare, and the apprehension about cardiovascular side effects from large doses continues to exist. PURPOSE: The aims of this study were (1) to demonstrate the feasibility of treating aggression with high-dose propranolol using telehealth study visits and (2) to document cardiac safety. METHODS: This study utilized a randomized, double-blind, placebo-controlled, crossover design. Dosing was titrated up in a flexible but stepwise fashion until therapeutic response was obtained or up to 200 mg tid. Following washout, those who were assigned propranolol were crossed over to placebo and vice versa. Six participants between the ages 12-19 participated. The primary outcome measures were the final Clinical Global Impression Improvement Scale (CGI-I) and the Aberrant Behavior Checklist-Community Irritability (ABC-C/I) scores at 200 mg tid. RESULTS: The CGI-I indicated a 50% reduction in symptoms in the propranolol phase, while the ABC-I indicated a 37% reduction in comparison to placebo. The effect sizes ( r ) for the CGI-I and the ABC-C/I were large, -0.74 and -0.64, respectively. The average blood pressure was 122/68 during the placebo phase and 109/72 during the propranolol phase. All Holter monitor exams were unremarkable. CONCLUSION: These results suggest that propranolol is an effective option in decreasing aggression in individuals with ASD. As this was a small study, a larger clinical trial is needed.


Sujet(s)
Antagonistes bêta-adrénergiques , Agressivité , Trouble du spectre autistique , Études croisées , Propranolol , Humains , Trouble du spectre autistique/traitement médicamenteux , Méthode en double aveugle , Propranolol/administration et posologie , Propranolol/effets indésirables , Agressivité/effets des médicaments et des substances chimiques , Mâle , Adolescent , Enfant , Antagonistes bêta-adrénergiques/administration et posologie , Antagonistes bêta-adrénergiques/effets indésirables , Antagonistes bêta-adrénergiques/pharmacologie , Projets pilotes , Jeune adulte , Femelle , Résultat thérapeutique , Adulte
4.
Int J Biochem Cell Biol ; 174: 106634, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39094731

RÉSUMÉ

Postmortem studies have revealed that brains of individuals with autism spectrum disorder (ASD) exhibit abnormalities in various components of the cholinergic system including cholinergic receptors, projections, and nuclei. Deletions in the 15q13.3 region which encompasses CHRNA7, the gene that encodes the α7-nACh receptor, have been linked to various neurodevelopmental disorders, including ASD. In addition, the involvement of α7-nACh receptors in biological phenomena known to play a role in the pathophysiology of ASD such as cognitive functions, learning, memory, neuroinflammation, and oxidative stress, as well as the excitation-inhibition balance in neuronal circuits and maternal immune activation have been reported in previous studies. Furthermore, evolving preclinical and clinical literature supports the potential therapeutic benefits of using selectively acting cholinergic compounds, particularly those targeting the α7-nACh receptor subtype, in the treatment of ASD. This study reviews the previous literature on the involvement of nACh receptors in the pathophysiology of ASD and focuses on the α7-nACh receptor as a potential therapeutic target.


Sujet(s)
Trouble du spectre autistique , Récepteur nicotinique de l'acétylcholine alpha7 , Animaux , Humains , Récepteur nicotinique de l'acétylcholine alpha7/métabolisme , Récepteur nicotinique de l'acétylcholine alpha7/génétique , Trouble du spectre autistique/traitement médicamenteux , Trouble du spectre autistique/métabolisme , Trouble du spectre autistique/génétique , Trouble du spectre autistique/physiopathologie , Trouble du spectre autistique/anatomopathologie
5.
J Korean Med Sci ; 39(26): e201, 2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38978488

RÉSUMÉ

BACKGROUND: Oral retinoids are used to treat various dermatological conditions, and their use is increasing in women of childbearing age. However, there is limited knowledge on the incidence of adverse outcomes after retinoid exposure during pregnancy. We aimed to evaluate the risk of adverse outcomes associated with oral retinoid exposure during pregnancy. METHODS: We conducted a retrospective cohort study using the NHIS mother-child linked healthcare database in South Korea. We included all women who gave live birth from April 1, 2009 to December 31, 2020 and their children. The exposure was defined as having ≥ 1 prescription of isotretinoin, alitretinoin, and acitretin from one month before pregnancy to the delivery. The outcomes of interest were adverse child outcomes including major congenital malformations, low birth weight, and neurodevelopmental disorders (autism spectrum disorder and intellectual disorder), and adverse pregnancy outcomes including gestational diabetes mellitus, preeclampsia, and postpartum hemorrhage. Propensity score-based matching weights were used to control for various potential confounders. For congenital malformation, low birth weight, and adverse pregnancy outcomes, we calculated relative risk (RR) with 95% confidence interval (CI) using a generalized linear model and for neurodevelopmental disorders, we estimated hazard ratio (HR) with 95% CI using the Cox proportional hazard model. RESULTS: Of 3,894,184 pregnancies, we identified 720 pregnancies (0.02%) as the oral retinoid-exposed group. The incidence of major congenital malformation was 400.6 per 10,000 births for oral retinoid-exposed group and 357.9 per 10,000 births for unexposed group and the weighted RR was 1.10 (95% CI, 0.65-1.85) in oral retinoid-exposed group compared with unexposed group. The neurodevelopmental disorder showed a potential increased risk, with the weighted HR of 1.63 (95% CI, 0.60-4.41) for autism spectrum disorder and 1.71 (95% CI, 0.60-4.93) for the intellectual disorder, although it did not reach statistical significance. For low birth weight and adverse pregnancy outcomes, no association was observed with oral retinoid exposure during pregnancy. CONCLUSION: This study found no significantly increased risk of congenital malformations, autism spectrum disorders, and intellectual disability associated with oral retinoid exposure during pregnancy; however, given the limitations such as including only the live births and increased point estimate, potential risk cannot be fully excluded.


Sujet(s)
Issue de la grossesse , Rétinoïdes , Humains , Femelle , Grossesse , Études rétrospectives , Adulte , République de Corée/épidémiologie , Rétinoïdes/effets indésirables , Rétinoïdes/usage thérapeutique , Administration par voie orale , Nouveau-né , Nourrisson à faible poids de naissance , Isotrétinoïne/effets indésirables , Isotrétinoïne/usage thérapeutique , Complications de la grossesse/traitement médicamenteux , Acitrétine/effets indésirables , Acitrétine/usage thérapeutique , Bases de données factuelles , Modèles des risques proportionnels , Jeune adulte , Trouble du spectre autistique/épidémiologie , Trouble du spectre autistique/traitement médicamenteux
6.
Brief Bioinform ; 25(4)2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-39038939

RÉSUMÉ

Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder for which current treatments are limited and drug development costs are prohibitive. Identifying drug targets for ASD is crucial for the development of targeted therapies. Summary-level data of expression quantitative trait loci obtained from GTEx, protein quantitative trait loci data from the ROSMAP project, and two ASD genome-wide association studies datasets were utilized for discovery and replication. We conducted a combined analysis using Mendelian randomization (MR), transcriptome-wide association studies, Bayesian colocalization, and summary-data-based MR to identify potential therapeutic targets associated with ASD and examine whether there are shared causal variants among them. Furthermore, pathway and drug enrichment analyses were performed to further explore the underlying mechanisms and summarize the current status of pharmacological targets for developing drugs to treat ASD. The protein-protein interaction (PPI) network and mouse knockout models were performed to estimate the effect of therapeutic targets. A total of 17 genes revealed causal associations with ASD and were identified as potential targets for ASD patients. Cathepsin B (CTSB) [odd ratio (OR) = 2.66 95, confidence interval (CI): 1.28-5.52, P = 8.84 × 10-3], gamma-aminobutyric acid type B receptor subunit 1 (GABBR1) (OR = 1.99, 95CI: 1.06-3.75, P = 3.24 × 10-2), and formin like 1 (FMNL1) (OR = 0.15, 95CI: 0.04-0.58, P = 5.59 × 10-3) were replicated in the proteome-wide MR analyses. In Drugbank, two potential therapeutic drugs, Acamprosate (GABBR1 inhibitor) and Bryostatin 1 (CASP8 inhibitor), were inferred as potential influencers of autism. Knockout mouse models suggested the involvement of the CASP8, GABBR1, and PLEKHM1 genes in neurological processes. Our findings suggest 17 candidate therapeutic targets for ASD and provide novel drug targets for therapy development and critical drug repurposing opportunities.


Sujet(s)
Trouble du spectre autistique , Étude d'association pangénomique , Protéomique , Humains , Trouble du spectre autistique/traitement médicamenteux , Trouble du spectre autistique/génétique , Trouble du spectre autistique/métabolisme , Animaux , Souris , Transcriptome , Locus de caractère quantitatif , Cartes d'interactions protéiques/effets des médicaments et des substances chimiques , Souris knockout , Thérapie moléculaire ciblée
7.
Int Rev Neurobiol ; 177: 149-203, 2024.
Article de Anglais | MEDLINE | ID: mdl-39029984

RÉSUMÉ

Autism Spectrum Disorder (ASD) is a neurodevelopmental condition characterized by persistent deficits in social communication and interaction, as well as restricted and repetitive patterns of behavior. Despite extensive research, effective pharmacological interventions for ASD remain limited. Cannabidiol (CBD), a non-psychotomimetic compound of the Cannabis sativa plant, has potential therapeutic effects on several neurological and psychiatric disorders. CBD interacts with the endocannabinoid system, a complex cell-signaling system that plays a crucial role in regulating various physiological processes, maintaining homeostasis, participating in social and behavioral processing, and neuronal development and maturation with great relevance to ASD. Furthermore, preliminary findings from clinical trials indicate that CBD may have a modulatory effect on specific ASD symptoms and comorbidities in humans. Interestingly, emerging evidence suggests that CBD may influence the gut microbiota, with implications for the bidirectional communication between the gut and the central nervous system. CBD is a safe drug with low induction of side effects. As it has a multi-target pharmacological profile, it becomes a candidate compound for treating the central symptoms and comorbidities of ASD.


Sujet(s)
Trouble du spectre autistique , Cannabidiol , Humains , Trouble du spectre autistique/traitement médicamenteux , Cannabidiol/usage thérapeutique , Cannabidiol/pharmacologie , Animaux , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques
8.
Article de Anglais | MEDLINE | ID: mdl-39004333

RÉSUMÉ

OBJECTIVES: This systematic review sought to provide evidence for the effectiveness of common pharmacological interventions used for treating attention deficit hyperactivity disorder (ADHD) symptoms in the autism spectrum disorder (ASD) population, considering studies attempting to find safe and effective drugs. METHODS: We searched for randomized controlled trials describing the effectiveness and/or safety profile of pharmacological interventions for treating ASD and ADHD or ASD with ADHD symptoms using three bibliographic databases: PubMed, Cochrane Library, and Embase. We have chosen ADHD symptoms measured by any clinical scale as the primary outcome. As additional outcomes, we have used other symptoms of aberrant behavior measured by the aberrant behavior checklist, satisfaction with treatment, and peer satisfaction. RESULTS: Twenty-two publications met the inclusion criteria for the systematic review and eight for the meta-analysis. In our investigation, we found a few articles using clonidine, modafinil, and bupropion as interventions when compared to methylphenidate (MPH). Our meta-analysis showed that MPH had positive changes compared to placebo in symptoms such as hyperactivity, irritability, or inattention. However, no effect was found in stereotyped symptoms, and our data's quantitative analysis revealed a large effect of MPH-induced adverse effects on the dropout rate. On the other hand, atomoxetine initiation had positive effects when compared to placebo on symptoms of hyperactivity and inattention. We have found no effect of atomoxetine on stereotypes or irritability. Furthermore, atomoxetine did not influence side effects that caused dropouts from studies. CONCLUSION: Our results indicated that atomoxetine has a modest effect on hyperactivity and inattention symptoms, with a relatively benign profile of side effects. MPH appears to be effective in handling hyperactivity, inattention, and irritability symptoms. However, our results on atomoxetine revealed increased dropouts due to adverse effects when compared to MPH or placebo. Evidence for other substances such as guanfacine, clonidine, bupropion, or modafinil is either preliminary or nonexistent.


Sujet(s)
Trouble déficitaire de l'attention avec hyperactivité , Trouble du spectre autistique , Humains , Trouble déficitaire de l'attention avec hyperactivité/traitement médicamenteux , Trouble du spectre autistique/traitement médicamenteux , Stimulants du système nerveux central/usage thérapeutique , Essais contrôlés randomisés comme sujet , Clonidine/usage thérapeutique , Méthylphénidate/usage thérapeutique , Résultat thérapeutique
9.
Transl Psychiatry ; 14(1): 307, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39054328

RÉSUMÉ

Activity-dependent neuroprotective protein (ADNP) syndrome is a rare neurodevelopmental disorder resulting in intellectual disability, developmental delay and autism spectrum disorder (ASD) and is due to mutations in the ADNP gene. Ketamine treatment has emerged as a promising therapeutic option for ADNP syndrome, showing safety and apparent behavioral improvements in a first open label study. However, the molecular perturbations induced by ketamine remain poorly understood. Here, we investigated the longitudinal effect of ketamine on the blood transcriptome of 10 individuals with ADNP syndrome. Transcriptomic profiling was performed before and at multiple time points after a single low-dose intravenous ketamine infusion (0.5 mg/kg). We show that ketamine triggers immediate and profound gene expression alterations, with specific enrichment of monocyte-related expression patterns. These acute alterations encompass diverse signaling pathways and co-expression networks, implicating upregulation of immune and inflammatory-related processes and down-regulation of RNA processing mechanisms and metabolism. Notably, these changes exhibit a transient nature, returning to baseline levels 24 hours to 1 week after treatment. These findings enhance our understanding of ketamine's molecular effects and lay the groundwork for further research elucidating its specific cellular and molecular targets. Moreover, they contribute to the development of therapeutic strategies for ADNP syndrome and potentially, ASD more broadly.


Sujet(s)
Trouble du spectre autistique , Kétamine , Transcriptome , Kétamine/pharmacologie , Kétamine/administration et posologie , Kétamine/usage thérapeutique , Humains , Mâle , Transcriptome/effets des médicaments et des substances chimiques , Enfant , Femelle , Trouble du spectre autistique/traitement médicamenteux , Trouble du spectre autistique/génétique , Trouble du spectre autistique/sang , Enfant d'âge préscolaire , Protéines de tissu nerveux/génétique , Déficience intellectuelle/traitement médicamenteux , Déficience intellectuelle/génétique , Incapacités de développement/génétique , Incapacités de développement/traitement médicamenteux , Analyse de profil d'expression de gènes , Adolescent , Protéines à homéodomaine
10.
Transl Psychiatry ; 14(1): 312, 2024 Jul 29.
Article de Anglais | MEDLINE | ID: mdl-39075076

RÉSUMÉ

Over the last decade, a number of clinical trials have reported effects of chronic treatment with intranasal oxytocin on autistic symptoms but with inconsistent findings. Autism is a heterogeneous disorder and one factor which may influence treatment outcome is whether a subtype of individuals is more sensitive to oxytocin. In a recent cross-over trial on 41 young autistic children we reported that 44% showed a reliable improvement in clinical symptoms (Autism Diagnostic Observation Schedule, ADOS-2) after a placebo-controlled, 6-week intranasal oxytocin intervention where treatment was given every other day followed by a period of positive social interaction. In the current re-assessment of the data, we used an unsupervised data-driven cluster analysis approach to identify autism subtypes using 23 different demographic, social subtype, endocrine, eye-tracking and clinical symptom measures taken before treatment and this revealed an optimum of two different subtypes. We then assessed the proportion of identified responders to oxytocin and found that while 61.5% of one subtype included responders only 13.3% of the other did so. During the placebo phase there was no difference between the two subtypes for the small proportion of responders (19.2% vs 6.7%). This oxytocin-sensitive subtype also showed overall significant post-treatment clinical and eye-tracking measure changes. The oxytocin-sensitive subtype was primarily characterized at baseline by lower initial clinical severity (ADOS-2) and greater interest in the eye-region of emotional faces. These features alone were nearly as efficient in identifying the two subtypes as all 23 baseline measures and this easy-to-conduct approach may help rapidly and objectively screen for oxytocin responders. Future clinical trials using oxytocin interventions may therefore achieve greater success by focusing on children with this specific autism subtype and help develop individualized oxytocin intervention.


Sujet(s)
Administration par voie nasale , Trouble du spectre autistique , Ocytocine , Humains , Ocytocine/administration et posologie , Ocytocine/usage thérapeutique , Trouble du spectre autistique/traitement médicamenteux , Mâle , Femelle , Enfant , Analyse de regroupements , Résultat thérapeutique , Études croisées , Enfant d'âge préscolaire
11.
Int J Dev Neurosci ; 84(5): 454-468, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38961588

RÉSUMÉ

In this study, we delved into the intricate world of autism spectrum disorder (ASD) and its connection to the disturbance in the Wnt signaling pathway and immunological abnormalities. Our aim was to evaluate the impact of silibinin, a remarkable modulator of both the Wnt signaling pathway and the immune system, on the neurobehavioral and molecular patterns observed in a zebrafish model of ASD induced by valproic acid (VPA). Because silibinin is a hydrophobic molecule and highly insoluble in water, it was used in the form of silibinin nanoparticles (nanosilibinin, NS). After assessing survival, hatching rate, and morphology of zebrafish larvae exposed to different concentrations of NS, the appropriate concentrations were chosen. Then, zebrafish embryos were exposed to VPA (1 µM) and NS (100 and 200 µM) at the same time for 120 h. Next, anxiety and inattentive behaviors and the expression of CHD8, CTNNB, GSK3beta, LRP6, TNFalpha, IL1beta, and BDNF genes were assessed 7 days post fertilization. The results indicated that higher concentrations of NS had adverse effects on survival, hatching, and morphological development. The concentrations of 100 and 200 µM of NS could ameliorate the anxiety-like behavior and learning deficit and decrease ASD-related cytokines (IL1beta and TNFalpha) in VPA-treated larvae. In addition, only 100 µM of NS prevented raising the gene expression of Wnt signaling-related factors (CHD8, CTNNB, GSK3beta, and LRP6). In conclusion, NS treatment for the first 120 h showed therapeutic effect on an autism-like phenotype probably via reducing the expression of pro-inflammatory cytokines genes and changing the expression of Wnt signaling components genes.


Sujet(s)
Trouble du spectre autistique , Cytokines , Modèles animaux de maladie humaine , Acide valproïque , Voie de signalisation Wnt , Danio zébré , Animaux , Acide valproïque/pharmacologie , Voie de signalisation Wnt/effets des médicaments et des substances chimiques , Trouble du spectre autistique/traitement médicamenteux , Cytokines/métabolisme , Embryon non mammalien/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments
12.
Neuroscience ; 554: 16-25, 2024 Aug 30.
Article de Anglais | MEDLINE | ID: mdl-39004410

RÉSUMÉ

The biological effects of dapagliflozin, a sodium-glucose cotransporter-2 (SGLT2) inhibitor, reveal its antioxidant and anti-inflammatory properties, suggesting therapeutic benefits beyond glycemic control. This study explores the neuroprotective effects of dapagliflozin in a rat model of autism spectrum disorder (ASD) induced by propionic acid (PPA), characterized by social interaction deficits, communication challenges, repetitive behaviors, cognitive impairments, and oxidative stress. Our research aims to find effective treatments for ASD, a condition with limited therapeutic options and significant impacts on individuals and families. PPA induces ASD-like symptoms in rodents, mimicking biochemical and behavioral features of human ASD. This study explores dapagliflozin's potential to mitigate these symptoms, providing insights into novel therapeutic avenues. The findings demonstrate that dapagliflozin enhances the activation of the nuclear factor erythroid 2-related factor 2 (Nrf2) antioxidant pathway and increases levels of neurotrophic and growth factors such as brain-derived neurotrophic factor (BDNF), insulin-like growth factor-1 (IGF-1), and insulin-like growth factor-binding protein-3 (IGFBP-3). Additionally, dapagliflozin reduces pro-inflammatory cytokines, including tumor necrosis factor-alpha (TNF-α) and interleukin-17 (IL-17), and decreases the oxidative stress marker malondialdehyde (MDA). Dapagliflozin's antioxidant properties support cognitive functions by modulating apoptotic mechanisms and enhancing antioxidant capacity. These combined effects contribute to reducing learning and memory impairments in PPA-induced ASD, highlighting dapagliflozin's potential as an adjunctive therapy for oxidative stress and inflammation-related cognitive decline in ASD. This study underscores the importance of exploring new therapeutic strategies targeting molecular pathways involved in the pathophysiology of ASD, potentially improving the quality of life for individuals affected by this disorder.


Sujet(s)
Antioxydants , Trouble du spectre autistique , Composés benzhydryliques , Glucosides , Facteur de croissance IGF-I , Facteur-2 apparenté à NF-E2 , Propionates , Animaux , Glucosides/pharmacologie , Trouble du spectre autistique/traitement médicamenteux , Trouble du spectre autistique/métabolisme , Trouble du spectre autistique/induit chimiquement , Facteur de croissance IGF-I/métabolisme , Facteur-2 apparenté à NF-E2/métabolisme , Mâle , Composés benzhydryliques/pharmacologie , Propionates/pharmacologie , Antioxydants/pharmacologie , Rats , Stress oxydatif/effets des médicaments et des substances chimiques , Neuroprotecteurs/pharmacologie , Modèles animaux de maladie humaine , Rat Sprague-Dawley , Inhibiteurs du cotransporteur sodium-glucose de type 2/pharmacologie
13.
Neuropharmacology ; 258: 110089, 2024 Nov 01.
Article de Anglais | MEDLINE | ID: mdl-39033904

RÉSUMÉ

Autism spectrum disorder (ASD) is neurodevelopmental disorder with a high incidence rate, characterized by social deficits and repetitive behaviors. There is currently no effective management available to treat the core symptoms of ASD; however, oxidative stress has been implicated in its pathogenesis. Edaravone (EDA), a free-radical scavenger, is used to treat amyotrophic lateral sclerosis (ALS) and acute ischemic stroke (AIS). Here, we hypothesized that an oral formula of EDA may have therapeutic efficacy in the treatment of core ASD symptoms. A rat model of autism was established by prenatal exposure to valproic acid (VPA), and the offsprings were orally treated with EDA at low (3 mg/kg), medium (10 mg/kg), and high (30 mg/kg) doses once daily for 28 days starting from postnatal day 25 (PND25). Oral EDA administration alleviated the core symptoms in VPA rats in a dose-dependent manner, including repetitive stereotypical behaviors and impaired social interaction. Furthermore, oral administration of EDA significantly reduced oxidative stress in a dose-dependent manner, as evidenced by a reduction in oxidative stress markers and an increase in antioxidants in the blood and brain. In addition, oral EDA significantly attenuated downstream pathologies, including synaptic and mitochondrial damage in the brain. Proteomic analysis further revealed that EDA corrected the imbalance in brain oxidative reduction and mitochondrial proteins induced by prenatal VPA administration. Overall, these findings demonstrate that oral EDA has therapeutic potential for ASD by targeting the oxidative stress pathway of disease pathogenesis and paves the way towards clinical studies.


Sujet(s)
Trouble du spectre autistique , Modèles animaux de maladie humaine , Édaravone , Stress oxydatif , Acide valproïque , Animaux , Acide valproïque/pharmacologie , Acide valproïque/administration et posologie , Édaravone/pharmacologie , Trouble du spectre autistique/traitement médicamenteux , Trouble du spectre autistique/induit chimiquement , Femelle , Stress oxydatif/effets des médicaments et des substances chimiques , Mâle , Administration par voie orale , Grossesse , Rats , Rat Sprague-Dawley , Encéphale/effets des médicaments et des substances chimiques , Encéphale/métabolisme , Encéphale/anatomopathologie , Effets différés de l'exposition prénatale à des facteurs de risque/induit chimiquement , Piégeurs de radicaux libres/pharmacologie , Piégeurs de radicaux libres/administration et posologie , Piégeurs de radicaux libres/usage thérapeutique , Relation dose-effet des médicaments , Comportement stéréotypé/effets des médicaments et des substances chimiques , Comportement animal/effets des médicaments et des substances chimiques , Interaction sociale/effets des médicaments et des substances chimiques
14.
Neuropharmacology ; 257: 110057, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-38964596

RÉSUMÉ

Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by alterations and imbalances in multiple brain neurochemical systems, particularly the serotonergic neurotransmission. This includes changes in serotonin (5-HT) levels, aberrations in 5-HT transporter activity, and decreased synthesis and expression of 5-HT receptors (5-HT7Rs). The exact role of the brain 5-HT system in the development of ASD remains unclear, with conflicting evidence on its involvement. Recently, we have reported research has shown a significant decrease in serotonergic neurons originating from the raphe nuclei and projecting to the CA1 region of the dorsal hippocampus in autistic-like rats. Additionally, we have shown that chronic activation of 5-HT7Rs reverses the effects of autism induction on synaptic plasticity. However, the functional significance of 5-HT7Rs at the cellular level is still not fully understood. This study presents new evidence indicating an upregulation of 5-HT7R in the CA1 subregion of the hippocampus following the induction of autism. The present account also demonstrates that activation of 5-HT7R with its agonist LP-211 can reverse electrophysiological abnormalities in hippocampal pyramidal neurons in a rat model of autism induced by prenatal exposure to VPA. Additionally, in vivo administration of LP-211 resulted in improvements in motor coordination, novel object recognition, and a reduction in stereotypic behaviors in autistic-like offspring. The findings suggest that dysregulated expression of 5-HT7Rs may play a role in the pathophysiology of ASD, and that agonists like LP-211 could potentially be explored as a pharmacological treatment for autism spectrum disorder.


Sujet(s)
Modèles animaux de maladie humaine , Effets différés de l'exposition prénatale à des facteurs de risque , Récepteurs sérotoninergiques , Régulation positive , Acide valproïque , Animaux , Récepteurs sérotoninergiques/métabolisme , Acide valproïque/pharmacologie , Grossesse , Effets différés de l'exposition prénatale à des facteurs de risque/métabolisme , Effets différés de l'exposition prénatale à des facteurs de risque/induit chimiquement , Femelle , Régulation positive/effets des médicaments et des substances chimiques , Mâle , Région CA1 de l'hippocampe/métabolisme , Région CA1 de l'hippocampe/effets des médicaments et des substances chimiques , Rats , Pipérazines/pharmacologie , Trouble du spectre autistique/métabolisme , Trouble du spectre autistique/traitement médicamenteux , Rat Wistar , Trouble autistique/métabolisme , Trouble autistique/traitement médicamenteux
15.
J Appl Res Intellect Disabil ; 37(4): e13265, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38859732

RÉSUMÉ

BACKGROUND: In people with intellectual disabilities and/or autism spectrum disorder, oral midazolam (OM) is very effective as premedication for facilitating medical treatment. In this retrospective study, we investigated the optimal dosage of OM for premedication. METHODS: Patients with intellectual disability and/or autism spectrum disorder who were given OM as a premedication were selected from anaesthesia records. The primary outcome variable was the dose of OM (mg/kg) required to produce an adequate sedation. RESULTS: The mean OM dose required was 0.32 ± 0.10 mg/kg. The required OM dose decreased significantly as age and weight increased, and age and weight were also shown to be significantly associated with the dose of OM in the multivariate linear regression analysis. CONCLUSION: The dosage of OM to achieve adequate sedation should decrease as the patient ages. Furthermore, adequate sedation can be achieved with even lower doses of OM in obese people.


Sujet(s)
Trouble du spectre autistique , Hypnotiques et sédatifs , Déficience intellectuelle , Midazolam , Humains , Trouble du spectre autistique/traitement médicamenteux , Midazolam/administration et posologie , Mâle , Femelle , Adulte , Jeune adulte , Études rétrospectives , Hypnotiques et sédatifs/administration et posologie , Adolescent , Enfant , Adulte d'âge moyen , Administration par voie orale , Relation dose-effet des médicaments , Prémédication
16.
Nutrients ; 16(11)2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38892585

RÉSUMÉ

Autism spectrum disorder (ASD) is a neurodevelopmental disorder identified by impairments in common social interactions and repetitive behaviors. In ASD patients, substantial morphological alterations have been observed in the hippocampus, which represents an important region for the development of social skills. Melatonin, commonly found in many foods and plants, is also produced by the pineal gland. This indolamine, known to regulate the circadian rhythm, shows antioxidant and anti-inflammatory properties. We therefore hypothesized that melatonin may reduce oxidative stress and inflammation in the hippocampus of ASD patients. We explored our hypothesis using the BTBR mouse, a well-regarded murine transgenic model for ASD. Immediately after weaning, male BTBR and C57BL/6 mice underwent an 8-week treatment with melatonin or vehicle. Later, through immunohistochemistry and the immunoblotting analysis of the hippocampus, we evaluated the overall expression and cellular localization of Nrf2 and SOD1, two enzymes involved in the oxidative stress response. Similarly, we evaluated NLRP3 and NFkB, two mediators of inflammation, and GAD67, an enzyme responsible for the synthesis of GABA. Ultimately, we addressed melatonin's potential to regulate iron metabolism through a DAB-enhanced Perls reaction assay. Results showed melatonin's potential for modulating the analyzed markers in BTBR mice, suggesting a potential neuroprotective effect in ASD patients.


Sujet(s)
Trouble du spectre autistique , Modèles animaux de maladie humaine , Hippocampe , Mélatonine , Souris de lignée C57BL , Neuroprotecteurs , Stress oxydatif , Animaux , Mélatonine/pharmacologie , Hippocampe/métabolisme , Hippocampe/effets des médicaments et des substances chimiques , Mâle , Neuroprotecteurs/pharmacologie , Souris , Stress oxydatif/effets des médicaments et des substances chimiques , Trouble du spectre autistique/traitement médicamenteux , Trouble du spectre autistique/métabolisme , Antioxydants/pharmacologie , Souris transgéniques , Facteur-2 apparenté à NF-E2/métabolisme , Inflammation/métabolisme , Inflammation/prévention et contrôle
17.
Int J Nanomedicine ; 19: 6035-6055, 2024.
Article de Anglais | MEDLINE | ID: mdl-38911505

RÉSUMÉ

Background: Autism Spectrum Disorder (ASD) is a neurodevelopmental condition that affects social interaction and communication and can cause stereotypic behavior. Fullerenols, a type of carbon nanomaterial known for its neuroprotective properties, have not yet been studied for their potential in treating ASD. We aimed to investigate its role in improving autistic behaviors in BTBR T+Itpr3tf/J (BTBR) mice and its underlying mechanism, which could provide reliable clues for future ASD treatments. Methods: Our research involved treating C57BL/6J (C57) and BTBR mice with either 0.9% NaCl or fullerenols (10 mg/kg) daily for one week at seven weeks of age. We then conducted ASD-related behavioral tests in the eighth week and used RNA-seq to screen for vital pathways in the mouse hippocampus. Additionally, we used real-time quantitative PCR (RT-qPCR) to verify related pathway genes and evaluated the number of stem cells in the hippocampal dentate gyrus (DG) by Immunofluorescence staining. Results: Our findings revealed that fullerenols treatment significantly improved the related ASD-like behaviors of BTBR mice, manifested by enhanced social ability and improved cognitive deficits. Immunofluorescence results showed that fullerenols treatment increased the number of DCX+ and SOX2+/GFAP+ cells in the DG region of BTBR mice, indicating an expanded neural progenitor cell (NPC) pool of BTBR mice. RNA-seq analysis of the mouse hippocampus showed that VEGFA was involved in the rescued hippocampal neurogenesis by fullerenols treatment. Conclusion: In conclusion, our findings suggest that fullerenols treatment improves ASD-like behavior in BTBR mice by upregulating VEGFA, making nanoparticle- fullerenols a promising drug for ASD treatment.


Sujet(s)
Trouble du spectre autistique , Dysfonctionnement cognitif , Modèles animaux de maladie humaine , Protéine doublecortine , Fullerènes , Souris de lignée C57BL , Animaux , Souris , Fullerènes/pharmacologie , Fullerènes/composition chimique , Trouble du spectre autistique/traitement médicamenteux , Dysfonctionnement cognitif/traitement médicamenteux , Mâle , Comportement social , Comportement animal/effets des médicaments et des substances chimiques , Hippocampe/effets des médicaments et des substances chimiques , Facteur de croissance endothéliale vasculaire de type A/génétique , Neuroprotecteurs/pharmacologie , Neurogenèse/effets des médicaments et des substances chimiques , Trouble autistique/traitement médicamenteux
19.
Folia Biol (Praha) ; 70(1): 45-52, 2024.
Article de Anglais | MEDLINE | ID: mdl-38830122

RÉSUMÉ

Effective treatment of patients with autism spectrum disorder (ASD) is still absent so far. Taurine exhibits therapeutic effects towards the autism-like behaviour in ASD model animals. Here, we determined the mechanism of taurine effect on hippocampal neurogenesis in genetically inbred BTBR T+ tf/J (BTBR) mice, a proposed model of ASD. In this ASD mouse model, we explored the effect of oral taurine supplementation on ASD-like behaviours in an open field test, elevated plus maze, marble burying test, self-grooming test, and three-chamber test. The mice were divided into four groups of normal controls (WT) and models (BTBR), who did or did not receive 6-week taurine supplementation in water (WT, WT+ Taurine, BTBR, and BTBR+Taurine). Neurogenesis-related effects were determined by Ki67 immunofluorescence staining. Western blot analysis was performed to detect the expression of phosphatase and tensin homologue deleted from chromosome 10 (PTEN)/mTOR/AKT pathway-associated proteins. Our results showed that taurine improved the autism-like behaviour, increased the proliferation of hippocampal cells, promoted PTEN expression, and reduced phosphorylation of mTOR and AKT in hippocampal tissue of the BTBR mice. In conclusion, taurine reduced the autism-like behaviour in partially inherited autism model mice, which may be associa-ted with improving the defective neural precursor cell proliferation and enhancing the PTEN-associated pathway in hippocampal tissue.


Sujet(s)
Trouble autistique , Hippocampe , Neurogenèse , Phosphohydrolase PTEN , Protéines proto-oncogènes c-akt , Transduction du signal , Sérine-thréonine kinases TOR , Taurine , Animaux , Taurine/pharmacologie , Hippocampe/métabolisme , Hippocampe/effets des médicaments et des substances chimiques , Sérine-thréonine kinases TOR/métabolisme , Phosphohydrolase PTEN/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Neurogenèse/effets des médicaments et des substances chimiques , Trouble autistique/métabolisme , Trouble autistique/traitement médicamenteux , Mâle , Comportement animal/effets des médicaments et des substances chimiques , Souris , Modèles animaux de maladie humaine , Trouble du spectre autistique/métabolisme , Trouble du spectre autistique/traitement médicamenteux , Prolifération cellulaire/effets des médicaments et des substances chimiques
20.
Clin Neuropharmacol ; 47(3): 97-100, 2024.
Article de Anglais | MEDLINE | ID: mdl-38743603

RÉSUMÉ

OBJECTIVE: Ketamine and esketamine have been used in the field of psychiatry to alleviate conditions such as major depressive disorder. Our objective was to evaluate the current literature on the use of ketamine for symptoms of social withdrawal in autism spectrum disorder (ASD) and autism-like conditions. METHODS: A comprehensive search of PubMed and Web of Science was conducted to identify literature involving the use of ketamine to treat symptoms of autism and social withdrawal. Patients with comorbid disorders were also included. RESULTS: Two original studies were found, showing mixed results on the use of ketamine for ASD. The use of esketamine found no statistically significant results, whereas the use of intravenous ketamine was shown to alleviate symptoms of social withdrawal especially in the short term. Neither study reported a significant amount of serious adverse events. Five case reports were also included, showing decreased depressive symptoms and evidence of increased social condition. CONCLUSIONS: Research on the use of ketamine for ASD and ASD-related conditions is limited. Evidence of improved social condition exists, but further studies should be conducted to increase sample power and test various doses and methods of administration.


Sujet(s)
Kétamine , Kétamine/usage thérapeutique , Humains , Trouble du spectre autistique/traitement médicamenteux , Trouble autistique/traitement médicamenteux
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE