Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.704
Filter
1.
JAMA Netw Open ; 7(6): e2414709, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38833248

ABSTRACT

Importance: Concerns exist about teratogenic and long-term neurodevelopmental outcomes of paternal use of valproate during spermatogenesis. Objective: To evaluate the association between paternal use of valproate during spermatogenesis and offspring risk of congenital malformations and neurodevelopmental disorders. Design, Setting, and Participants: This nationwide cohort study included 1 235 353 singletons born in Denmark between January 1, 1997, and December 31, 2017, identified in the Medical Birth Register; 1336 children had fathers who had filled prescriptions for valproate during spermatogenesis. Congenital malformations were identified in the first year of life and neurodevelopmental disorders were identified from 1 year of age until December 31, 2018. Statistical analysis was performed March 2024. Exposures: Paternal valproate exposure was defined as fathers who filled 1 or more prescriptions for valproate immediately before or during the time of spermatogenesis (ie, 3 months prior to conception). Main Outcomes and Measures: Children with major congenital malformations in the first year of life and with neurodevelopmental disorders before death or end of follow-up were identified in Danish health registers. Log-binomial regression was used to estimate adjusted relative risks (ARRs) of congenital malformations, and Cox proportional hazards regression was used to estimate adjusted hazards ratios (AHRs) of neurodevelopmental disorders, adjusted for relevant confounders. Results: Among 1 235 353 live births (634 415 boys [51.4%] and 600 938 girls [48.6%]), 1336 children (0.1%) had fathers who filled prescriptions for valproate during spermatogenesis. The median follow-up was 10.1 years (IQR, 5.1-14.8 years) for valproate-exposed children and 10.3 years (IQR, 5.2-15.6 years) for valproate-unexposed children. A total of 43 903 children (3.6%) received a diagnosis of major congenital malformations in the first year of life, and 51 633 children (4.2%) received a diagnosis of neurodevelopmental disorders during follow-up. When comparing the risk among valproate-exposed children with that among unexposed children, the ARR of major congenital malformations was 0.89 (95% CI, 0.67-1.18), the AHR of neurodevelopmental disorders was 1.10 (95% CI, 0.88-1.37), and the AHR of autism spectrum disorder was 0.92 (95% CI, 0.65-1.30). In analyses addressing the robustness of the findings (ie, dose-response analyses, sibling analyses, analyses restricted to children of fathers with epilepsy, analyses that used children with paternal lamotrigine exposure as active comparator, and analyses that used children with paternal exposure to valproate only before spermatogenesis as a negative control exposure), there still was no increased risk of any of the included end points. Conclusions and Relevance: In all analyses based on this large Danish cohort study, results suggest that exposure to valproate during spermatogenesis was not associated with offspring risk of congenital malformations or neurodevelopmental disorders, including autism spectrum disorder.


Subject(s)
Neurodevelopmental Disorders , Spermatogenesis , Valproic Acid , Humans , Valproic Acid/adverse effects , Valproic Acid/therapeutic use , Male , Denmark/epidemiology , Spermatogenesis/drug effects , Female , Neurodevelopmental Disorders/epidemiology , Neurodevelopmental Disorders/chemically induced , Infant , Adult , Cohort Studies , Child, Preschool , Child , Paternal Exposure/adverse effects , Anticonvulsants/adverse effects , Anticonvulsants/therapeutic use , Registries , Infant, Newborn , Abnormalities, Drug-Induced/epidemiology , Risk Factors , Congenital Abnormalities/epidemiology , Prenatal Exposure Delayed Effects/epidemiology , Prenatal Exposure Delayed Effects/chemically induced
4.
Sci Rep ; 14(1): 11435, 2024 05 19.
Article in English | MEDLINE | ID: mdl-38763939

ABSTRACT

Autism spectrum disorder (ASD) exhibits a gender bias, with boys more frequently affected than girls. Similarly, in mouse models induced by prenatal exposure to valproic acid (VPA), males typically display reduced sociability, while females are less affected. Although both males and females exhibit VPA effects on neuroinflammatory parameters, these effects are sex-specific. Notably, females exposed to VPA show increased microglia and astrocyte density during the juvenile period. We hypothesized that these distinct neuroinflammatory patterns contribute to the resilience of females to VPA. To investigate this hypothesis, we treated juvenile animals with intraperitoneal bacterial lipopolysaccharides (LPS), a treatment known to elicit brain neuroinflammation. We thus evaluated the impact of juvenile LPS-induced inflammation on adult sociability and neuroinflammation in female mice prenatally exposed to VPA. Our results demonstrate that VPA-LPS females exhibit social deficits in adulthood, overriding the resilience observed in VPA-saline littermates. Repetitive behavior and anxiety levels were not affected by either treatment. We also evaluated whether the effect on sociability was accompanied by heightened neuroinflammation in the cerebellum and hippocampus. Surprisingly, we observed reduced astrocyte and microglia density in the cerebellum of VPA-LPS animals. These findings shed light on the complex interactions between prenatal insults, juvenile inflammatory stimuli, and sex-specific vulnerability in ASD-related social deficits, providing insights into potential therapeutic interventions for ASD.


Subject(s)
Autism Spectrum Disorder , Lipopolysaccharides , Prenatal Exposure Delayed Effects , Social Behavior , Valproic Acid , Animals , Female , Prenatal Exposure Delayed Effects/chemically induced , Pregnancy , Mice , Valproic Acid/adverse effects , Male , Autism Spectrum Disorder/chemically induced , Autism Spectrum Disorder/etiology , Microglia/drug effects , Microglia/metabolism , Disease Models, Animal , Behavior, Animal/drug effects , Astrocytes/drug effects , Astrocytes/metabolism , Mice, Inbred C57BL
5.
JAMA Netw Open ; 7(5): e2412680, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38776082

ABSTRACT

Importance: Teratogenic outcomes associated with valproic acid use represent a substantial concern for persons of childbearing age. Regulatory agencies worldwide have enhanced warnings or implemented risk minimization programs to reduce exposure during pregnancy. Objectives: To determine pregnancy rates during valproic acid use and concomitant contraception use across indications. Design, Setting, and Participants: This retrospective cohort study used data from the Merative MarketScan commercial claims databases from January 1, 2005, to December 31, 2020, to identify female patients aged 12 to 44 years who initiated valproic acid treatment and had continuous insurance enrollment 6 months before initiation and 9 months after treatment end. A treatment episode included consecutive prescription fills that occurred within 7 days from the end of the days' supply of the previous dispensing. Data were analyzed from March 1 to September 10, 2023. Main Outcomes and Measures: Treatment episodes were categorized by inferred indication using diagnoses preceding treatment initiation, including epilepsy, migraine or headache, mood disorders, and unknown or off-label uses. Pregnancy incidence rate ratios (IRRs) were calculated and were adjusted for age and calendar year. Contraceptive use (prescription contraceptives, intrauterine devices, and implants) during treatment was examined. Results: The cohort included 165 772 valproic acid treatment episodes among 69 390 women (mean [SD] age, 29.8 [10.0] years). Mood disorders (42.5%) were the most common indication, followed by migraine or headache (20.1%), with epilepsy playing a minor role (14.9%). Pregnancy incidence rates during valproic acid use remained unchanged, with a rate of 1.74 (95% CI, 1.14-2.53) per 100 person-years in 2005 and a rate of 1.90 (95% CI, 1.16-3.12) per 100 person-years in 2019. Compared with epilepsy, pregnancy rates were more than double for mood disorder (IRR, 2.16 [95% CI, 1.93-2.42]) and migraine or headache (IRR, 2.01 [95% CI, 1.92-2.09]). Few treatment episodes coincided with contraceptive use (37 012 [22.3%]), and oral dosage forms were the most common (27 069 [73.1%]). Conclusions and Relevance: In this cohort study of patients of childbearing age who used valproic acid, pregnancy rates during valproic acid use did not decrease despite enhanced US Food and Drug Administration safety communications, and contraception use remained low. Patients with migraine and mood disorders accounted for the largest proportion of valproic acid use and had the highest pregnancy rates, while patients with epilepsy had the lowest. These findings suggest a need to enhance efforts to mitigate prenatal exposure to valproic acid, especially for indications where the risk of use during pregnancy outweighs the benefit.


Subject(s)
Epilepsy , Prenatal Exposure Delayed Effects , Valproic Acid , Humans , Female , Valproic Acid/adverse effects , Valproic Acid/therapeutic use , Pregnancy , Adult , Retrospective Studies , Adolescent , Prenatal Exposure Delayed Effects/epidemiology , Epilepsy/drug therapy , Young Adult , Anticonvulsants/adverse effects , Anticonvulsants/therapeutic use , Child , Pregnancy Rate , Mood Disorders/drug therapy , Mood Disorders/epidemiology , Migraine Disorders/drug therapy , United States/epidemiology
6.
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
7.
Birth Defects Res ; 116(4): e2336, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38624050

ABSTRACT

BACKGROUND: According to reports, prenatal exposure to valproic acid can induce autism spectrum disorder (ASD)-like symptoms in both humans and rodents. However, the exact cause and therapeutic method of ASD is not fully understood. Agmatine (AGM) is known for its neuroprotective effects, and this study aims to explore whether giving agmatine hydrochloride before birth can prevent autism-like behaviors in mouse offspring exposed prenatally to valproic acid. METHODS: In this study, we investigated the effects of AGM prenatally on valproate (VPA)-exposed mice. We established a mouse model of ASD by prenatally administering VPA. From birth to weaning, we evaluated mouse behavior using the marble burying test, open-field test, and three-chamber social interaction test on male offspring. RESULTS: The results showed prenatal use of AGM relieved anxiety and hyperactivity behaviors as well as ameliorated sociability of VPA-exposed mice in the marble burying test, open-field test, and three-chamber social interaction test, and this protective effect might be attributed to the activation of the ERK/CREB/BDNF signaling pathway. CONCLUSION: Therefore, AGM can effectively reduce the likelihood of offspring developing autism to a certain extent when exposed to VPA during pregnancy, serving as a potential therapeutic drug.


Subject(s)
Agmatine , Autism Spectrum Disorder , Animals , Female , Male , Mice , Pregnancy , Agmatine/pharmacology , Autism Spectrum Disorder/chemically induced , Autism Spectrum Disorder/drug therapy , Autism Spectrum Disorder/prevention & control , Brain-Derived Neurotrophic Factor , Calcium Carbonate , Rodentia , Signal Transduction , Social Behavior , Valproic Acid/adverse effects
8.
Biomolecules ; 14(4)2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38672454

ABSTRACT

Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by severe deficits in social communication and interaction, repetitive movements, abnormal focusing on objects, or activity that can significantly affect the quality of life of the afflicted. Neuronal and glial cells have been implicated. It has a genetic component but can also be triggered by environmental factors or drugs. For example, prenatal exposure to valproic acid or acetaminophen, or ingestion of propionic acid, can increase the risk of ASD. Recently, epigenetic influences on ASD have come to the forefront of investigations on the etiology, prevention, and treatment of this disorder. Epigenetics refers to DNA modifications that alter gene expression without making any changes to the DNA sequence. Although an increasing number of pharmaceuticals and environmental chemicals are being implicated in the etiology of ASD, here, we specifically focus on the molecular influences of the abovementioned chemicals on epigenetic alterations in neuronal and glial cells and their potential connection to ASD. We conclude that a better understanding of these phenomena can lead to more effective interventions in ASD.


Subject(s)
Autism Spectrum Disorder , Epigenesis, Genetic , Neuroglia , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/chemically induced , Humans , Epigenesis, Genetic/drug effects , Neuroglia/metabolism , Neuroglia/drug effects , Valproic Acid/pharmacology , Valproic Acid/adverse effects , Propionates/pharmacology , Animals , Acetaminophen/adverse effects , Neurons/metabolism , Neurons/drug effects , Neurons/pathology , DNA Methylation/drug effects
10.
Epilepsy Behav ; 155: 109802, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38657483

ABSTRACT

OBJECTIVE: Epilepsy is a chronic disease that requires long-term monitoring and treatment. It is suspected that there is a interaction between the use of anti-seizure medications and the risk of cardiovascular disease. The aim of the study is to investigate the association between the intake of phenobarbital, carbamazepine and valproic acid and their serum drug concentrations (SDC) with various cardiovascular risk parameters (homocysteine, folic acid, vitamin B12, total cholesterol (TC), triglycerides, high- and low-density lipoprotein (LDL)). METHODS: This is a cross-sectional study. Data (demographic characteristics and laboratory results) of patients treated for epilepsy in a tertiary care hospital between January 2020 and February 2022 were analyzed retrospectively (n = 2014). Kruskal Wallis, Mann-Whitney U, correlation analysis was used, p < 0.05 was considered statistically significant. RESULTS: The median age of patients was 15 years (IQR:8-31) and 48.3 % were women. The highest homocysteine level was found in patients receiving valproic acid, but it was not statistically significant. Patients receiving phenobarbital had the highest levels of folic acid and B12 and the lowest levels of total cholesterol and low-density lipoprotein cholesterol, which was statistically significant. In patients receiving carbamazepine, a moderately negative significant association was found between serum drug concentration and folic acid levels and a moderately positive significant association was found between TC and LDL levels. CONCLUSION: In our study, the majority of patients were children and adolescents. Regular monitoring of drug serum concentrations and metabolic parameters may be useful to select the safest drug in terms of cardiovascular disease risk. Randomized controlled trials on the long-term effects of anti-seizure treatment are needed.


Subject(s)
Anticonvulsants , Carbamazepine , Cardiovascular Diseases , Epilepsy , Valproic Acid , Humans , Female , Anticonvulsants/therapeutic use , Anticonvulsants/blood , Anticonvulsants/adverse effects , Cross-Sectional Studies , Male , Adult , Epilepsy/drug therapy , Epilepsy/blood , Adolescent , Young Adult , Valproic Acid/therapeutic use , Valproic Acid/adverse effects , Valproic Acid/blood , Cardiovascular Diseases/blood , Child , Carbamazepine/therapeutic use , Carbamazepine/blood , Carbamazepine/adverse effects , Homocysteine/blood , Phenobarbital/therapeutic use , Phenobarbital/blood , Retrospective Studies , Vitamin B 12/blood , Heart Disease Risk Factors , Folic Acid/blood
12.
Biomolecules ; 14(3)2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38540788

ABSTRACT

(1) Background: Valproic acid (VPA) is one of the frequently prescribed antiepileptic drugs and is generally considered well tolerated. However, VPA neurologic adverse effects in the absence of liver failure are fairly common, suggesting that in the mechanism for the development of VPA-induced encephalopathy, much more is involved than merely the exposure to hyperammonemia (HA) caused by liver insufficiency to perform detoxification. Taking into account the importance of the relationship between an impaired brain energy metabolism and elevated ammonia production, and based on the ability of VPA to interfere with neuronal oxidative pathways, the current study intended to investigate a potential regional ammoniagenic effect of VPA on rats' brains by determining activities of the enzymes responsible for ammonia production and neutralization. (2) Methods: Rats received a single intraperitoneal injection of VPA (50, 100, 250, 500 mg/kg). Plasma, the neocortex, the cerebellum, and the hippocampus were collected at 30 min after injection. The levels of ammonia, urea, aspartate aminotransferase (AST), and alanine aminotransferase (ALT) were measured in blood plasma. The activities of glutaminase and glutamate dehydrogenase (GDH) in mitochondria and the activities of AMP deaminase (AMPD), adenosine deaminase (ADA), and glutamine synthetase (GS) in cytosolic fractions isolated from rat brain regions were measured. Ammonia, ALT, and AST values were determined in the mitochondrial and cytosolic fractions. (3) Results: Multi-dose VPA treatment did not significantly affect the plasma levels of ammonia and urea or the ALT and AST liver enzymes. Significant dose-independent increases in the accumulation of ammonia were found only in the cytosol from the cerebellum and there was a strong correlation between the ammonia level and the ADA activity in this brain structure. A significant decrease in the AMPD and AST activities was observed, while the ALT activity was unaffected. Only the highest VPA dose (500 mg/kg) was associated with significantly less activity of GS compared to the control in all studied brain structures. In the mitochondria of all studied brain structures, VPA caused a dose-independent increases in ammonia levels, a high concentration of which was strongly and positively correlated with the increased GDH and ALT activity, while glutaminase activity remained unchanged, and AST activity significantly decreased compared to the control in all studied brain structures. (4) Conclusions: This study highlights the rat brain region-specific ammoniagenic effects of VPA, which may manifest themselves in the absence of hyperammonemia. Further research should analyze how the responsiveness of the different brain regions may vary in VPA-treated animals that exhibit compromised energy metabolism, leading to increased ammoniagenesis.


Subject(s)
Hyperammonemia , Valproic Acid , Rats , Animals , Valproic Acid/adverse effects , Glutaminase , Hyperammonemia/chemically induced , Hyperammonemia/metabolism , Ammonia/metabolism , Urea
13.
Palliat Med ; 38(4): 492-497, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38444061

ABSTRACT

BACKGROUND: Seizures are an important palliative symptom, the management of which can be complicated by patients' capacity to swallow oral medications. In this setting, and the wish to avoid intravenous access, subcutaneous infusions may be employed. Options for antiseizure medications that can be provided subcutaneously may be limited. Subcutaneous sodium valproate may be an additional management strategy. AIM: To evaluate the published experience of subcutaneous valproate use in palliative care, namely with respect to effectiveness and tolerability. DESIGN: A systematic review was registered (PROSPERO CRD42023453427), conducted and reported according to PRISMA reporting guidelines. DATA SOURCES: The databases PubMed, EMBASE and Scopus were searched for publications until August 11, 2023. RESULTS: The searches returned 429 results, of which six fulfilled inclusion criteria. Case series were the most common study design, and most studies included <10 individuals who received subcutaneous sodium valproate. There were three studies that presented results on the utility of subcutaneous sodium valproate for seizure control, which described it to be an effective strategy. One study also described it as an effective treatment for neuropathic pain. The doses were often based on presumed 1:1 oral to subcutaneous conversion ratios. Only one study described a local site adverse reaction, which resolved with a change of administration site. CONCLUSIONS: There are limited data on the use of subcutaneous sodium valproate in palliative care. However, palliative symptoms for which subcutaneous sodium valproate have been used successfully are seizures and neuropathic pain. The available data have described few adverse effects, supporting its use with an appropriate degree of caution.


Subject(s)
Neuralgia , Valproic Acid , Humans , Valproic Acid/adverse effects , Palliative Care , Seizures/chemically induced , Seizures/drug therapy , Neuralgia/drug therapy
15.
Clin Toxicol (Phila) ; 62(2): 101-106, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38512019

ABSTRACT

BACKGROUND: Valproic acid has been widely used as an antiepileptic drug for several decades. Long-term valproic acid treatment is usually accompanied by liver injury. Although both men and women are susceptible to valproic acid-associated liver injury, hepatotoxicity differs between the sexes. However, the mechanisms underlying sex differences in valproic acid-associated liver injury remain unclear. METHODS: To explore potential risk factors for the susceptibility to valproic acid-associated liver injury, 231 pediatric patients with epilepsy (119 males, 112 females) were enrolled for laboratory and genetic analysis. RESULTS: Heterozygous genotype of catalase C-262T (P = 0.045) and the concentrations of glutathione (P = 0.002) and thiobarbituric acid-reactive substances (P = 0.011) were associated with the sex-specific susceptibility to valproic acid-associated liver injury. Meanwhile, logistic regression analysis revealed that carriers of heterozygous genotype of catalase C-262T (P = 0.010, odds ratio: 4.163; 95 percent confidence interval 1.400 - 7.378), glutathione concentration (P = 0.001, odds ratio: 2.421; 95 percent confidence interval 2.262 - 2.591) and male patients (P = 0.005, odds ratio: 1.344; 95% confidence interval 0.782 - 2.309) had a higher risk for valproic acid-associated liver injury. DISCUSSION: The mechanism underlying valproic acid-induced hepatotoxicity remains unclear. Additionally, factors that may contribute to the observed differences in the incidence of hepatotoxicity between males and females have yet to be defined. This study identifies several genetic factors that may predispose patients to valproic acid-associated hepatotoxicity. LIMITATIONS: This relatively small sample size of children with one ethnicity some of whom were taking other antiepileptics that are potentially hepatotoxic. CONCLUSION: Catalase C-262T genotype, glutathione concentration and gender (male) are potential risk factors for the susceptibility to valproic acid-associated liver injury.


Subject(s)
Chemical and Drug Induced Liver Injury , Epilepsy , Humans , Female , Male , Child , Valproic Acid/adverse effects , Sex Characteristics , Catalase/genetics , Epilepsy/drug therapy , Glutathione , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/genetics
16.
Epilepsy Behav ; 153: 109733, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38447300

ABSTRACT

OBJECTIVE: This study aimed to evaluate the impact of prolonged sodium valproate use on bone mineral density (BMD) and Vitamin D levels in pediatric epilepsy patients. METHODS: In a cross-sectional study conducted at the Epilepsy Clinic of Niloufer Hospital, Hyderabad, India, 50 pediatric patients (aged 4-10 years) were recruited. The cohort comprised 30 epilepsy patients on sodium valproate treatment (cases) and 20 healthy siblings without epilepsy or valproate use (controls). BMD was assessed using dual-energy X-ray absorptiometry to measure height-adjusted total body less head Z-scores (TBLH Z-scores), and serum 25-hydroxyvitamin D levels were measured. Statistical analysis included independent samples t-tests, Mann-Whitney U tests, and Pearson correlation, with a preliminary power analysis ensuring adequate sample size. RESULTS: Cases exhibited significantly lower BMD TBLH Z-scores (Mean = -1.543) compared to controls (Mean = 0.515, p <.001) and reduced Vitamin D levels (Mean = 9.17 for cases vs. 27.80 for controls, p <.001). A negative correlation was observed between the duration of sodium valproate use and both BMD Z-scores (r = -0.626, p <.001) and Vitamin D levels (r = -0.707, p <.001). CONCLUSIONS: The findings suggest a significant negative impact of prolonged sodium valproate use on both bone density and Vitamin D levels in pediatric patients. These results underscore the importance of monitoring and managing bone health in children receiving long-term sodium valproate therapy.


Subject(s)
Bone Density , Epilepsy , Humans , Child , Valproic Acid/adverse effects , Cross-Sectional Studies , Absorptiometry, Photon , Vitamin D , Epilepsy/drug therapy , Epilepsy/chemically induced , Vitamins
17.
JAMA Neurol ; 81(5): 481-489, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38497990

ABSTRACT

Importance: Women with epilepsy (WWE) require treatment with antiseizure medications (ASMs) during pregnancy, which may be associated with an increased risk of major congenital malformations (MCMs) in their offspring. Objective: To investigate the prevalence of MCMs after prenatal exposure to 8 commonly used ASM monotherapies and changes in MCM prevalence over time. Design, Setting, and Participants: This was a prospective, observational, longitudinal cohort study conducted from June 1999 to October 2022. Since 1999, physicians from more than 40 countries enrolled ASM-treated WWE before pregnancy outcome was known and followed up their offspring until 1 year after birth. Participants aged 14 to 55 years who were exposed to 8 of the most frequently used ASMs during pregnancy were included in this study. Data were analyzed from April to September 2023. Exposure: Maternal use of ASMs at conception. Main Outcomes and Measures: MCMs were assessed 1 year after birth by a committee blinded to type of exposure. Teratogenic outcomes across exposures were compared by random-effects logistic regression adjusting for potential confounders and prognostic factors. Results: A total of 10 121 prospective pregnancies exposed to ASM monotherapy met eligibility criteria. Of those, 9840 were exposed to the 8 most frequently used ASMs. The 9840 pregnancies occurred in 8483 women (mean [range] age, 30.1 [14.1-55.2] years). MCMs occurred in 153 of 1549 pregnancies for valproate (9.9%; 95% CI, 8.5%-11.5%), 9 of 142 for phenytoin (6.3%; 95% CI, 3.4%-11.6%), 21 of 338 for phenobarbital (6.2%; 95% CI, 4.1%-9.3%), 121 of 2255 for carbamazepine (5.4%; 95% CI, 4.5%-6.4%), 10 of 204 for topiramate (4.9%; 95% CI, 2.7%-8.8%), 110 of 3584 for lamotrigine (3.1%; 95% CI, 2.5%-3.7%), 13 of 443 for oxcarbazepine (2.9%; 95% CI, 1.7%-5.0%), and 33 of 1325 for levetiracetam (2.5%; 95% CI, 1.8%-3.5%). For valproate, phenobarbital, and carbamazepine, there was a significant increase in the prevalence of MCMs associated with increasing dose of the ASM. Overall prevalence of MCMs decreased from 6.1% (153 of 2505) during the period 1998 to 2004 to 3.7% (76 of 2054) during the period 2015 to 2022. This decrease over time was significant in univariable logistic analysis but not after adjustment for changes in ASM exposure pattern. Conclusions and Relevance: Of all ASMs with meaningful data, the lowest prevalence of MCMs was observed in offspring exposed to levetiracetam, oxcarbazepine, and lamotrigine. Prevalence of MCMs was higher with phenytoin, valproate, carbamazepine, and phenobarbital, and dose dependent for the latter 3 ASMs. The shift in exposure pattern over time with a declining exposure to valproate and carbamazepine and greater use of lamotrigine and levetiracetam was associated with a 39% decline in prevalence of MCMs, a finding that has major public health implications.


Subject(s)
Abnormalities, Drug-Induced , Anticonvulsants , Epilepsy , Pregnancy Complications , Humans , Female , Anticonvulsants/adverse effects , Anticonvulsants/therapeutic use , Adult , Pregnancy , Young Adult , Adolescent , Epilepsy/drug therapy , Epilepsy/epidemiology , Abnormalities, Drug-Induced/epidemiology , Abnormalities, Drug-Induced/etiology , Middle Aged , Longitudinal Studies , Pregnancy Complications/drug therapy , Pregnancy Complications/epidemiology , Prospective Studies , Valproic Acid/adverse effects , Valproic Acid/therapeutic use , Prenatal Exposure Delayed Effects/epidemiology , Prenatal Exposure Delayed Effects/chemically induced , Phenytoin/adverse effects , Phenytoin/therapeutic use , Lamotrigine/adverse effects , Lamotrigine/therapeutic use , Carbamazepine/adverse effects , Phenobarbital/adverse effects , Phenobarbital/therapeutic use , Cohort Studies , Oxcarbazepine/adverse effects , Oxcarbazepine/therapeutic use , Prevalence
18.
Auton Neurosci ; 253: 103161, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38461695

ABSTRACT

Autism spectrum disorder (ASD) has increased in incidence over the past several decades, and is associated with a range of co-morbidities including gastrointestinal (GI) dysfunctions including gastroesophageal reflux, abdominal pain, bloating, constipation and/or diarrhea. Several animal models have been used that replicate several aspects of ASD but no single model has been able to replicate the entire disease pathophysiology. In humans, prenatal exposure to valproic acid (VPA) has been identified as a significant risk factor and rodent models have shown that in utero VPA exposure leads to behavioral deficits in offspring. The present study aimed to investigate whether in utero exposure to VPA induces GI dysfunction in rats. Timed pregnant Sprague-Dawley rats were injected with a single dose of VPA at embryonic day 12.5. Both male and female offspring subsequently underwent behavioral studies and assessment of GI function in adulthood. In utero VPA treatment induced social deficits in both male and female offspring, decreasing sociability and social novelty. Histological examination showed that VPA treated offspring had decreased thickness of GI muscle and mucosa, while immunohistochemical studies showed a decrease in myenteric neuron number in the fundus. Functional studies showed that both male and female VPA offspring had a delay in gastric emptying compared to vehicle treated offspring. Results of the current study suggest that the rat VPA model of behavioral deficits may be a convenient model by which both mechanistic and functional insights into GI dysfunction may be studied.


Subject(s)
Disease Models, Animal , Gastrointestinal Diseases , Prenatal Exposure Delayed Effects , Rats, Sprague-Dawley , Valproic Acid , Animals , Valproic Acid/toxicity , Valproic Acid/adverse effects , Female , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/physiopathology , Male , Gastrointestinal Diseases/chemically induced , Gastrointestinal Diseases/physiopathology , Rats , Social Behavior , Autism Spectrum Disorder/chemically induced , Autism Spectrum Disorder/physiopathology , Gastric Emptying/drug effects , Gastric Emptying/physiology
19.
N Engl J Med ; 390(12): 1069-1079, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38507750

ABSTRACT

BACKGROUND: Maternal use of valproate during pregnancy has been associated with an increased risk of neurodevelopmental disorders in children. Although most studies of other antiseizure medications have not shown increased risks of these disorders, there are limited and conflicting data regarding the risk of autism spectrum disorder associated with maternal topiramate use. METHODS: We identified a population-based cohort of pregnant women and their children within two health care utilization databases in the United States, with data from 2000 through 2020. Exposure to specific antiseizure medications was defined on the basis of prescription fills from gestational week 19 until delivery. Children who had been exposed to topiramate during the second half of pregnancy were compared with those unexposed to any antiseizure medication during pregnancy with respect to the risk of autism spectrum disorder. Valproate was used as a positive control, and lamotrigine was used as a negative control. RESULTS: The estimated cumulative incidence of autism spectrum disorder at 8 years of age was 1.9% for the full population of children who had not been exposed to antiseizure medication (4,199,796 children). With restriction to children born to mothers with epilepsy, the incidence was 4.2% with no exposure to antiseizure medication (8815 children), 6.2% with exposure to topiramate (1030 children), 10.5% with exposure to valproate (800 children), and 4.1% with exposure to lamotrigine (4205 children). Propensity score-adjusted hazard ratios in a comparison with no exposure to antiseizure medication were 0.96 (95% confidence interval [CI], 0.56 to 1.65) for exposure to topiramate, 2.67 (95% CI, 1.69 to 4.20) for exposure to valproate, and 1.00 (95% CI, 0.69 to 1.46) for exposure to lamotrigine. CONCLUSIONS: The incidence of autism spectrum disorder was higher among children prenatally exposed to the studied antiseizure medications than in the general population. However, after adjustment for indication and other confounders, the association was substantially attenuated for topiramate and lamotrigine, whereas an increased risk remained for valproate. (Funded by the National Institute of Mental Health.).


Subject(s)
Anticonvulsants , Autism Spectrum Disorder , Lamotrigine , Prenatal Exposure Delayed Effects , Topiramate , Valproic Acid , Child , Female , Humans , Pregnancy , Anticonvulsants/adverse effects , Anticonvulsants/therapeutic use , Autism Spectrum Disorder/chemically induced , Autism Spectrum Disorder/epidemiology , Autism Spectrum Disorder/etiology , Autistic Disorder/chemically induced , Autistic Disorder/epidemiology , Autistic Disorder/etiology , Lamotrigine/adverse effects , Lamotrigine/therapeutic use , Prenatal Exposure Delayed Effects/epidemiology , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/drug therapy , Topiramate/adverse effects , Topiramate/therapeutic use , Valproic Acid/adverse effects , Valproic Acid/therapeutic use , Epilepsy/drug therapy
20.
Int J Dev Neurosci ; 84(2): 134-142, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38304999

ABSTRACT

Autism spectrum disorder (ASD) is a widespread neurodevelopmental disorder with unknown etiology. Dysfunction of several brain areas including the prefrontal cortex (PFC), hippocampus, and cerebellum is involved in cognitive and behavioral deficits associated with ASD. Several studies have reported a reduction in the number of parvalbumin-immunoreactive (PV+) neurons in brain areas of ASD patients and animal models such as a shank mutant mouse model and rodents receiving fetal valproic acid (VPA) administration. Developing therapeutic interventions that restore PV interneurons can be the future therapeutic approach to ASD. The present study examined the possible effect of agmatine (AG), an endogenous NMDA antagonist, on the number of PV+ neurons in a VPA animal model of autism. The therapeutic effects of AG in ameliorating ASD-like behaviors were previously reported in VPA rats. AG was gavaged at dosages of 0.001, 0.01, and 0.1 mg/kg from gestational day (GD) 6.5 to 18.5, and the number of PV interneurons was analyzed by immunohistochemistry in the 1-month-old rats. Prenatal VPA (GD 12.5) or AG led to a decrease of PV neurons in the PFC, Cornu ammonia (CA1), and molecular layers (MLs) of the cerebellum. However, exposure to AG restored the PV population induced by VPA. AG may modify underlying neuronal mechanisms resulting in the increased survival or restoration of the PV population.


Subject(s)
Agmatine , Autism Spectrum Disorder , Parvalbumins , Prenatal Exposure Delayed Effects , Valproic Acid , Animals , Female , Humans , Infant , Mice , Pregnancy , Rats , Agmatine/therapeutic use , Autism Spectrum Disorder/chemically induced , Behavior, Animal , Disease Models, Animal , Neurons , Parvalbumins/metabolism , Prenatal Exposure Delayed Effects/chemically induced , Valproic Acid/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...