1
|
Oliveira MS, Fernandes RA, Pinto LS, Moreira FA, Castro OWD, Santos VR. Balancing efficacy and safety: The dual impact of antiseizure medications on the developing brain. Epilepsy Behav 2025; 167:110400. [PMID: 40187052 DOI: 10.1016/j.yebeh.2025.110400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 02/25/2025] [Accepted: 03/21/2025] [Indexed: 04/07/2025]
Abstract
The number of neurons in the developing brain is greater than typically found in adulthood, and the brain possesses delicate mechanisms to induce the death of excess cells and refine neural circuitry. The correct tuning between the processes of neuronal death and survival generates a mature and functional brain in its complexity and plastic capacity. Epilepsy is a highly prevalent neurological condition worldwide, including among young individuals. However, exposure to the main treatment approaches, the long-term use of Antiseizure Medication (ASM), during the critical period of development can induce a series of changes in this delicate balance. Acting by various mechanisms of action, ASMs may induce an increase in neuronal death, something that translates into deleterious neuropsychiatric effects in adulthood. Several investigations conducted in recent years have brought to light new aspects related to this dynamic, yet many questions, such as the cellular mechanisms of death and the pathophysiology of late effects, still have unresolved elements. In this review, we aimed to explore the mechanisms of action of the most widely used ASMs in the treatment of neonatal epilepsy, the broad aspects of neuronal death in the developing brain and the repercussions of this death and other effects in adulthood. We review the evidence indicating a relationship between exposure to ASMs and the manifestation of associated psychiatric comorbidities in adulthood and discuss some possible mechanisms underlying the induction of this process by morphological and physiological changes in the related behaviors.
Collapse
Affiliation(s)
- M S Oliveira
- Department of Morphology, Institute of Biological Science, Universidade Federal de Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | - R A Fernandes
- Department of Morphology, Institute of Biological Science, Universidade Federal de Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | - L S Pinto
- Department of Morphology, Institute of Biological Science, Universidade Federal de Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | - F A Moreira
- Department of Pharmacology, Institute of Biological Science, Universidade Federal de Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | - O W de Castro
- Departament of Physiology, Institute of Biological Science and Health, Universidade Federal de Alagoas - UFAL, Brazil
| | - V R Santos
- Department of Morphology, Institute of Biological Science, Universidade Federal de Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| |
Collapse
|
2
|
Di Maio A, Yahyavi I, Buzzelli V, Motta Z, Ascone F, Putignani L, Usiello A, Pollegioni L, Trezza V, Errico F. Prenatal Exposure to Lipopolysaccharide or Valproate Leads to Abnormal Accumulation of the NMDA Receptor Agonist D-Aspartate in the Adolescent Rat Brain. J Neurochem 2025; 169:e70095. [PMID: 40437860 PMCID: PMC12120390 DOI: 10.1111/jnc.70095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 05/09/2025] [Accepted: 05/12/2025] [Indexed: 06/01/2025]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental psychiatric condition linked to glutamatergic neurotransmission disruption. Although endogenous D-serine and D-aspartate modulate glutamatergic N-methyl D-aspartate receptor (NMDAR) activity, their involvement in ASD remains elusive. We measured the levels of D-aspartate, D-serine, and other key neuroactive amino acids, and their direct precursors in brain regions, plasma, and feces of environmental ASD rat models prenatally exposed to lipopolysaccharide or valproate, both during adolescence and early adulthood, as well as in a genetic ASD model, the Fmr1-Δexon8 rat. No significant changes were found in plasma and feces. Conversely, we observed a prominent accumulation of D-aspartate in several brain regions of lipopolysaccharide- and valproate-exposed rats, selectively during adolescence, while D-serine level variations were more limited. No significant amino acid changes were observed in the Fmr1-Δexon8 rat brain. We also assayed the activity of the main enzymes involved in cerebral D-serine and D-aspartate metabolism, suggesting that their regulation extends beyond their metabolic enzymes. These findings highlight that prenatal environmental stressors disrupt D-amino acid levels selectively in ASD rat brains, emphasizing the role of early NMDAR dysfunction in ASD-related phenotypes.
Collapse
Affiliation(s)
- Anna Di Maio
- CEINGE Biotecnologie Avanzate “Franco Salvatore”NaplesItaly
- Department of Environmental, Biological and Pharmaceutical Sciences and TechnologiesUniversità degli Studi della Campania “Luigi Vanvitelli”CasertaItaly
| | - Isar Yahyavi
- CEINGE Biotecnologie Avanzate “Franco Salvatore”NaplesItaly
- Department of Environmental, Biological and Pharmaceutical Sciences and TechnologiesUniversità degli Studi della Campania “Luigi Vanvitelli”CasertaItaly
| | | | - Zoraide Motta
- “The Protein Factory 2.0”, Dipartimento di Biotecnologie e Scienze della VitaUniversità degli Studi dell'insubriaVareseItaly
| | | | - Lorenza Putignani
- Unit of Microbiomics and Unit of Research of MicrobiomeBambino Gesù Children's Hospital, IRCCSRomeItaly
- Department of Life Science, Health, and Health Professions, Link Campus UniversityRomeItaly
| | - Alessandro Usiello
- CEINGE Biotecnologie Avanzate “Franco Salvatore”NaplesItaly
- Department of Environmental, Biological and Pharmaceutical Sciences and TechnologiesUniversità degli Studi della Campania “Luigi Vanvitelli”CasertaItaly
| | - Loredano Pollegioni
- “The Protein Factory 2.0”, Dipartimento di Biotecnologie e Scienze della VitaUniversità degli Studi dell'insubriaVareseItaly
| | - Viviana Trezza
- Department of ScienceRoma Tre UniversityRomeItaly
- Neuroendocrinology, Metabolism and Neuropharmacology UnitIRCCS Fondazione Santa LuciaRomeItaly
| | - Francesco Errico
- CEINGE Biotecnologie Avanzate “Franco Salvatore”NaplesItaly
- Dipartimento di AgrariaUniversità degli Studi di Napoli “Federico II”PorticiItaly
| |
Collapse
|
3
|
Zhang Q, Zheng J, Sun H, Zheng J, Ma Y, Ji Q, Chen D, Tang Z, Zhang J, He Y, Song T. The Notch Signaling Pathway: A Potential Target for Mental Disorders. Mol Neurobiol 2025:10.1007/s12035-025-05034-w. [PMID: 40372672 DOI: 10.1007/s12035-025-05034-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 05/02/2025] [Indexed: 05/16/2025]
Abstract
The highly conserved Notch signaling pathway plays a critical role in cell fate determination during metazoan development through cell-to-cell communication. The classical pathway consists of Notch receptors, ligands, intracellular effectors, DNA-binding proteins, and other regulatory molecules. Recent research has highlighted its involvement in the pathogenesis of several diseases. In autism, bipolar disorder, and schizophrenia, the Notch signaling pathway is implicated in key processes such as neuronal development and synaptic plasticity. Furthermore, it has been shown to play significant roles in other mental health conditions, including anxiety, depression, post-traumatic stress disorder, and neurocognitive disorders. However, the precise mechanisms underlying the contribution of Notch to these conditions remain poorly understood. This review examines the current understanding of the Notch signaling pathway in mental disorders, highlighting its role in their pathophysiology and summarizing therapeutic strategies aimed at modulating this pathway to improve mental health outcomes.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Immunology, Zunyi Medical University, Zunyi, China
| | - Jingxuan Zheng
- The Eighth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Hongqin Sun
- Department of Immunology, Zunyi Medical University, Zunyi, China
| | - Jishan Zheng
- Department of Immunology, Zunyi Medical University, Zunyi, China
| | - Yunyan Ma
- Department of Immunology, Zunyi Medical University, Zunyi, China
| | - Qinglu Ji
- School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Dengwang Chen
- Department of Immunology, Zunyi Medical University, Zunyi, China
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Zhengzhen Tang
- Department of Pediatrics, Third Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jidong Zhang
- Department of Immunology, Zunyi Medical University, Zunyi, China
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, China
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Yuqi He
- School of Pharmacy, Zunyi Medical University, Zunyi, China
- Engineering Research Center of Key Technologies for Industrial Development of Dendrobium in Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Tao Song
- Department of Immunology, Zunyi Medical University, Zunyi, China.
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi, China.
| |
Collapse
|
4
|
Amaral de Lara IC, de Souza Wagner PH, Freitas Uchôa Matheus GT, Eduardo Campos L, de Almeida Souza Miranda C, Cavalcanti Souza ME, Aquino de Moraes FC, Alves Kelly F, Rodrigues Fernandes L. Association of prenatal exposure to antiseizure medication with risk of autism: a systematic review and meta-analysis. Seizure 2025; 130:41-47. [PMID: 40354746 DOI: 10.1016/j.seizure.2025.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/29/2025] [Accepted: 05/02/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Antiseizure medications (ASMs) can affect neurodevelopment and cause congenital malformations. Autism spectrum disorder (ASD) is characterized by challenges in communication, behavior, and learning. This study evaluated the association between prenatal ASM exposure and ASD development. METHODS A systematic review and meta-analysis was conducted using PubMed, Scopus, Web of Science, and Cochrane databases to identify studies on fetal ASM exposure and ASD. Hazard ratio (HR) and risk ratio (RR) with 95 % confidence interval (CI) were pooled using a random-effects model. Heterogeneity was assessed using I² statistic. A p-value < 0.05 was considered significant. RESULTS 10 studies were included in our meta-analysis, comprising 54,747 patients exposed to ASM. Prenatal ASM exposure significantly increased the risk of ASD (HR 1.8082; 95 % CI 1.2616 to 2.5916; P = 0.001; RR 2.0401; 95 % CI 1.7588 to 2.3664; P < 0.0001). Subgroup analyses identified elevated risks with specific ASMs, including carbamazepine (HR 1.2213; 95 % CI 1.0047 to 1.4847; P = 0.045; I² = 0 %), valproate (HR 2.8306; 95 % CI 2.3881 to 3.3550; P < 0.001; I² = 0 %), and oxcarbazepine (HR 1.6141; 95 % CI 1.1500 to 2.2655; P = 0.006; I² = 27 %). Among women with epilepsy, prenatal ASM exposure increased ASD risk (RR 1.4174; 95 % CI 1.2345 to 1.6273; P < 0.0001; I² = 0 %). CONCLUSIONS This meta-analysis showed that prenatal exposure to antiseizures, particularly valproate, carbamazepine, and oxcarbazepine, significantly increases ASD risk. These findings emphasize the need for cautious ASM use during pregnancy.
Collapse
Affiliation(s)
| | | | | | - Lara Eduardo Campos
- Federal University of The State of Rio de Janeiro, Rio de Janeiro, 20271-062, Brazil
| | | | | | | | | | | |
Collapse
|
5
|
Brown LTL, Pereira D, Winn LM. A Narrative Review on the Effect of Valproic Acid on the Placenta. Birth Defects Res 2025; 117:e2471. [PMID: 40211937 PMCID: PMC11986804 DOI: 10.1002/bdr2.2471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/10/2025] [Accepted: 03/24/2025] [Indexed: 04/13/2025]
Abstract
BACKGROUND Valproic acid (VPA) is an antiepileptic and mood-stabilizing drug with well-established teratogenic risks when taken during pregnancy. While its harmful effects on fetal development are well known, less attention has been given to its impact on placental development and function, despite the placenta's critical role in pregnancy. AIM This narrative review examines how VPA exposure affects placental growth, morphology, nutrient transport, and epigenetic modifications. It also considers whether placental dysfunction may contribute VPA's teratogenic effects. RESULTS Evidence suggests that VPA disrupts placental structure and growth, alters the expression of nutrient transporters, such as those for folate, glucose, and amino acids, and modifies the placental epigenome, including globally decreased DNA methylation and increased histone acetylation. DISCUSSION It is hypothesized that these epigenetic changes may influence chromatin remodelling and trophoblast gene expression, though this connection has not been fully established. Such epigenetic dysregulation may result in aberrant gene expression that underlies the structural and functional impairments observed in the placenta, potentially compromising its ability to support fetal development and contributing to VPA's teratogenic effects. Findings across studies, however, are inconsistent, varying with dose, timing of exposure, and model system. Furthermore, there is a lack of research examining sex-specific differences in placental responses to VPA, despite evidence that male and female placentas exhibit distinct growth patterns, gene expression profiles, and susceptibilities to environmental insults. CONCLUSION Addressing these knowledge gaps through targeted research will improve our understanding of how VPA affects the placenta and its role in teratogenesis.
Collapse
Affiliation(s)
- Lauren T. L. Brown
- Department of Biomedical and Molecular SciencesQueen's University at KingstonKingstonOntarioCanada
| | - Delaine Pereira
- Department of Biomedical and Molecular SciencesQueen's University at KingstonKingstonOntarioCanada
| | - Louise M. Winn
- Department of Biomedical and Molecular SciencesQueen's University at KingstonKingstonOntarioCanada
- School of Environmental SciencesQueen's University at KingstonKingstonOntarioCanada
| |
Collapse
|
6
|
Practice Guideline From the AAN, AES, and SMFM, Pack AM, Oskoui M, Roberson SW, Donley DK, French J, Gerard EE, Gloss D, Miller WR, Clary HMM, Osmundson SS, McFadden B, Parratt K, Pennell PB, Saade G, Smith DB, Sullivan K, Thomas SV, Tomson T, O’Brien MD, Botchway-Doe K, Silsbee HM, Keezer MR. Teratogenesis, Perinatal, and Neurodevelopmental Outcomes After In Utero Exposure to Antiseizure Medication. Epilepsy Curr 2025:15357597241258514. [PMID: 40083742 PMCID: PMC11897993 DOI: 10.1177/15357597241258514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2025] Open
Abstract
This practice guideline provides updated evidence-based conclusions and recommendations regarding the effects of antiseizure medications (ASMs) and folic acid supplementation on the prevalence of major congenital malformations (MCMs), adverse perinatal outcomes, and neuro-developmental outcomes in children born to people with epilepsy of childbearing potential (PWECP). A multidisciplinary panel conducted a systematic review and developed practice recommendations following the process outlined in the 2017 edition of the American Academy of Neurology Clinical Practice Guideline Process Manual. The systematic review includes studies through August 2022. Recommendations are supported by structured rationales that integrate evidence from the systematic review, related evidence, principles of care, and inferences from evidence. The following are some of the major recommendations. When treating PWECP, clinicians should recommend ASMs and doses that optimize both seizure control and fetal outcomes should pregnancy occur, at the earliest possible opportunity preconceptionally. Clinicians must minimize the occurrence of convulsive seizures in PWECP during pregnancy to minimize potential risks to the birth parent and to the fetus. Once a PWECP is already pregnant, clinicians should exercise caution in attempting to remove or replace an ASM that is effective in controlling generalized tonic-clonic or focal-to-bilateral tonic-clonic seizures. Clinicians must consider using lamotrigine, levetiracetam, or oxcarbazepine in PWECP when appropriate based on the patient's epilepsy syndrome, likelihood of achieving seizure control, and comorbidities, to minimize the risk of MCMs. Clinicians must avoid the use of valproic acid in PWECP to minimize the risk of MCMs or neural tube defects (NTDs), if clinically feasible. Clinicians should avoid the use of valproic acid or topiramate in PWECP to minimize the risk of offspring being born small for gestational age, if clinically feasible. To reduce the risk of poor neurodevelopmental outcomes, including autism spectrum disorder and lower IQ, in children born to PWECP, clinicians must avoid the use of valproic acid in PWECP, if clinically feasible. Clinicians should prescribe at least 0.4 mg of folic acid supplementation daily preconceptionally and during pregnancy to any PWECP treated with an ASM to decrease the risk of NTDs and possibly improve neurodevelopmental outcomes in the offspring.
Collapse
|
7
|
Kaiser J, Risteska A, Muller AG, Sun H, Lei B, Nay K, Means AR, Cousin MA, Drewry DH, Oakhill JS, Kemp BE, Hannan AJ, Berk M, Febbraio MA, Gundlach AL, Hill-Yardin EL, Scott JW. Convergence on CaMK4: A Key Modulator of Autism-Associated Signaling Pathways in Neurons. Biol Psychiatry 2025; 97:439-449. [PMID: 39442785 DOI: 10.1016/j.biopsych.2024.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/03/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024]
Abstract
Although the precise underlying cause(s) of autism spectrum disorder remain unclear, more than 1000 rare genetic variations are associated with the condition. For many people living with profound autism, this genetic heterogeneity has impeded the identification of common biological targets for therapy development for core and comorbid traits that include significant impairments in social communication and repetitive and restricted behaviors. A substantial number of genes associated with autism encode proteins involved in signal transduction and synaptic transmission that are critical for brain development and function. CAMK4 is an emerging risk gene for autism spectrum disorder that encodes the CaMK4 (calcium/calmodulin-dependent protein kinase 4) enzyme. CaMK4 is a key component of a Ca2+-activated signaling pathway that regulates neurodevelopment and synaptic plasticity. In this review, we discuss 3 genetic variants of CAMK4 found in individuals with hyperkinetic movement disorder and comorbid neurological symptoms including autism spectrum disorder that are likely pathogenic with monogenic effect. We also comment on 4 other genetic variations in CAMK4 that show associations with autism spectrum disorder, as well as 12 examples of autism-associated variations in other genes that impact CaMK4 signaling pathways. Finally, we highlight 3 environmental risk factors that impact CaMK4 signaling based on studies of preclinical models of autism and/or clinical cohorts. Overall, we review molecular, genetic, physiological, and environmental evidence that suggest that defects in the CaMK4 signaling pathway may play an important role in a common autism pathogenesis network across numerous patient groups, and we propose CaMK4 as a potential therapeutic target.
Collapse
Affiliation(s)
- Jacqueline Kaiser
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia; St. Vincent's Institute of Medical Research, Melbourne, Victoria, Australia; Mary McKillop Institute for Health Research, Australian Catholic University, Melbourne, Victoria, Australia
| | - Alana Risteska
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia
| | - Abbey G Muller
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia; Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia
| | - Haoxiong Sun
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia
| | - Bethany Lei
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia
| | - Kevin Nay
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia
| | - Anthony R Means
- Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Margot A Cousin
- Center for Individualized Medicine, College of Medicine, Mayo Clinic, Rochester, Minnesota
| | - David H Drewry
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, the University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jonathan S Oakhill
- St. Vincent's Institute of Medical Research, Melbourne, Victoria, Australia; Mary McKillop Institute for Health Research, Australian Catholic University, Melbourne, Victoria, Australia
| | - Bruce E Kemp
- St. Vincent's Institute of Medical Research, Melbourne, Victoria, Australia
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, Melbourne, Victoria, Australia; Department of Anatomy and Physiology, the University of Melbourne, Melbourne, Victoria, Australia
| | - Michael Berk
- Florey Institute of Neuroscience and Mental Health, Melbourne, Victoria, Australia; The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, Parkville, Melbourne, Australia
| | - Mark A Febbraio
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia
| | - Andrew L Gundlach
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia; St. Vincent's Institute of Medical Research, Melbourne, Victoria, Australia; Florey Institute of Neuroscience and Mental Health, Melbourne, Victoria, Australia; Department of Anatomy and Physiology, the University of Melbourne, Melbourne, Victoria, Australia
| | - Elisa L Hill-Yardin
- Department of Anatomy and Physiology, the University of Melbourne, Melbourne, Victoria, Australia; School of Health and Biomedical Sciences, STEM College, RMIT University, Melbourne, Victoria, Australia.
| | - John W Scott
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia; St. Vincent's Institute of Medical Research, Melbourne, Victoria, Australia; Florey Institute of Neuroscience and Mental Health, Melbourne, Victoria, Australia.
| |
Collapse
|
8
|
Bhaskaran D, Thomas S, Scaria LM. Neurobehavioral Outcomes of Children with Antenatal Exposure to Antiseizure Medications. Indian J Pediatr 2025; 92:252-259. [PMID: 38850358 DOI: 10.1007/s12098-024-05165-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 05/13/2024] [Indexed: 06/10/2024]
Abstract
OBJECTIVES To evaluate the effect of antiepileptic medications prescribed to mothers during pregnancy on the development and behavior of children. METHODS From the Kerala Registry of Epilepsy and Pregnancy, 98 children between the ages of 1½ to 2½ y were consecutively chosen. Children of mothers who did not have epilepsy during pregnancy and not exposed to antiseizure medications (ASMs) antenatally were selected as comparator group. Developmental assessment of the children was performed using Developmental Assessment Scale for Indian Infants (DASII) and Receptive-Expressive Emergent Language Scale (REELS). Behavior outcomes were assessed using Child Behavior Checklist. RESULTS A significant delay in expressive language skills was seen in children exposed to antiseizure medication with an odds ratio of 2.539 (95% CI 1.10, 5.85, P = 0.026). A delay in expressive language skills was seen in polytherapy with clobazam (odds ratio 6.83; 95% CI 2.17, 21.56, P < 0.001). Also, delay was seen in receptive language skills in the same polytherapy group (odds ratio of 7.333; 95% CI 2.16, 24.92, P < 0.001). There were no statistically significant differences between study and comparative groups in motor and mental quotient domains and behavioral outcomes. CONCLUSIONS The finding of speech delay in children exposed to ASMs is significant since individuals with a history of childhood speech or language disorders may experience long-term difficulties in mental health, social well-being, and academic outcomes.
Collapse
Affiliation(s)
- Deepa Bhaskaran
- Department of Developmental Pediatrics, Child Development Centre, Thiruvananthapuram Medical College, Thiruvananthapuram, 695 011, Kerala, India.
| | - Sanjeev Thomas
- Institute for Communicative and Cognitive Neurosciences (ICCONS), Thiruvananthapuram, Kerala, India
| | - Liss Maria Scaria
- Child Development Centre, Thiruvananthapuram Medical College, Thiruvananthapuram, Kerala, India
| |
Collapse
|
9
|
Matsushima T, Toji N, Wada K, Shikanai H, Izumi T. Embryonic exposure to valproic acid and neonicotinoid deteriorates the hyperpolarizing GABA shift and impairs long-term potentiation of excitatory transmission in the local circuit of intermediate medial mesopallium of chick telencephalon. Cereb Cortex 2025; 35:bhaf044. [PMID: 40037548 DOI: 10.1093/cercor/bhaf044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/20/2025] [Accepted: 01/22/2025] [Indexed: 03/06/2025] Open
Abstract
Embryonic exposure to valproic acid and imidacloprid (a neonicotinoid insecticide) impairs filial imprinting in hatchlings, and the deteriorating effects of valproic acid are mitigated by post-hatch injection of bumetanide, a blocker of the chloride intruder Na-K-2Cl cotransporter 1. Here, we report that these exposures depolarized the reversal potential of local GABAergic transmission in the neurons of the intermediate medial mesopallium, the pallial region critical for imprinting. Furthermore, exposure increased field excitatory post-synaptic potentials in pre-tetanus recordings and impaired long-term potentiation (LTP) by low-frequency tetanic stimulation. Bath-applied bumetanide rescued the impaired LTP in the valproic acid slices, whereas VU0463271, a blocker of the chloride extruder KCC2, suppressed LTP in the control slices, suggesting that hyperpolarizing GABA action is necessary for the potentiation of excitatory synaptic transmission. Whereas a steep increase in the gene expression of KCC2 appeared compared to NKCC1 during the peri-hatch development, significant differences were not found between valproic acid and control post-hatch chicks in these genes. Instead, both valproic acid and imidacloprid downregulated several transcriptional regulators (FOS, NR4A1, and NR4A2) and upregulated the RNA component of signal recognition particles (RN7SL1). Despite different chemical actions, valproic acid and imidacloprid could cause common neuronal effects that lead to impaired imprinting.
Collapse
Affiliation(s)
- Toshiya Matsushima
- Department of Biology, Faculty of Science, Hokkaido University, N10 W8 Kita-ku, 060-0810 Sapporo, Japan
- Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Kanazawa 1757, 061-0293 Tobetsu, Japan
- Center for Mind/Brain Sciences, University of Trento, Piazza Manifattura 1, 38068 Rovereto, Italy
| | - Noriyuki Toji
- Department of Biology, Faculty of Science, Hokkaido University, N10 W8 Kita-ku, 060-0810 Sapporo, Japan
| | - Kazuhiro Wada
- Department of Biology, Faculty of Science, Hokkaido University, N10 W8 Kita-ku, 060-0810 Sapporo, Japan
| | - Hiroki Shikanai
- Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Kanazawa 1757, 061-0293 Tobetsu, Japan
| | - Takeshi Izumi
- Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Kanazawa 1757, 061-0293 Tobetsu, Japan
| |
Collapse
|
10
|
Williams OOF, Coppolino M, Micelli CB, McCallum RT, Henry-Duru PT, Manduca JD, Lalonde J, Perreault ML. Prenatal exposure to valproic acid induces sex-specific alterations in rat cortical and hippocampal neuronal structure and function in vitro. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111222. [PMID: 39701172 DOI: 10.1016/j.pnpbp.2024.111222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/10/2024] [Accepted: 12/12/2024] [Indexed: 12/21/2024]
Abstract
There are substantial differences in the characteristics of males and females with an autism spectrum disorder (ASD), yet there is little knowledge surrounding the mechanistic underpinnings of these differences. The valproic acid (VPA) rodent model is based upon the human fetal valproate spectrum disorder, which is associated with increased risk of developing ASD. This model, which displays significant social, learning, and memory alterations, has therefore been widely used to further our understanding of specific biological features of ASD. However, to date, almost all of the studies employing this model have used male rodents. To fill this knowledge gap, we evaluated sex differences for neuronal activity, morphology, and glycogen synthase kinase-3 (GSK-3) signaling in primary cortical (CTX) and hippocampal (HIP) neurons prepared from rats exposed to VPA in utero. In vivo, sex-specific VPA-induced alterations in the frontal CTX transcriptome at birth were also determined. Overall, VPA induced more robust changes in neuronal function and structure in the CTX than in the HIP. Male- and female-derived primary CTX neurons from rats exposed to prenatal VPA had elevated activity and showed more disorganized firing. In the HIP, only the female VPA neurons showed elevated firing, while the male VPA neurons exhibited disorganized activity. Dendritic arborization of CTX neurons from VPA rats was less complex in both sexes, though this was more pronounced in the females. Conversely, both female and male HIP neurons from VPA rats showed elevated complexity distal to the soma. Female VPA CTX neurons also had an elevated number of dendritic spines. The relative activity of the α and β isoforms of GSK-3 were suppressed in both female and male VPA CTX neurons, with no changes in the HIP neurons. On postnatal day 0, alterations in CTX genes associated with neuropeptides (e.g., penk, pdyn) and receptors (e.g., drd1, adora2a) were seen in both sexes, though they were downregulated in females and upregulated in males. Together these findings suggest that substantial sex differences in neuronal structure and function in the VPA model may have relevance to the reported sex differences in idiopathic ASD.
Collapse
Affiliation(s)
- Olivia O F Williams
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, Ontario N1G 2W1, Canada.
| | - Madeleine Coppolino
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, Ontario N1G 2W1, Canada
| | - Cecilia B Micelli
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, Ontario N1G 2W1, Canada
| | - Ryan T McCallum
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, Ontario N1G 2W1, Canada
| | - Paula T Henry-Duru
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, Ontario N1G 2W1, Canada.
| | - Joshua D Manduca
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, Ontario N1G 2W1, Canada.
| | - Jasmin Lalonde
- Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Rd. E., Guelph, Ontario N1G 2W1, Canada.
| | - Melissa L Perreault
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, Ontario N1G 2W1, Canada.
| |
Collapse
|
11
|
He C, Zhou H, Chen L, Liu Z. NEAT1 Promotes Valproic Acid-Induced Autism Spectrum Disorder by Recruiting YY1 to Regulate UBE3A Transcription. Mol Neurobiol 2025; 62:846-860. [PMID: 38922486 DOI: 10.1007/s12035-024-04309-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 06/13/2024] [Indexed: 06/27/2024]
Abstract
Evidence suggests that long non-coding RNAs (lncRNAs) play a significant role in autism. Herein, we explored the functional role and possible molecular mechanisms of NEAT1 in valproic acid (VPA)-induced autism spectrum disorder (ASD). A VPA-induced ASD rat model was constructed, and a series of behavioral tests were performed to examine motor coordination and learning-memory abilities. qRT-PCR and western blot assays were used to evaluate target gene expression levels. Loss-and-gain-of-function assays were conducted to explore the functional role of NEAT1 in ASD development. Furthermore, a combination of mechanistic experiments and bioinformatic tools was used to assess the relationship and regulatory role of the NEAT1-YY1-UBE3A axis in ASD cellular processes. Results showed that VPA exposure induced autism-like developmental delays and behavioral abnormalities in the VPA-induced ASD rat model. We found that NEAT1 was elevated in rat hippocampal tissues after VPA exposure. NEAT1 promoted VPA-induced autism-like behaviors and mitigated apoptosis, oxidative stress, and inflammation in VPA-induced ASD rats. Notably, NEAT1 knockdown improved autism-related behaviors and ameliorated hippocampal neuronal damage. Mechanistically, it was observed that NEAT1 recruited the transcription factor YY1 to regulate UBE3A expression. Additionally, in vitro experiments further confirmed that NEAT1 knockdown mitigated hippocampal neuronal damage, oxidative stress, and inflammation through the YY1/UBE3A axis. In conclusion, our study demonstrates that NEAT1 is highly expressed in ASD, and its inhibition prominently suppresses hippocampal neuronal injury and oxidative stress through the YY1/UBE3A axis, thereby alleviating ASD development. This provides a new direction for ASD-targeted therapy.
Collapse
Affiliation(s)
- Chuping He
- Department of Children's Health, Chenzhou First People's Hospital, No. 6, Feihong Road, Suxian District, Chenzhou, 423000, Hunan, China
| | - Huimei Zhou
- Department of Children's Health, Chenzhou First People's Hospital, No. 6, Feihong Road, Suxian District, Chenzhou, 423000, Hunan, China.
| | - Lei Chen
- Department of Children's Health, Chenzhou First People's Hospital, No. 6, Feihong Road, Suxian District, Chenzhou, 423000, Hunan, China
| | - Zeying Liu
- Department of Children's Health, Chenzhou First People's Hospital, No. 6, Feihong Road, Suxian District, Chenzhou, 423000, Hunan, China
| |
Collapse
|
12
|
Ohnami S, Naito M, Kawase H, Higuchi M, Hasebe S, Takasu K, Kanemaru R, Azuma Y, Yokoyama R, Kochi T, Imado E, Tahara T, Kotake Y, Asano S, Oishi N, Takuma K, Hashimoto H, Ogawa K, Nakamura A, Yamakawa H, Ago Y. Brain region-specific neural activation by low-dose opioid promotes social behavior. JCI Insight 2024; 9:e182060. [PMID: 39641273 PMCID: PMC11623950 DOI: 10.1172/jci.insight.182060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 10/22/2024] [Indexed: 12/07/2024] Open
Abstract
The opioid system plays crucial roles in modulating social behaviors in both humans and animals. However, the pharmacological profiles of opioids regarding social behavior and their therapeutic potential remain unclear. Multiple pharmacological, behavioral, and immunohistological c-Fos mapping approaches were used to characterize the effects of μ-opioid receptor agonists on social behavior and investigate the mechanisms in naive mice and autism spectrum disorder-like (ASD-like) mouse models, such as prenatally valproic acid-treated mice and Fmr1-KO mice. Here, we report that low-dose morphine, a μ-opioid receptor agonist, promoted social behavior by selectively activating neurons in prosocial brain regions, including the nucleus accumbens, but not those in the dorsomedial periaqueductal gray (dmPAG), which are only activated by analgesic high-dose morphine. Critically, intra-dmPAG morphine injection counteracted the prosocial effect of low-dose morphine, suggesting that dmPAG neural activation suppresses social behavior. Moreover, buprenorphine, a μ-opioid receptor partial agonist with less abuse liability and a well-established safety profile, ameliorated social behavior deficits in two mouse models recapitulating ASD symptoms by selectively activating prosocial brain regions without dmPAG neural activation. Our findings highlight the therapeutic potential of brain region-specific neural activation induced by low-dose opioids for social behavior deficits in ASD.
Collapse
MESH Headings
- Animals
- Mice
- Social Behavior
- Morphine/pharmacology
- Morphine/administration & dosage
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/metabolism
- Analgesics, Opioid/pharmacology
- Analgesics, Opioid/administration & dosage
- Male
- Mice, Knockout
- Periaqueductal Gray/drug effects
- Periaqueductal Gray/metabolism
- Disease Models, Animal
- Female
- Nucleus Accumbens/drug effects
- Nucleus Accumbens/metabolism
- Neurons/drug effects
- Neurons/metabolism
- Buprenorphine/pharmacology
- Buprenorphine/administration & dosage
- Autism Spectrum Disorder/drug therapy
- Fragile X Mental Retardation Protein/genetics
- Fragile X Mental Retardation Protein/metabolism
- Brain/drug effects
- Brain/metabolism
- Behavior, Animal/drug effects
- Valproic Acid/pharmacology
- Valproic Acid/administration & dosage
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Soichiro Ohnami
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co. Ltd., Toyonaka, Osaka, Japan
- SK Project, Medical Innovation Center, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan
| | - Megumi Naito
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, and
| | - Haruki Kawase
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, and
| | - Momoko Higuchi
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, and
| | - Shigeru Hasebe
- Department of Pharmacology, Graduate School of Dentistry, Osaka University, Suita, Osaka, Japan
- Department of Social Pharmacy, Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata, Osaka, Japan
| | - Keiko Takasu
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co. Ltd., Toyonaka, Osaka, Japan
| | - Ryo Kanemaru
- Shionogi TechnoAdvance Research Co. Ltd., Toyonaka, Osaka, Japan
| | - Yuki Azuma
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co. Ltd., Toyonaka, Osaka, Japan
| | - Rei Yokoyama
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, and
- Department of Cellular and Molecular Pharmacology
| | | | - Eiji Imado
- Department of Cellular and Molecular Pharmacology
- Department of Dental Anesthesiology, and
| | - Takeru Tahara
- Department of Neurochemistry and Environmental Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima, Japan
| | - Yaichiro Kotake
- Department of Neurochemistry and Environmental Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima, Japan
| | | | - Naoya Oishi
- SK Project, Medical Innovation Center, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan
- Department of Psychiatry, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan
| | - Kazuhiro Takuma
- Department of Pharmacology, Graduate School of Dentistry, Osaka University, Suita, Osaka, Japan
- Molecular Research Center for Children’s Mental Development, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University, and University of Fukui, Suita, Osaka, Japan
| | - Hitoshi Hashimoto
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, and
- Molecular Research Center for Children’s Mental Development, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University, and University of Fukui, Suita, Osaka, Japan
- Division of Bioscience, Institute for Datability Science
- Open and Transdisciplinary Research Initiatives, and
- Department of Molecular Pharmaceutical Sciences, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Koichi Ogawa
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co. Ltd., Toyonaka, Osaka, Japan
- SK Project, Medical Innovation Center, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan
| | - Atsushi Nakamura
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co. Ltd., Toyonaka, Osaka, Japan
- Shionogi TechnoAdvance Research Co. Ltd., Toyonaka, Osaka, Japan
| | - Hidekuni Yamakawa
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co. Ltd., Toyonaka, Osaka, Japan
- SK Project, Medical Innovation Center, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan
- Ping An-Shionogi Co. Ltd., Xuhui District, Shanghai, China
| | - Yukio Ago
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, and
- Department of Cellular and Molecular Pharmacology
- Laboratory of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, and
- Global Center for Medical Engineering and Informatics, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
13
|
Wang L, Clark EA, Hanratty L, Koblan KS, Foley A, Dedic N, Bristow LJ. TAAR1 and 5-HT 1B receptor agonists attenuate autism-like irritability and aggression in rats prenatally exposed to valproic acid. Pharmacol Biochem Behav 2024; 245:173862. [PMID: 39197535 DOI: 10.1016/j.pbb.2024.173862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/02/2024] [Accepted: 08/23/2024] [Indexed: 09/01/2024]
Abstract
Despite the rising prevalence of autism spectrum disorder (ASD), there remains a significant unmet need for pharmacotherapies addressing its core and associative symptoms. While some atypical antipsychotics have been approved for managing associated irritability and aggression, their use is constrained by substantial side effects. This study aimed firstly to develop behavioral measures to explore frustration, irritability and aggression phenotypes in the rat prenatal valproic acid (VPA) model of ASD. Additionally, we investigated the potential of two novel mechanisms, 5-HT1B and TAAR1 agonism, to alleviate these behaviors. Male offspring exposed to prenatal VPA were trained to achieve stable performance on a cued operant task, followed by pharmacological assessment in an operant frustration test, bottle brush test and resident intruder test. VPA exposed rats demonstrated behaviors indicative of frustration and irritability, as well as increased aggression compared to controls. The irritability-like behavior and aggression were further exacerbated in animals previously experiencing a frustrative event during the operant test. Single administration of the 5-HT1B agonist CP-94253 or TAAR1 agonist RO5263397 attenuated the frustration-like behavior compared to vehicle. Additionally, both agonists reduced irritability-like behavior under both normal and frustrative conditions. While CP-94253 reduced aggression in the resident intruder test under both conditions, RO5263397 only produced effects in rats that previously experienced a frustrative event. Our study describes previously uncharacterized phenotypes of frustration, irritability, and aggression in the rat prenatal VPA model of ASD. Administration of selective TAAR1 or 5-HT1B receptor agonists alleviated these deficits, warranting further exploration of both targets in ASD treatment.
Collapse
Affiliation(s)
- Lien Wang
- Sumitomo Pharma America, Inc., Marlborough, MA, USA
| | - Erin A Clark
- Sumitomo Pharma America, Inc., Marlborough, MA, USA
| | | | | | | | - Nina Dedic
- Sumitomo Pharma America, Inc., Marlborough, MA, USA.
| | | |
Collapse
|
14
|
Madley-Dowd P, Ahlqvist VH, Forbes H, Rast JE, Martin FZ, Zhong C, Barry CJS, Berglind D, Lundberg M, Lyall K, Newschaffer CJ, Tomson T, Davies NM, Magnusson C, Rai D, Lee BK. Antiseizure medication use during pregnancy and children's neurodevelopmental outcomes. Nat Commun 2024; 15:9640. [PMID: 39548057 PMCID: PMC11568279 DOI: 10.1038/s41467-024-53813-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 10/22/2024] [Indexed: 11/17/2024] Open
Abstract
The teratogenic potential of valproate in pregnancy is well established; however, evidence regarding the long-term safety of other antiseizure medications (ASMs) during pregnancy remains limited. Using routinely collected primary care data from the UK and nationwide Swedish registries to create a cohort of 3,182,773 children, of which 17,495 were exposed to ASMs in pregnancy, we show that those exposed to valproate were more likely to receive a diagnosis of autism, intellectual disability, and ADHD, when compared to children not exposed to ASMs. Additionally, children exposed to topiramate were 2.5 times more likely to be diagnosed with intellectual disability (95% CI: 1.23-4.98), and those exposed to carbamazepine were 1.25 times more likely to be diagnosed with autism (95% CI: 1.05-1.48) and 1.30 times more likely to be diagnosed with intellectual disability (95% CI: 1.01-1.69). There was little evidence that children exposed to lamotrigine in pregnancy were more likely to receive neurodevelopmental diagnoses. While further research is needed, these findings may support considering safer treatment alternatives well before conception when clinically appropriate.
Collapse
Affiliation(s)
- Paul Madley-Dowd
- Centre for Academic Mental Health, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK.
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, Bristol, UK.
- NIHR Biomedical Research Centre, University of Bristol, Bristol, UK.
| | - Viktor H Ahlqvist
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
| | - Harriet Forbes
- Centre for Academic Mental Health, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, UK
| | - Jessica E Rast
- A.J. Drexel Autism Institute, Drexel University, Philadelphia, PA, USA
- Department of Epidemiology and Biostatistics, Drexel University Dornsife School of Public Health, Philadelphia, PA, USA
| | - Florence Z Martin
- Centre for Academic Mental Health, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
| | - Caichen Zhong
- A.J. Drexel Autism Institute, Drexel University, Philadelphia, PA, USA
| | - Ciarrah-Jane S Barry
- Centre for Academic Mental Health, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
| | - Daniel Berglind
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
- Centre for Epidemiology and Community Medicine, Region Stockholm, Stockholm, Sweden
| | - Michael Lundberg
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
| | - Kristen Lyall
- Department of Epidemiology and Biostatistics, Drexel University Dornsife School of Public Health, Philadelphia, PA, USA
| | - Craig J Newschaffer
- College of Health and Human Development, The Pennsylvania State University, Philadelphia, PA, USA
| | - Torbjörn Tomson
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Neil M Davies
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
- Division of Psychiatry, University College London, London, UK
- Department of Statistical Science, University College London, London, UK
| | - Cecilia Magnusson
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
- Centre for Epidemiology and Community Medicine, Region Stockholm, Stockholm, Sweden
| | - Dheeraj Rai
- Centre for Academic Mental Health, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- NIHR Biomedical Research Centre, University of Bristol, Bristol, UK
- Avon and Wiltshire Partnership NHS Mental Health Trust, Bristol, UK
| | - Brian K Lee
- A.J. Drexel Autism Institute, Drexel University, Philadelphia, PA, USA
- Department of Epidemiology and Biostatistics, Drexel University Dornsife School of Public Health, Philadelphia, PA, USA
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
15
|
Graeca M, Kulesza R. Impaired brainstem auditory evoked potentials after in utero exposure to high dose paracetamol exposure. Hear Res 2024; 454:109149. [PMID: 39550993 DOI: 10.1016/j.heares.2024.109149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 10/20/2024] [Accepted: 11/06/2024] [Indexed: 11/19/2024]
Abstract
Paracetamol is an analgesic and antipyretic medication regarded as the safest over-the-counter pain and fever relief option during pregnancy. Paracetamol and its metabolites are known to reach the developing fetus through direct placental transfer and can cross the blood brain barrier. Several recent, large-scale epidemiologic studies suggest that in utero paracetamol exposure can increase the risk of neurodevelopmental conditions, including autism spectrum disorder (ASD), attention deficit hyperactivity disorder (ADHD) and developmental delay (DD). Since auditory processing deficits are a common feature of ASD, we hypothesized that animals exposed to paracetamol in utero will have impaired auditory brainstem function. We investigated this hypothesis by recording and analyzing click-evoked auditory brainstem responses (ABR) at postnatal day 21 and 29 in Sprague-Dawley rats. In utero exposure to high dose paracetamol exposure had no impact on body or brain weight. However, high dose paracetamol exposure did significantly delay ear opening and resulted in elevated ABR thresholds, and longer wave and interwave latencies. These changes in wave latency extended to the highest click intensity tested but were most severe near threshold. This data suggests that development and function of the auditory brainstem may be impacted by high dose paracetamol exposure and that simple, non-invasive tests of auditory function have utility as an early screening tool for neurodevelopmental disorders.
Collapse
Affiliation(s)
- Meghan Graeca
- Department of Anatomy, Lake Erie College of Osteopathic Medicine, Erie, PA, United States
| | - Randy Kulesza
- Department of Anatomy, Lake Erie College of Osteopathic Medicine, Erie, PA, United States.
| |
Collapse
|
16
|
Yoshida F, Nagatomo R, Utsunomiya S, Kimura M, Shun S, Kono R, Kato Y, Nao Y, Maeda K, Koyama R, Ikegaya Y, Lichtenthaler SF, Takatori S, Takemoto H, Ogawa K, Ito G, Tomita T. Soluble form of Lingo2, an autism spectrum disorder-associated molecule, functions as an excitatory synapse organizer in neurons. Transl Psychiatry 2024; 14:448. [PMID: 39443477 PMCID: PMC11500186 DOI: 10.1038/s41398-024-03167-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 10/13/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024] Open
Abstract
Autism Spectrum Disorder (ASD) is a developmental disorder characterized by impaired social communication and repetitive behaviors. In recent years, a pharmacological mouse model of ASD involving maternal administration of valproic acid (VPA) has become widely used. Newborn pups in this model show an abnormal balance between excitatory and inhibitory (E/I) signaling in neurons and exhibit ASD-like behavior. However, the molecular basis of this model and its implications for the pathogenesis of ASD in humans remain unknown. Using quantitative secretome analysis, we found that the level of leucine-rich repeat and immunoglobulin domain-containing protein 2 (Lingo2) was upregulated in the conditioned medium of VPA model neurons. This upregulation was associated with excitatory synaptic organizer activity. The secreted form of the extracellular domain of Lingo2 (sLingo2) is produced by the transmembrane metalloprotease ADAM10 through proteolytic processing. sLingo2 was found to induce the formation of excitatory synapses in both mouse and human neurons, and treatment with sLingo2 resulted in an increased frequency of miniature excitatory postsynaptic currents in human neurons. These findings suggest that sLingo2 is an excitatory synapse organizer involved in ASD, and further understanding of the mechanisms by which sLingo2 induces excitatory synaptogenesis is expected to advance our understanding of the pathogenesis of ASD.
Collapse
Affiliation(s)
- Fumiaki Yoshida
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Ryota Nagatomo
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Shun Utsunomiya
- Neuroscience 2, Laboratory for Drug Discovery and Disease Research, Shionogi & Co. Ltd, Osaka, Japan
| | - Misaki Kimura
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Shiyori Shun
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Rena Kono
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yuma Kato
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yosuke Nao
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Kazuma Maeda
- Neuroscience 2, Laboratory for Drug Discovery and Disease Research, Shionogi & Co. Ltd, Osaka, Japan
| | - Ryuta Koyama
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
- Institute for AI and Beyond, The University of Tokyo, Tokyo, Japan
- 5Department of Translational Neurobiology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Yuji Ikegaya
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
- Institute for AI and Beyond, The University of Tokyo, Tokyo, Japan
- Center for Information and Neural Networks, National Institute of Information and Communications Technology, Osaka, Japan
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Sho Takatori
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Takemoto
- Neuroscience 2, Laboratory for Drug Discovery and Disease Research, Shionogi & Co. Ltd, Osaka, Japan
- Business-Academia Collaborative Laboratory (Shionogi), Graduate School of Pharmaceutical Science, The University of Tokyo, Tokyo, Japan
| | - Koichi Ogawa
- Neuroscience 2, Laboratory for Drug Discovery and Disease Research, Shionogi & Co. Ltd, Osaka, Japan
| | - Genta Ito
- Department of Biomolecular Chemistry, Faculty of Pharma-Science, Teikyo University, Tokyo, Japan
- Social Cooperation Program of Brain and Neurological Disorders, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Taisuke Tomita
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan.
- Social Cooperation Program of Brain and Neurological Disorders, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
17
|
Mansour Y, Kulesza R. Obliteration of a glycinergic projection to the medial geniculate in an animal model of autism. Front Cell Neurosci 2024; 18:1465255. [PMID: 39484183 PMCID: PMC11524938 DOI: 10.3389/fncel.2024.1465255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/07/2024] [Indexed: 11/03/2024] Open
Abstract
Auditory dysfunction affects the vast majority of people with autism spectrum disorder (ASD) and can range from deafness to hypersensitivity. In utero exposure to the antiepileptic valproic acid (VPA) is associated with significant risk of an ASD diagnosis in humans and timed in utero exposure to VPA is utilized as an animal model of ASD. VPA-exposed rats have significantly fewer neurons in their auditory brainstem, thalamus and cortex, reduced ascending projections to the midbrain and thalamus and reduced descending projections from the cortex to the auditory midbrain. Consistent with these anatomical changes, VPA-exposed animals also have abnormal auditory brainstem responses. We have recently described a significant ascending projection from calbindin-positive neurons in the medial nucleus of the trapezoid body (MNTB) to the ventral division of the medial geniculate (vMG) in rats that bypasses the central nucleus of the inferior colliculus (CNIC). Since we found that axonal projections to the vMG in VPA-exposed rats are reduced beyond what is predicted from neuron loss alone, we hypothesize that VPA exposure would result in a significant reduction in the MNTB projection to the vMG. We examined this hypothesis by quantifying the proportion of retrogradely-labeled neurons in the MNTB of control and VPA-exposed animals after injections of retrograde tracers in the CNIC and vMG in control and VPA-exposed animals. Our results indicate that in control animals, the MNTB forms the largest projection from the superior olivary complex to the MG and that this projection is nearly abolished by in utero VPA exposure.
Collapse
Affiliation(s)
- Yusra Mansour
- Department of Otolaryngology—Head and Neck Surgery, Detroit, MI, United States
| | - Randy Kulesza
- Department of Anatomy, Lake Erie College of Osteopathic Medicine, Erie, PA, United States
| |
Collapse
|
18
|
Mbizvo GK, Bucci T, Lip GYH, Marson AG. Morbidity and mortality risks associated with valproate withdrawal in young adults with epilepsy. Brain 2024; 147:3426-3441. [PMID: 38657115 PMCID: PMC11449131 DOI: 10.1093/brain/awae128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 02/19/2024] [Accepted: 03/13/2024] [Indexed: 04/26/2024] Open
Abstract
Valproate is the most effective treatment for idiopathic generalized epilepsy. Current guidance precludes its use in females of childbearing potential, unless other treatments are ineffective or not tolerated, because of high teratogenicity. This risk was recently extended to males. New guidance will limit use both in males and females aged <55 years, resulting in withdrawal of valproate from males already taking it, as occurs for females. Whether there are risks of personal harm (including injury or death) associated with valproate withdrawal has not yet been quantified for males or females ON valproate, meaning clinicians cannot reliably counsel either sex when discussing valproate withdrawal with them, despite that this concern may be at the forefront of patients' and clinicians' minds. We assessed whether there are any morbidity or mortality risks associated with valproate withdrawal in young males and females. We performed a retrospective cohort study of internationally derived electronic health data within the TriNetX Global Collaborative Network. Included were males and females aged 16-54 years with ≥1 epilepsy disease or symptom code between 1 December 2017 and 1 December 2018, and ≥2 valproate prescriptions over the preceding 2 years (1 January 2015-30 November 2017). Five-year propensity-matched risks of mortality and a range of morbidity outcomes were compared between those remaining ON versus withdrawn from valproate during the 1 December 2017-1 December 2018 recruitment period, regardless of whether switched to another antiseizure medication. Survival analysis was undertaken using Cox-proportional hazard models, generating hazard ratios (HRs) with 95% confidence intervals (CIs). In total, 8991 males and 5243 females taking valproate were recruited. Twenty-eight per cent of males and 36% of females were subsequently withdrawn from valproate. Valproate withdrawal was associated with significantly increased risks of emergency department attendance [HRs overall: 1.236 (CI 1.159-1.319), males: 1.181 (CI 1.083-1.288), females: 1.242 (CI 1.125-1.371)], hospital admission [HRs overall: 1.160 (CI 1.081-1.246), males: 1.132 (CI 1.027-1.249), females: 1.147 (CI 1.033-1.274)], falls [HRs overall: 1.179 (CI 1.041-1.336), males: 1.298 (CI 1.090-1.546)], injuries [HRs overall: 1.095 (CI 1.021-1.174), males: 1.129 (CI 1.029-1.239)], burns [HRs overall: 1.592 (CI 1.084-2.337)] and new-onset depression [HRs overall 1.323 (CI 1.119-1.565), females: 1.359 (CI 1.074-1.720)]. The risk of these outcomes occurring was 1%-7% higher in those withdrawn from valproate than in those remaining ON valproate. Overall, valproate withdrawal was not associated with increased mortality. These results may help patients and clinicians have a more informed discussion about personal safety when considering valproate withdrawal.
Collapse
Affiliation(s)
- Gashirai K Mbizvo
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart and Chest Hospital, Liverpool, L69 7TX, UK
- Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L69 7BE, UK
- Neurology Service, The Walton Centre NHS Foundation Trust, Liverpool, L9 7LJ, UK
| | - Tommaso Bucci
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart and Chest Hospital, Liverpool, L69 7TX, UK
- Department of General and Specialized Surgery, Sapienza University of Rome, Rome, 00185, Italy
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart and Chest Hospital, Liverpool, L69 7TX, UK
- Department of Clinical Medicine, Danish Centre for Health Services Research, Aalborg University, Aalborg, 9220, Denmark
| | - Anthony G Marson
- Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L69 7BE, UK
- Neurology Service, The Walton Centre NHS Foundation Trust, Liverpool, L9 7LJ, UK
| |
Collapse
|
19
|
Nyakato M, Naggayi SK, Akun PR, Ononge S, Odong A, Baguma EA, Nansiiro H, Kalibbala D, Ouma S, Besigye I, Idro R. Poor neurodevelopment, nutritional and physical growth outcomes among children born to mothers with nodding syndrome. Seizure 2024:S1059-1311(24)00260-7. [PMID: 39343705 DOI: 10.1016/j.seizure.2024.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024] Open
Abstract
INTRODUCTION Nodding syndrome (NS), a poorly understood severe neurological disorder develops in children. In Uganda, some NS cases have grown into child-bearing adults. Babies born to mothers with NS may be prone to impaired neurodevelopmental outcomes. Cognitive deficits in mothers with NS may further inhibit care offered to their children hence compromising neurocognitive development, physical growth, and behaviour. OBJECTIVES The study aimed to determine the neurodevelopmental, behavioural, nutritional, and physical growth outcomes of children whose mothers have nodding syndrome. METHODS A comparative cross-sectional study was conducted between May 2021 and April 2022 in Northern Uganda. Children aged 0-5 years of mothers with NS were compared to those of mothers without NS, matched by age, gender and neighbouring residence. Neurodevelopment, behaviour, nutrition, and physical growth were assessed using standardized measures and t-tests employed for group comparisons of outcomes. RESULTS Overall,106 children participated. Fifty-three (53) were offspring of mothers with NS and 53 of mothers without NS; having a mean age of 26.9 (2.22) and 27.5(2.12) months respectively. Children whose mothers have NS had significantly lower neurodevelopmental scores than those of NS-unaffected mothers in fine motor (37.5(12.1) vs 44.2(14.3), p = 0.011), receptive language (37.8(10.8) vs 43.9(12.9), p = 0.010), overall cognitive development (74.36(17.8) vs 83.34(19.6), p = 0.015), and attention (0.64(0.20) vs 0.76(0.15), p = 0.001). There were no differences in the behaviour scores. Children of mothers with NS also had significantly lower weight-for-age z scores (WAZ) (p = 0.003) and length/height-for-age z scores (LAZ/HAZ) (p = 0.001); with 19(35.9 %) of them stunted. CONCLUSION Children whose mothers have NS have poorer neurodevelopmental, nutritional, and physical growth outcomes. Interventions to improve outcomes in these children are warranted.
Collapse
Affiliation(s)
- Mary Nyakato
- College of Health Sciences, Makerere University, P.O. Box 7072, Kampala-Uganda; Global Health Uganda, P.O. Box 33842, Kampala-Uganda.
| | - Shubaya Kasule Naggayi
- College of Health Sciences, Makerere University, P.O. Box 7072, Kampala-Uganda; Global Health Uganda, P.O. Box 33842, Kampala-Uganda
| | - Pamela Rosemary Akun
- College of Health Sciences, Makerere University, P.O. Box 7072, Kampala-Uganda; Centre of Tropical Neuroscience, Kitgum Site, P.O. Box 27520, Kampala-Uganda
| | - Sam Ononge
- College of Health Sciences, Makerere University, P.O. Box 7072, Kampala-Uganda
| | - Alfred Odong
- Global Health Uganda, P.O. Box 33842, Kampala-Uganda
| | - Erias Adams Baguma
- Makerere University-John Hopkins University Research Collaboration, P.O. Box 23491, Kampala-Uganda
| | - Hellen Nansiiro
- College of Health Sciences, Makerere University, P.O. Box 7072, Kampala-Uganda
| | | | - Simple Ouma
- College of Health Sciences, Makerere University, P.O. Box 7072, Kampala-Uganda
| | - Innocent Besigye
- College of Health Sciences, Makerere University, P.O. Box 7072, Kampala-Uganda
| | - Richard Idro
- College of Health Sciences, Makerere University, P.O. Box 7072, Kampala-Uganda; Global Health Uganda, P.O. Box 33842, Kampala-Uganda; Centre of Tropical Neuroscience, Kitgum Site, P.O. Box 27520, Kampala-Uganda
| |
Collapse
|
20
|
Karimi Z, Zarifkar A, Mirzaei E, Dianatpour M, Dara M, Aligholi H. Therapeutic effects of nanosilibinin in valproic acid-zebrafish model of autism spectrum disorder: Focusing on Wnt signaling pathway and autism spectrum disorder-related cytokines. Int J Dev Neurosci 2024; 84:454-468. [PMID: 38961588 DOI: 10.1002/jdn.10348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 05/08/2024] [Accepted: 05/13/2024] [Indexed: 07/05/2024] Open
Abstract
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.
Collapse
Affiliation(s)
- Zahra Karimi
- Department of Neuroscience, School of Advanced Medical Science and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Asadollah Zarifkar
- Department of Physiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Esmaeil Mirzaei
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Dianatpour
- Stem Cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Medical Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahintaj Dara
- Stem Cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hadi Aligholi
- Department of Neuroscience, School of Advanced Medical Science and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
21
|
Roh SH, Mendez-Vazquez H, Sathler MF, Doolittle MJ, Zaytseva A, Brown H, Sainsbury M, Kim S. Prenatal exposure to valproic acid reduces synaptic δ-catenin levels and disrupts ultrasonic vocalization in neonates. Neuropharmacology 2024; 253:109963. [PMID: 38657945 PMCID: PMC11127754 DOI: 10.1016/j.neuropharm.2024.109963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 04/26/2024]
Abstract
Valproic acid (VPA) is an effective and commonly prescribed drug for epilepsy and bipolar disorder. However, children born from mothers treated with VPA during pregnancy exhibit an increased incidence of autism spectrum disorder (ASD). Although VPA may impair brain development at the cellular level, the mechanism of VPA-induced ASD has not been completely addressed. A previous study has found that VPA treatment strongly reduces δ-catenin mRNA levels in cultured human neurons. δ-catenin is important for the control of glutamatergic synapses and is strongly associated with ASD. VPA inhibits dendritic morphogenesis in developing neurons, an effect that is also found in neurons lacking δ-catenin expression. We thus hypothesize that prenatal exposure to VPA significantly reduces δ-catenin levels in the brain, which impairs glutamatergic synapses to cause ASD. Here, we found that prenatal exposure to VPA markedly reduced δ-catenin levels in the brain of mouse pups. VPA treatment also impaired dendritic branching in developing mouse cortical neurons, which was partially reversed by elevating δ-catenin expression. Prenatal VPA exposure significantly reduced synaptic α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor levels and postsynaptic density 95 (PSD95) in the brain of mouse pups, indicating dysfunctions in glutamatergic synaptic transmission. VPA exposure also significantly altered ultrasonic vocalization (USV) in newly born pups when they were isolated from their nest. Moreover, VPA-exposed pups show impaired hypothalamic response to isolation, which is required to produce animals' USVs following isolation from the nest. Therefore, these results suggest that VPA-induced ASD pathology can be mediated by the loss of δ-catenin functions.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Morgan Sainsbury
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, 80523, USA
| | - Seonil Kim
- Department of Biomedical Sciences, USA; Molecular, Cellular and Integrative Neurosciences Program, USA.
| |
Collapse
|
22
|
Chen B, Xu X, Wang Y, Yang Z, Liu C, Zhang T. VPA-induced autism impairs memory ability through disturbing neural oscillatory patterns in offspring rats. Cogn Neurodyn 2024; 18:1563-1574. [PMID: 39104704 PMCID: PMC11297858 DOI: 10.1007/s11571-023-09996-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 06/17/2023] [Accepted: 08/07/2023] [Indexed: 08/07/2024] Open
Abstract
Autism spectrum disorder (ASD) is a general neurodevelopmental disease characterized by unusual social communication and rigid, repetitive behavior patterns. The purpose of this study was to investigate the effects of ASD on the alteration of neural oscillatory patterns and synaptic plasticity, which commonly supported a wide range of basic and higher memory activities. Accordingly, a prenatal valproic acid (VPA) exposure rat model was established for studying autism. The behavioral experiments showed that the social orientation declined and the memory ability was significantly impaired in VPA rats, which was closely associated with the synaptic plasticity deficits. Neural oscillation is the rhythmic neuron-activity, and the pathological characteristics and neurological changes in autism may be peeped at the neural oscillatory analysis. Interestingly, neural oscillatory analysis showed that prenatal VPA exposure reduced the low-frequency power but increased high-frequency gamma (HG) power in the hippocampus CA1 area. Meanwhile, the coherence and synchronization between CA3 and CA1 were abnormally increased in the VPA group, especially in theta and HG rhythms. Furthermore, the cross-frequency coupling strength of theta-LG in the CA1 and CA3 → CA1 pathway was significantly attenuated, but the theta-HG coupling strength was increased. Additionally, prenatal VPA exposure inhibited the expression of SYP and NR2B but enhanced the expression of PSD-95 along with decreased synaptic plasticity. The neural oscillatory patterns in VPA-induced offspring were disturbed with the intensity and direction of neural information flow disordered, which are consistent with the changes in synaptic plasticity, suggesting that the decline in synaptic plasticity is the underlying mechanism.
Collapse
Affiliation(s)
- Bin Chen
- College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, 300071 People’s Republic of China
| | - Xinxin Xu
- College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, 300071 People’s Republic of China
| | - Yue Wang
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071 People’s Republic of China
| | - Zhuo Yang
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071 People’s Republic of China
| | - Chunhua Liu
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071 People’s Republic of China
| | - Tao Zhang
- College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, 300071 People’s Republic of China
| |
Collapse
|
23
|
Xu SC, Zhong Y, Jiang HY, Tang J. Exposure to anti-seizure medication during pregnancy and the risk of autism and ADHD in offspring: a systematic review and meta-analysis. Front Neurol 2024; 15:1440145. [PMID: 39105059 PMCID: PMC11298387 DOI: 10.3389/fneur.2024.1440145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 07/10/2024] [Indexed: 08/07/2024] Open
Abstract
Background Evidence of an association between maternal use of anti-seizure medication (ASM) during pregnancy and the risk of autism spectrum disorder (ASD) or attention-deficit/hyperactivity disorder (ADHD) in children is conflicting. This systematic review and meta-analysis aimed to summarize the relationship between fetal exposure to ASM and the development of ASD or ADHD in offspring. Methods A comprehensive literature search was conducted in PubMed and other databases to identify relevant epidemiological studies published from inception until 1 March 2024. Results Seven cohort studies were included in the meta-analysis. The results showed that maternal exposure to ASMs during pregnancy was associated with an increased risk of ASD [odds ratio (OR): 2.1, 95% confidence interval (CI): 1.63-2.71; p < 0.001] in the general population. This association became weaker (ASD: OR: 1.38, 95% CI: 1.11-1.73; p = 0.004) when the reference group was mothers with a psychiatric disorder or epilepsy not treated during pregnancy. Furthermore, an increased risk of ADHD was observed when the study data adjusted for drug indications were pooled (OR: 1.43, 95% CI: 1.07-1.92; p = 0.015). In subgroup analyses based on individual ASM use, only exposure to valproate preconception was significantly associated with an increased risk of ASD or ADHD. Conclusion The significant association between maternal ASM use during pregnancy and ASD or ADHD in offspring may be partially explained by the drug indication or driven by valproate.
Collapse
Affiliation(s)
- Shan-Chun Xu
- Department of General Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China
| | - Ying Zhong
- Department of Orthopaedics, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China
| | - Hai-Yin Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jun Tang
- Department of General Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China
| |
Collapse
|
24
|
Pack AM, Oskoui M, Williams Roberson S, Donley DK, French J, Gerard EE, Gloss D, Miller WR, Munger Clary HM, Osmundson SS, McFadden B, Parratt K, Pennell PB, Saade G, Smith DB, Sullivan K, Thomas SV, Tomson T, Dolan O'Brien M, Botchway-Doe K, Silsbee HM, Keezer MR. Teratogenesis, Perinatal, and Neurodevelopmental Outcomes After In Utero Exposure to Antiseizure Medication: Practice Guideline From the AAN, AES, and SMFM. Neurology 2024; 102:e209279. [PMID: 38748979 PMCID: PMC11175651 DOI: 10.1212/wnl.0000000000209279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 02/21/2024] [Indexed: 06/15/2024] Open
Abstract
This practice guideline provides updated evidence-based conclusions and recommendations regarding the effects of antiseizure medications (ASMs) and folic acid supplementation on the prevalence of major congenital malformations (MCMs), adverse perinatal outcomes, and neurodevelopmental outcomes in children born to people with epilepsy of childbearing potential (PWECP). A multidisciplinary panel conducted a systematic review and developed practice recommendations following the process outlined in the 2017 edition of the American Academy of Neurology Clinical Practice Guideline Process Manual. The systematic review includes studies through August 2022. Recommendations are supported by structured rationales that integrate evidence from the systematic review, related evidence, principles of care, and inferences from evidence. The following are some of the major recommendations. When treating PWECP, clinicians should recommend ASMs and doses that optimize both seizure control and fetal outcomes should pregnancy occur, at the earliest possible opportunity preconceptionally. Clinicians must minimize the occurrence of convulsive seizures in PWECP during pregnancy to minimize potential risks to the birth parent and to the fetus. Once a PWECP is already pregnant, clinicians should exercise caution in attempting to remove or replace an ASM that is effective in controlling generalized tonic-clonic or focal-to-bilateral tonic-clonic seizures. Clinicians must consider using lamotrigine, levetiracetam, or oxcarbazepine in PWECP when appropriate based on the patient's epilepsy syndrome, likelihood of achieving seizure control, and comorbidities, to minimize the risk of MCMs. Clinicians must avoid the use of valproic acid in PWECP to minimize the risk of MCMs or neural tube defects (NTDs), if clinically feasible. Clinicians should avoid the use of valproic acid or topiramate in PWECP to minimize the risk of offspring being born small for gestational age, if clinically feasible. To reduce the risk of poor neurodevelopmental outcomes, including autism spectrum disorder and lower IQ, in children born to PWECP, clinicians must avoid the use of valproic acid in PWECP, if clinically feasible. Clinicians should prescribe at least 0.4 mg of folic acid supplementation daily preconceptionally and during pregnancy to any PWECP treated with an ASM to decrease the risk of NTDs and possibly improve neurodevelopmental outcomes in the offspring.
Collapse
Affiliation(s)
- Alison M Pack
- From the Department of Neurology (A.M.P.), Columbia University, New York City; Departments of Pediatrics and Neurology & Neurosurgery (M.O.), McGill University, Montreal, Quebec, Canada; Departments of Neurology (S.W.R.), Biomedical Engineering (S.W.R.), and Obstetrics and Gynecology (S.S.O.), Vanderbilt University Medical Center, Nashville, TN; Northern Michigan Neurology and Munson Medical Center (D.K.D.), Traverse City, MI; Department of Neurology (J.F.), NYU Grossman School of Medicine, New York City; Feinberg School of Medicine (E.E.G.), Northwestern University, Chicago, IL; The NeuroMedical Center (D.G.), Baton Rouge, LA; Epilepsy Foundation (W.R.M.), Bowie, MD; Department of Neurology (H.M.M.C.), Wake Forest University School of Medicine, Winston-Salem, NC; My Epilepsy Story (B.M.), Nashville, TN; Institute of Clinical Neurosciences (K.P.), Royal Prince Alfred Hospital, Sydney, Australia; Department of Neurology (P.B.P.), University of Pittsburgh School of Medicine, PA; Department of Ob-Gyn (G.S.), Eastern Virginia Medical School, Norfolk; Department of Neurology (D.B.S.), University of Colorado School of Medicine, Aurora; Department of Biostatistics, Epidemiology, and Environmental Health Sciences (K.S.), Jiann-Ping Hsu College of Public Health, Georgia Southern University, Statesboro; Department of Neurology (S.V.T.), Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India; Department of Clinical Neuroscience (T.T.), Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden; American Academy of Neurology (M.D.O.B., K.B.-D., H.M.S.), Minneapolis, MN; and Centre Hospitalier de l'Université de Montréal Research Centre (CRCHUM) (M.R.K.), Quebec, Canada
| | - Maryam Oskoui
- From the Department of Neurology (A.M.P.), Columbia University, New York City; Departments of Pediatrics and Neurology & Neurosurgery (M.O.), McGill University, Montreal, Quebec, Canada; Departments of Neurology (S.W.R.), Biomedical Engineering (S.W.R.), and Obstetrics and Gynecology (S.S.O.), Vanderbilt University Medical Center, Nashville, TN; Northern Michigan Neurology and Munson Medical Center (D.K.D.), Traverse City, MI; Department of Neurology (J.F.), NYU Grossman School of Medicine, New York City; Feinberg School of Medicine (E.E.G.), Northwestern University, Chicago, IL; The NeuroMedical Center (D.G.), Baton Rouge, LA; Epilepsy Foundation (W.R.M.), Bowie, MD; Department of Neurology (H.M.M.C.), Wake Forest University School of Medicine, Winston-Salem, NC; My Epilepsy Story (B.M.), Nashville, TN; Institute of Clinical Neurosciences (K.P.), Royal Prince Alfred Hospital, Sydney, Australia; Department of Neurology (P.B.P.), University of Pittsburgh School of Medicine, PA; Department of Ob-Gyn (G.S.), Eastern Virginia Medical School, Norfolk; Department of Neurology (D.B.S.), University of Colorado School of Medicine, Aurora; Department of Biostatistics, Epidemiology, and Environmental Health Sciences (K.S.), Jiann-Ping Hsu College of Public Health, Georgia Southern University, Statesboro; Department of Neurology (S.V.T.), Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India; Department of Clinical Neuroscience (T.T.), Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden; American Academy of Neurology (M.D.O.B., K.B.-D., H.M.S.), Minneapolis, MN; and Centre Hospitalier de l'Université de Montréal Research Centre (CRCHUM) (M.R.K.), Quebec, Canada
| | - Shawniqua Williams Roberson
- From the Department of Neurology (A.M.P.), Columbia University, New York City; Departments of Pediatrics and Neurology & Neurosurgery (M.O.), McGill University, Montreal, Quebec, Canada; Departments of Neurology (S.W.R.), Biomedical Engineering (S.W.R.), and Obstetrics and Gynecology (S.S.O.), Vanderbilt University Medical Center, Nashville, TN; Northern Michigan Neurology and Munson Medical Center (D.K.D.), Traverse City, MI; Department of Neurology (J.F.), NYU Grossman School of Medicine, New York City; Feinberg School of Medicine (E.E.G.), Northwestern University, Chicago, IL; The NeuroMedical Center (D.G.), Baton Rouge, LA; Epilepsy Foundation (W.R.M.), Bowie, MD; Department of Neurology (H.M.M.C.), Wake Forest University School of Medicine, Winston-Salem, NC; My Epilepsy Story (B.M.), Nashville, TN; Institute of Clinical Neurosciences (K.P.), Royal Prince Alfred Hospital, Sydney, Australia; Department of Neurology (P.B.P.), University of Pittsburgh School of Medicine, PA; Department of Ob-Gyn (G.S.), Eastern Virginia Medical School, Norfolk; Department of Neurology (D.B.S.), University of Colorado School of Medicine, Aurora; Department of Biostatistics, Epidemiology, and Environmental Health Sciences (K.S.), Jiann-Ping Hsu College of Public Health, Georgia Southern University, Statesboro; Department of Neurology (S.V.T.), Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India; Department of Clinical Neuroscience (T.T.), Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden; American Academy of Neurology (M.D.O.B., K.B.-D., H.M.S.), Minneapolis, MN; and Centre Hospitalier de l'Université de Montréal Research Centre (CRCHUM) (M.R.K.), Quebec, Canada
| | - Diane K Donley
- From the Department of Neurology (A.M.P.), Columbia University, New York City; Departments of Pediatrics and Neurology & Neurosurgery (M.O.), McGill University, Montreal, Quebec, Canada; Departments of Neurology (S.W.R.), Biomedical Engineering (S.W.R.), and Obstetrics and Gynecology (S.S.O.), Vanderbilt University Medical Center, Nashville, TN; Northern Michigan Neurology and Munson Medical Center (D.K.D.), Traverse City, MI; Department of Neurology (J.F.), NYU Grossman School of Medicine, New York City; Feinberg School of Medicine (E.E.G.), Northwestern University, Chicago, IL; The NeuroMedical Center (D.G.), Baton Rouge, LA; Epilepsy Foundation (W.R.M.), Bowie, MD; Department of Neurology (H.M.M.C.), Wake Forest University School of Medicine, Winston-Salem, NC; My Epilepsy Story (B.M.), Nashville, TN; Institute of Clinical Neurosciences (K.P.), Royal Prince Alfred Hospital, Sydney, Australia; Department of Neurology (P.B.P.), University of Pittsburgh School of Medicine, PA; Department of Ob-Gyn (G.S.), Eastern Virginia Medical School, Norfolk; Department of Neurology (D.B.S.), University of Colorado School of Medicine, Aurora; Department of Biostatistics, Epidemiology, and Environmental Health Sciences (K.S.), Jiann-Ping Hsu College of Public Health, Georgia Southern University, Statesboro; Department of Neurology (S.V.T.), Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India; Department of Clinical Neuroscience (T.T.), Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden; American Academy of Neurology (M.D.O.B., K.B.-D., H.M.S.), Minneapolis, MN; and Centre Hospitalier de l'Université de Montréal Research Centre (CRCHUM) (M.R.K.), Quebec, Canada
| | - Jacqueline French
- From the Department of Neurology (A.M.P.), Columbia University, New York City; Departments of Pediatrics and Neurology & Neurosurgery (M.O.), McGill University, Montreal, Quebec, Canada; Departments of Neurology (S.W.R.), Biomedical Engineering (S.W.R.), and Obstetrics and Gynecology (S.S.O.), Vanderbilt University Medical Center, Nashville, TN; Northern Michigan Neurology and Munson Medical Center (D.K.D.), Traverse City, MI; Department of Neurology (J.F.), NYU Grossman School of Medicine, New York City; Feinberg School of Medicine (E.E.G.), Northwestern University, Chicago, IL; The NeuroMedical Center (D.G.), Baton Rouge, LA; Epilepsy Foundation (W.R.M.), Bowie, MD; Department of Neurology (H.M.M.C.), Wake Forest University School of Medicine, Winston-Salem, NC; My Epilepsy Story (B.M.), Nashville, TN; Institute of Clinical Neurosciences (K.P.), Royal Prince Alfred Hospital, Sydney, Australia; Department of Neurology (P.B.P.), University of Pittsburgh School of Medicine, PA; Department of Ob-Gyn (G.S.), Eastern Virginia Medical School, Norfolk; Department of Neurology (D.B.S.), University of Colorado School of Medicine, Aurora; Department of Biostatistics, Epidemiology, and Environmental Health Sciences (K.S.), Jiann-Ping Hsu College of Public Health, Georgia Southern University, Statesboro; Department of Neurology (S.V.T.), Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India; Department of Clinical Neuroscience (T.T.), Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden; American Academy of Neurology (M.D.O.B., K.B.-D., H.M.S.), Minneapolis, MN; and Centre Hospitalier de l'Université de Montréal Research Centre (CRCHUM) (M.R.K.), Quebec, Canada
| | - Elizabeth E Gerard
- From the Department of Neurology (A.M.P.), Columbia University, New York City; Departments of Pediatrics and Neurology & Neurosurgery (M.O.), McGill University, Montreal, Quebec, Canada; Departments of Neurology (S.W.R.), Biomedical Engineering (S.W.R.), and Obstetrics and Gynecology (S.S.O.), Vanderbilt University Medical Center, Nashville, TN; Northern Michigan Neurology and Munson Medical Center (D.K.D.), Traverse City, MI; Department of Neurology (J.F.), NYU Grossman School of Medicine, New York City; Feinberg School of Medicine (E.E.G.), Northwestern University, Chicago, IL; The NeuroMedical Center (D.G.), Baton Rouge, LA; Epilepsy Foundation (W.R.M.), Bowie, MD; Department of Neurology (H.M.M.C.), Wake Forest University School of Medicine, Winston-Salem, NC; My Epilepsy Story (B.M.), Nashville, TN; Institute of Clinical Neurosciences (K.P.), Royal Prince Alfred Hospital, Sydney, Australia; Department of Neurology (P.B.P.), University of Pittsburgh School of Medicine, PA; Department of Ob-Gyn (G.S.), Eastern Virginia Medical School, Norfolk; Department of Neurology (D.B.S.), University of Colorado School of Medicine, Aurora; Department of Biostatistics, Epidemiology, and Environmental Health Sciences (K.S.), Jiann-Ping Hsu College of Public Health, Georgia Southern University, Statesboro; Department of Neurology (S.V.T.), Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India; Department of Clinical Neuroscience (T.T.), Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden; American Academy of Neurology (M.D.O.B., K.B.-D., H.M.S.), Minneapolis, MN; and Centre Hospitalier de l'Université de Montréal Research Centre (CRCHUM) (M.R.K.), Quebec, Canada
| | - David Gloss
- From the Department of Neurology (A.M.P.), Columbia University, New York City; Departments of Pediatrics and Neurology & Neurosurgery (M.O.), McGill University, Montreal, Quebec, Canada; Departments of Neurology (S.W.R.), Biomedical Engineering (S.W.R.), and Obstetrics and Gynecology (S.S.O.), Vanderbilt University Medical Center, Nashville, TN; Northern Michigan Neurology and Munson Medical Center (D.K.D.), Traverse City, MI; Department of Neurology (J.F.), NYU Grossman School of Medicine, New York City; Feinberg School of Medicine (E.E.G.), Northwestern University, Chicago, IL; The NeuroMedical Center (D.G.), Baton Rouge, LA; Epilepsy Foundation (W.R.M.), Bowie, MD; Department of Neurology (H.M.M.C.), Wake Forest University School of Medicine, Winston-Salem, NC; My Epilepsy Story (B.M.), Nashville, TN; Institute of Clinical Neurosciences (K.P.), Royal Prince Alfred Hospital, Sydney, Australia; Department of Neurology (P.B.P.), University of Pittsburgh School of Medicine, PA; Department of Ob-Gyn (G.S.), Eastern Virginia Medical School, Norfolk; Department of Neurology (D.B.S.), University of Colorado School of Medicine, Aurora; Department of Biostatistics, Epidemiology, and Environmental Health Sciences (K.S.), Jiann-Ping Hsu College of Public Health, Georgia Southern University, Statesboro; Department of Neurology (S.V.T.), Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India; Department of Clinical Neuroscience (T.T.), Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden; American Academy of Neurology (M.D.O.B., K.B.-D., H.M.S.), Minneapolis, MN; and Centre Hospitalier de l'Université de Montréal Research Centre (CRCHUM) (M.R.K.), Quebec, Canada
| | - Wendy R Miller
- From the Department of Neurology (A.M.P.), Columbia University, New York City; Departments of Pediatrics and Neurology & Neurosurgery (M.O.), McGill University, Montreal, Quebec, Canada; Departments of Neurology (S.W.R.), Biomedical Engineering (S.W.R.), and Obstetrics and Gynecology (S.S.O.), Vanderbilt University Medical Center, Nashville, TN; Northern Michigan Neurology and Munson Medical Center (D.K.D.), Traverse City, MI; Department of Neurology (J.F.), NYU Grossman School of Medicine, New York City; Feinberg School of Medicine (E.E.G.), Northwestern University, Chicago, IL; The NeuroMedical Center (D.G.), Baton Rouge, LA; Epilepsy Foundation (W.R.M.), Bowie, MD; Department of Neurology (H.M.M.C.), Wake Forest University School of Medicine, Winston-Salem, NC; My Epilepsy Story (B.M.), Nashville, TN; Institute of Clinical Neurosciences (K.P.), Royal Prince Alfred Hospital, Sydney, Australia; Department of Neurology (P.B.P.), University of Pittsburgh School of Medicine, PA; Department of Ob-Gyn (G.S.), Eastern Virginia Medical School, Norfolk; Department of Neurology (D.B.S.), University of Colorado School of Medicine, Aurora; Department of Biostatistics, Epidemiology, and Environmental Health Sciences (K.S.), Jiann-Ping Hsu College of Public Health, Georgia Southern University, Statesboro; Department of Neurology (S.V.T.), Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India; Department of Clinical Neuroscience (T.T.), Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden; American Academy of Neurology (M.D.O.B., K.B.-D., H.M.S.), Minneapolis, MN; and Centre Hospitalier de l'Université de Montréal Research Centre (CRCHUM) (M.R.K.), Quebec, Canada
| | - Heidi M Munger Clary
- From the Department of Neurology (A.M.P.), Columbia University, New York City; Departments of Pediatrics and Neurology & Neurosurgery (M.O.), McGill University, Montreal, Quebec, Canada; Departments of Neurology (S.W.R.), Biomedical Engineering (S.W.R.), and Obstetrics and Gynecology (S.S.O.), Vanderbilt University Medical Center, Nashville, TN; Northern Michigan Neurology and Munson Medical Center (D.K.D.), Traverse City, MI; Department of Neurology (J.F.), NYU Grossman School of Medicine, New York City; Feinberg School of Medicine (E.E.G.), Northwestern University, Chicago, IL; The NeuroMedical Center (D.G.), Baton Rouge, LA; Epilepsy Foundation (W.R.M.), Bowie, MD; Department of Neurology (H.M.M.C.), Wake Forest University School of Medicine, Winston-Salem, NC; My Epilepsy Story (B.M.), Nashville, TN; Institute of Clinical Neurosciences (K.P.), Royal Prince Alfred Hospital, Sydney, Australia; Department of Neurology (P.B.P.), University of Pittsburgh School of Medicine, PA; Department of Ob-Gyn (G.S.), Eastern Virginia Medical School, Norfolk; Department of Neurology (D.B.S.), University of Colorado School of Medicine, Aurora; Department of Biostatistics, Epidemiology, and Environmental Health Sciences (K.S.), Jiann-Ping Hsu College of Public Health, Georgia Southern University, Statesboro; Department of Neurology (S.V.T.), Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India; Department of Clinical Neuroscience (T.T.), Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden; American Academy of Neurology (M.D.O.B., K.B.-D., H.M.S.), Minneapolis, MN; and Centre Hospitalier de l'Université de Montréal Research Centre (CRCHUM) (M.R.K.), Quebec, Canada
| | - Sarah S Osmundson
- From the Department of Neurology (A.M.P.), Columbia University, New York City; Departments of Pediatrics and Neurology & Neurosurgery (M.O.), McGill University, Montreal, Quebec, Canada; Departments of Neurology (S.W.R.), Biomedical Engineering (S.W.R.), and Obstetrics and Gynecology (S.S.O.), Vanderbilt University Medical Center, Nashville, TN; Northern Michigan Neurology and Munson Medical Center (D.K.D.), Traverse City, MI; Department of Neurology (J.F.), NYU Grossman School of Medicine, New York City; Feinberg School of Medicine (E.E.G.), Northwestern University, Chicago, IL; The NeuroMedical Center (D.G.), Baton Rouge, LA; Epilepsy Foundation (W.R.M.), Bowie, MD; Department of Neurology (H.M.M.C.), Wake Forest University School of Medicine, Winston-Salem, NC; My Epilepsy Story (B.M.), Nashville, TN; Institute of Clinical Neurosciences (K.P.), Royal Prince Alfred Hospital, Sydney, Australia; Department of Neurology (P.B.P.), University of Pittsburgh School of Medicine, PA; Department of Ob-Gyn (G.S.), Eastern Virginia Medical School, Norfolk; Department of Neurology (D.B.S.), University of Colorado School of Medicine, Aurora; Department of Biostatistics, Epidemiology, and Environmental Health Sciences (K.S.), Jiann-Ping Hsu College of Public Health, Georgia Southern University, Statesboro; Department of Neurology (S.V.T.), Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India; Department of Clinical Neuroscience (T.T.), Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden; American Academy of Neurology (M.D.O.B., K.B.-D., H.M.S.), Minneapolis, MN; and Centre Hospitalier de l'Université de Montréal Research Centre (CRCHUM) (M.R.K.), Quebec, Canada
| | - Brandy McFadden
- From the Department of Neurology (A.M.P.), Columbia University, New York City; Departments of Pediatrics and Neurology & Neurosurgery (M.O.), McGill University, Montreal, Quebec, Canada; Departments of Neurology (S.W.R.), Biomedical Engineering (S.W.R.), and Obstetrics and Gynecology (S.S.O.), Vanderbilt University Medical Center, Nashville, TN; Northern Michigan Neurology and Munson Medical Center (D.K.D.), Traverse City, MI; Department of Neurology (J.F.), NYU Grossman School of Medicine, New York City; Feinberg School of Medicine (E.E.G.), Northwestern University, Chicago, IL; The NeuroMedical Center (D.G.), Baton Rouge, LA; Epilepsy Foundation (W.R.M.), Bowie, MD; Department of Neurology (H.M.M.C.), Wake Forest University School of Medicine, Winston-Salem, NC; My Epilepsy Story (B.M.), Nashville, TN; Institute of Clinical Neurosciences (K.P.), Royal Prince Alfred Hospital, Sydney, Australia; Department of Neurology (P.B.P.), University of Pittsburgh School of Medicine, PA; Department of Ob-Gyn (G.S.), Eastern Virginia Medical School, Norfolk; Department of Neurology (D.B.S.), University of Colorado School of Medicine, Aurora; Department of Biostatistics, Epidemiology, and Environmental Health Sciences (K.S.), Jiann-Ping Hsu College of Public Health, Georgia Southern University, Statesboro; Department of Neurology (S.V.T.), Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India; Department of Clinical Neuroscience (T.T.), Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden; American Academy of Neurology (M.D.O.B., K.B.-D., H.M.S.), Minneapolis, MN; and Centre Hospitalier de l'Université de Montréal Research Centre (CRCHUM) (M.R.K.), Quebec, Canada
| | - Kaitlyn Parratt
- From the Department of Neurology (A.M.P.), Columbia University, New York City; Departments of Pediatrics and Neurology & Neurosurgery (M.O.), McGill University, Montreal, Quebec, Canada; Departments of Neurology (S.W.R.), Biomedical Engineering (S.W.R.), and Obstetrics and Gynecology (S.S.O.), Vanderbilt University Medical Center, Nashville, TN; Northern Michigan Neurology and Munson Medical Center (D.K.D.), Traverse City, MI; Department of Neurology (J.F.), NYU Grossman School of Medicine, New York City; Feinberg School of Medicine (E.E.G.), Northwestern University, Chicago, IL; The NeuroMedical Center (D.G.), Baton Rouge, LA; Epilepsy Foundation (W.R.M.), Bowie, MD; Department of Neurology (H.M.M.C.), Wake Forest University School of Medicine, Winston-Salem, NC; My Epilepsy Story (B.M.), Nashville, TN; Institute of Clinical Neurosciences (K.P.), Royal Prince Alfred Hospital, Sydney, Australia; Department of Neurology (P.B.P.), University of Pittsburgh School of Medicine, PA; Department of Ob-Gyn (G.S.), Eastern Virginia Medical School, Norfolk; Department of Neurology (D.B.S.), University of Colorado School of Medicine, Aurora; Department of Biostatistics, Epidemiology, and Environmental Health Sciences (K.S.), Jiann-Ping Hsu College of Public Health, Georgia Southern University, Statesboro; Department of Neurology (S.V.T.), Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India; Department of Clinical Neuroscience (T.T.), Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden; American Academy of Neurology (M.D.O.B., K.B.-D., H.M.S.), Minneapolis, MN; and Centre Hospitalier de l'Université de Montréal Research Centre (CRCHUM) (M.R.K.), Quebec, Canada
| | - Page B Pennell
- From the Department of Neurology (A.M.P.), Columbia University, New York City; Departments of Pediatrics and Neurology & Neurosurgery (M.O.), McGill University, Montreal, Quebec, Canada; Departments of Neurology (S.W.R.), Biomedical Engineering (S.W.R.), and Obstetrics and Gynecology (S.S.O.), Vanderbilt University Medical Center, Nashville, TN; Northern Michigan Neurology and Munson Medical Center (D.K.D.), Traverse City, MI; Department of Neurology (J.F.), NYU Grossman School of Medicine, New York City; Feinberg School of Medicine (E.E.G.), Northwestern University, Chicago, IL; The NeuroMedical Center (D.G.), Baton Rouge, LA; Epilepsy Foundation (W.R.M.), Bowie, MD; Department of Neurology (H.M.M.C.), Wake Forest University School of Medicine, Winston-Salem, NC; My Epilepsy Story (B.M.), Nashville, TN; Institute of Clinical Neurosciences (K.P.), Royal Prince Alfred Hospital, Sydney, Australia; Department of Neurology (P.B.P.), University of Pittsburgh School of Medicine, PA; Department of Ob-Gyn (G.S.), Eastern Virginia Medical School, Norfolk; Department of Neurology (D.B.S.), University of Colorado School of Medicine, Aurora; Department of Biostatistics, Epidemiology, and Environmental Health Sciences (K.S.), Jiann-Ping Hsu College of Public Health, Georgia Southern University, Statesboro; Department of Neurology (S.V.T.), Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India; Department of Clinical Neuroscience (T.T.), Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden; American Academy of Neurology (M.D.O.B., K.B.-D., H.M.S.), Minneapolis, MN; and Centre Hospitalier de l'Université de Montréal Research Centre (CRCHUM) (M.R.K.), Quebec, Canada
| | - George Saade
- From the Department of Neurology (A.M.P.), Columbia University, New York City; Departments of Pediatrics and Neurology & Neurosurgery (M.O.), McGill University, Montreal, Quebec, Canada; Departments of Neurology (S.W.R.), Biomedical Engineering (S.W.R.), and Obstetrics and Gynecology (S.S.O.), Vanderbilt University Medical Center, Nashville, TN; Northern Michigan Neurology and Munson Medical Center (D.K.D.), Traverse City, MI; Department of Neurology (J.F.), NYU Grossman School of Medicine, New York City; Feinberg School of Medicine (E.E.G.), Northwestern University, Chicago, IL; The NeuroMedical Center (D.G.), Baton Rouge, LA; Epilepsy Foundation (W.R.M.), Bowie, MD; Department of Neurology (H.M.M.C.), Wake Forest University School of Medicine, Winston-Salem, NC; My Epilepsy Story (B.M.), Nashville, TN; Institute of Clinical Neurosciences (K.P.), Royal Prince Alfred Hospital, Sydney, Australia; Department of Neurology (P.B.P.), University of Pittsburgh School of Medicine, PA; Department of Ob-Gyn (G.S.), Eastern Virginia Medical School, Norfolk; Department of Neurology (D.B.S.), University of Colorado School of Medicine, Aurora; Department of Biostatistics, Epidemiology, and Environmental Health Sciences (K.S.), Jiann-Ping Hsu College of Public Health, Georgia Southern University, Statesboro; Department of Neurology (S.V.T.), Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India; Department of Clinical Neuroscience (T.T.), Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden; American Academy of Neurology (M.D.O.B., K.B.-D., H.M.S.), Minneapolis, MN; and Centre Hospitalier de l'Université de Montréal Research Centre (CRCHUM) (M.R.K.), Quebec, Canada
| | - Don B Smith
- From the Department of Neurology (A.M.P.), Columbia University, New York City; Departments of Pediatrics and Neurology & Neurosurgery (M.O.), McGill University, Montreal, Quebec, Canada; Departments of Neurology (S.W.R.), Biomedical Engineering (S.W.R.), and Obstetrics and Gynecology (S.S.O.), Vanderbilt University Medical Center, Nashville, TN; Northern Michigan Neurology and Munson Medical Center (D.K.D.), Traverse City, MI; Department of Neurology (J.F.), NYU Grossman School of Medicine, New York City; Feinberg School of Medicine (E.E.G.), Northwestern University, Chicago, IL; The NeuroMedical Center (D.G.), Baton Rouge, LA; Epilepsy Foundation (W.R.M.), Bowie, MD; Department of Neurology (H.M.M.C.), Wake Forest University School of Medicine, Winston-Salem, NC; My Epilepsy Story (B.M.), Nashville, TN; Institute of Clinical Neurosciences (K.P.), Royal Prince Alfred Hospital, Sydney, Australia; Department of Neurology (P.B.P.), University of Pittsburgh School of Medicine, PA; Department of Ob-Gyn (G.S.), Eastern Virginia Medical School, Norfolk; Department of Neurology (D.B.S.), University of Colorado School of Medicine, Aurora; Department of Biostatistics, Epidemiology, and Environmental Health Sciences (K.S.), Jiann-Ping Hsu College of Public Health, Georgia Southern University, Statesboro; Department of Neurology (S.V.T.), Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India; Department of Clinical Neuroscience (T.T.), Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden; American Academy of Neurology (M.D.O.B., K.B.-D., H.M.S.), Minneapolis, MN; and Centre Hospitalier de l'Université de Montréal Research Centre (CRCHUM) (M.R.K.), Quebec, Canada
| | - Kelly Sullivan
- From the Department of Neurology (A.M.P.), Columbia University, New York City; Departments of Pediatrics and Neurology & Neurosurgery (M.O.), McGill University, Montreal, Quebec, Canada; Departments of Neurology (S.W.R.), Biomedical Engineering (S.W.R.), and Obstetrics and Gynecology (S.S.O.), Vanderbilt University Medical Center, Nashville, TN; Northern Michigan Neurology and Munson Medical Center (D.K.D.), Traverse City, MI; Department of Neurology (J.F.), NYU Grossman School of Medicine, New York City; Feinberg School of Medicine (E.E.G.), Northwestern University, Chicago, IL; The NeuroMedical Center (D.G.), Baton Rouge, LA; Epilepsy Foundation (W.R.M.), Bowie, MD; Department of Neurology (H.M.M.C.), Wake Forest University School of Medicine, Winston-Salem, NC; My Epilepsy Story (B.M.), Nashville, TN; Institute of Clinical Neurosciences (K.P.), Royal Prince Alfred Hospital, Sydney, Australia; Department of Neurology (P.B.P.), University of Pittsburgh School of Medicine, PA; Department of Ob-Gyn (G.S.), Eastern Virginia Medical School, Norfolk; Department of Neurology (D.B.S.), University of Colorado School of Medicine, Aurora; Department of Biostatistics, Epidemiology, and Environmental Health Sciences (K.S.), Jiann-Ping Hsu College of Public Health, Georgia Southern University, Statesboro; Department of Neurology (S.V.T.), Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India; Department of Clinical Neuroscience (T.T.), Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden; American Academy of Neurology (M.D.O.B., K.B.-D., H.M.S.), Minneapolis, MN; and Centre Hospitalier de l'Université de Montréal Research Centre (CRCHUM) (M.R.K.), Quebec, Canada
| | - Sanjeev V Thomas
- From the Department of Neurology (A.M.P.), Columbia University, New York City; Departments of Pediatrics and Neurology & Neurosurgery (M.O.), McGill University, Montreal, Quebec, Canada; Departments of Neurology (S.W.R.), Biomedical Engineering (S.W.R.), and Obstetrics and Gynecology (S.S.O.), Vanderbilt University Medical Center, Nashville, TN; Northern Michigan Neurology and Munson Medical Center (D.K.D.), Traverse City, MI; Department of Neurology (J.F.), NYU Grossman School of Medicine, New York City; Feinberg School of Medicine (E.E.G.), Northwestern University, Chicago, IL; The NeuroMedical Center (D.G.), Baton Rouge, LA; Epilepsy Foundation (W.R.M.), Bowie, MD; Department of Neurology (H.M.M.C.), Wake Forest University School of Medicine, Winston-Salem, NC; My Epilepsy Story (B.M.), Nashville, TN; Institute of Clinical Neurosciences (K.P.), Royal Prince Alfred Hospital, Sydney, Australia; Department of Neurology (P.B.P.), University of Pittsburgh School of Medicine, PA; Department of Ob-Gyn (G.S.), Eastern Virginia Medical School, Norfolk; Department of Neurology (D.B.S.), University of Colorado School of Medicine, Aurora; Department of Biostatistics, Epidemiology, and Environmental Health Sciences (K.S.), Jiann-Ping Hsu College of Public Health, Georgia Southern University, Statesboro; Department of Neurology (S.V.T.), Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India; Department of Clinical Neuroscience (T.T.), Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden; American Academy of Neurology (M.D.O.B., K.B.-D., H.M.S.), Minneapolis, MN; and Centre Hospitalier de l'Université de Montréal Research Centre (CRCHUM) (M.R.K.), Quebec, Canada
| | - Torbjörn Tomson
- From the Department of Neurology (A.M.P.), Columbia University, New York City; Departments of Pediatrics and Neurology & Neurosurgery (M.O.), McGill University, Montreal, Quebec, Canada; Departments of Neurology (S.W.R.), Biomedical Engineering (S.W.R.), and Obstetrics and Gynecology (S.S.O.), Vanderbilt University Medical Center, Nashville, TN; Northern Michigan Neurology and Munson Medical Center (D.K.D.), Traverse City, MI; Department of Neurology (J.F.), NYU Grossman School of Medicine, New York City; Feinberg School of Medicine (E.E.G.), Northwestern University, Chicago, IL; The NeuroMedical Center (D.G.), Baton Rouge, LA; Epilepsy Foundation (W.R.M.), Bowie, MD; Department of Neurology (H.M.M.C.), Wake Forest University School of Medicine, Winston-Salem, NC; My Epilepsy Story (B.M.), Nashville, TN; Institute of Clinical Neurosciences (K.P.), Royal Prince Alfred Hospital, Sydney, Australia; Department of Neurology (P.B.P.), University of Pittsburgh School of Medicine, PA; Department of Ob-Gyn (G.S.), Eastern Virginia Medical School, Norfolk; Department of Neurology (D.B.S.), University of Colorado School of Medicine, Aurora; Department of Biostatistics, Epidemiology, and Environmental Health Sciences (K.S.), Jiann-Ping Hsu College of Public Health, Georgia Southern University, Statesboro; Department of Neurology (S.V.T.), Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India; Department of Clinical Neuroscience (T.T.), Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden; American Academy of Neurology (M.D.O.B., K.B.-D., H.M.S.), Minneapolis, MN; and Centre Hospitalier de l'Université de Montréal Research Centre (CRCHUM) (M.R.K.), Quebec, Canada
| | - Mary Dolan O'Brien
- From the Department of Neurology (A.M.P.), Columbia University, New York City; Departments of Pediatrics and Neurology & Neurosurgery (M.O.), McGill University, Montreal, Quebec, Canada; Departments of Neurology (S.W.R.), Biomedical Engineering (S.W.R.), and Obstetrics and Gynecology (S.S.O.), Vanderbilt University Medical Center, Nashville, TN; Northern Michigan Neurology and Munson Medical Center (D.K.D.), Traverse City, MI; Department of Neurology (J.F.), NYU Grossman School of Medicine, New York City; Feinberg School of Medicine (E.E.G.), Northwestern University, Chicago, IL; The NeuroMedical Center (D.G.), Baton Rouge, LA; Epilepsy Foundation (W.R.M.), Bowie, MD; Department of Neurology (H.M.M.C.), Wake Forest University School of Medicine, Winston-Salem, NC; My Epilepsy Story (B.M.), Nashville, TN; Institute of Clinical Neurosciences (K.P.), Royal Prince Alfred Hospital, Sydney, Australia; Department of Neurology (P.B.P.), University of Pittsburgh School of Medicine, PA; Department of Ob-Gyn (G.S.), Eastern Virginia Medical School, Norfolk; Department of Neurology (D.B.S.), University of Colorado School of Medicine, Aurora; Department of Biostatistics, Epidemiology, and Environmental Health Sciences (K.S.), Jiann-Ping Hsu College of Public Health, Georgia Southern University, Statesboro; Department of Neurology (S.V.T.), Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India; Department of Clinical Neuroscience (T.T.), Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden; American Academy of Neurology (M.D.O.B., K.B.-D., H.M.S.), Minneapolis, MN; and Centre Hospitalier de l'Université de Montréal Research Centre (CRCHUM) (M.R.K.), Quebec, Canada
| | - Kylie Botchway-Doe
- From the Department of Neurology (A.M.P.), Columbia University, New York City; Departments of Pediatrics and Neurology & Neurosurgery (M.O.), McGill University, Montreal, Quebec, Canada; Departments of Neurology (S.W.R.), Biomedical Engineering (S.W.R.), and Obstetrics and Gynecology (S.S.O.), Vanderbilt University Medical Center, Nashville, TN; Northern Michigan Neurology and Munson Medical Center (D.K.D.), Traverse City, MI; Department of Neurology (J.F.), NYU Grossman School of Medicine, New York City; Feinberg School of Medicine (E.E.G.), Northwestern University, Chicago, IL; The NeuroMedical Center (D.G.), Baton Rouge, LA; Epilepsy Foundation (W.R.M.), Bowie, MD; Department of Neurology (H.M.M.C.), Wake Forest University School of Medicine, Winston-Salem, NC; My Epilepsy Story (B.M.), Nashville, TN; Institute of Clinical Neurosciences (K.P.), Royal Prince Alfred Hospital, Sydney, Australia; Department of Neurology (P.B.P.), University of Pittsburgh School of Medicine, PA; Department of Ob-Gyn (G.S.), Eastern Virginia Medical School, Norfolk; Department of Neurology (D.B.S.), University of Colorado School of Medicine, Aurora; Department of Biostatistics, Epidemiology, and Environmental Health Sciences (K.S.), Jiann-Ping Hsu College of Public Health, Georgia Southern University, Statesboro; Department of Neurology (S.V.T.), Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India; Department of Clinical Neuroscience (T.T.), Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden; American Academy of Neurology (M.D.O.B., K.B.-D., H.M.S.), Minneapolis, MN; and Centre Hospitalier de l'Université de Montréal Research Centre (CRCHUM) (M.R.K.), Quebec, Canada
| | - Heather M Silsbee
- From the Department of Neurology (A.M.P.), Columbia University, New York City; Departments of Pediatrics and Neurology & Neurosurgery (M.O.), McGill University, Montreal, Quebec, Canada; Departments of Neurology (S.W.R.), Biomedical Engineering (S.W.R.), and Obstetrics and Gynecology (S.S.O.), Vanderbilt University Medical Center, Nashville, TN; Northern Michigan Neurology and Munson Medical Center (D.K.D.), Traverse City, MI; Department of Neurology (J.F.), NYU Grossman School of Medicine, New York City; Feinberg School of Medicine (E.E.G.), Northwestern University, Chicago, IL; The NeuroMedical Center (D.G.), Baton Rouge, LA; Epilepsy Foundation (W.R.M.), Bowie, MD; Department of Neurology (H.M.M.C.), Wake Forest University School of Medicine, Winston-Salem, NC; My Epilepsy Story (B.M.), Nashville, TN; Institute of Clinical Neurosciences (K.P.), Royal Prince Alfred Hospital, Sydney, Australia; Department of Neurology (P.B.P.), University of Pittsburgh School of Medicine, PA; Department of Ob-Gyn (G.S.), Eastern Virginia Medical School, Norfolk; Department of Neurology (D.B.S.), University of Colorado School of Medicine, Aurora; Department of Biostatistics, Epidemiology, and Environmental Health Sciences (K.S.), Jiann-Ping Hsu College of Public Health, Georgia Southern University, Statesboro; Department of Neurology (S.V.T.), Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India; Department of Clinical Neuroscience (T.T.), Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden; American Academy of Neurology (M.D.O.B., K.B.-D., H.M.S.), Minneapolis, MN; and Centre Hospitalier de l'Université de Montréal Research Centre (CRCHUM) (M.R.K.), Quebec, Canada
| | - Mark R Keezer
- From the Department of Neurology (A.M.P.), Columbia University, New York City; Departments of Pediatrics and Neurology & Neurosurgery (M.O.), McGill University, Montreal, Quebec, Canada; Departments of Neurology (S.W.R.), Biomedical Engineering (S.W.R.), and Obstetrics and Gynecology (S.S.O.), Vanderbilt University Medical Center, Nashville, TN; Northern Michigan Neurology and Munson Medical Center (D.K.D.), Traverse City, MI; Department of Neurology (J.F.), NYU Grossman School of Medicine, New York City; Feinberg School of Medicine (E.E.G.), Northwestern University, Chicago, IL; The NeuroMedical Center (D.G.), Baton Rouge, LA; Epilepsy Foundation (W.R.M.), Bowie, MD; Department of Neurology (H.M.M.C.), Wake Forest University School of Medicine, Winston-Salem, NC; My Epilepsy Story (B.M.), Nashville, TN; Institute of Clinical Neurosciences (K.P.), Royal Prince Alfred Hospital, Sydney, Australia; Department of Neurology (P.B.P.), University of Pittsburgh School of Medicine, PA; Department of Ob-Gyn (G.S.), Eastern Virginia Medical School, Norfolk; Department of Neurology (D.B.S.), University of Colorado School of Medicine, Aurora; Department of Biostatistics, Epidemiology, and Environmental Health Sciences (K.S.), Jiann-Ping Hsu College of Public Health, Georgia Southern University, Statesboro; Department of Neurology (S.V.T.), Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India; Department of Clinical Neuroscience (T.T.), Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden; American Academy of Neurology (M.D.O.B., K.B.-D., H.M.S.), Minneapolis, MN; and Centre Hospitalier de l'Université de Montréal Research Centre (CRCHUM) (M.R.K.), Quebec, Canada
| |
Collapse
|
25
|
Mkhitaryan M, Avetisyan T, Mkhoyan A, Avetisyan L, Yenkoyan K. A case-control study on pre-, peri-, and neonatal risk factors associated with autism spectrum disorder among Armenian children. Sci Rep 2024; 14:12308. [PMID: 38811666 PMCID: PMC11137108 DOI: 10.1038/s41598-024-63240-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 05/27/2024] [Indexed: 05/31/2024] Open
Abstract
We aimed to investigate the role of pre-, peri- and neonatal risk factors in the development of autism spectrum disorder (ASD) among Armenian children with the goal of detecting and addressing modifiable risk factors to reduce ASD incidence. For this purpose a retrospective case-control study using a random proportional sample of Armenian children with ASD to assess associations between various factors and ASD was conducted. The study was approved by the local ethical committee, and parental written consent was obtained. A total of 168 children with ASD and 329 controls were included in the analysis. Multivariable logistic regression analysis revealed that male gender, maternal weight gain, use of MgB6, self-reported stress during the pregnancy, pregnancy with complications, as well as use of labor-inducing drugs were associated with a significant increase in the odds of ASD, whereas Duphaston use during pregnancy, the longer interpregnancy interval and birth height were associated with decreased odds of ASD. These findings are pertinent as many identified factors may be preventable or modifiable, underscoring the importance of timely and appropriate public health strategies aimed at disease prevention in pregnant women to reduce ASD incidence.
Collapse
Affiliation(s)
- Meri Mkhitaryan
- Neuroscience Laboratory, Cobrain Center, Yerevan State Medical University Named After M. Heratsi, 0025, Yerevan, Armenia
| | - Tamara Avetisyan
- Cobrain Center, Yerevan State Medical University Named After M. Heratsi, 0025, Yerevan, Armenia
- Muratsan University Hospital Complex, Yerevan State Medical University Named After M. Heratsi, 0075, Yerevan, Armenia
| | - Anna Mkhoyan
- Department of Infectious Diseases, Yerevan State Medical University Named After M. Heratsi, 0025, Yerevan, Armenia
| | - Larisa Avetisyan
- Cobrain Center, Yerevan State Medical University Named After M. Heratsi, 0025, Yerevan, Armenia
- Department of Hygiene, Yerevan State Medical University Named After M. Heratsi, 0025, Yerevan, Armenia
| | - Konstantin Yenkoyan
- Neuroscience Laboratory, Cobrain Center, Yerevan State Medical University Named After M. Heratsi, 0025, Yerevan, Armenia.
| |
Collapse
|
26
|
Kosmer K, Kulesza R. Cortical dysmorphology and reduced cortico-collicular projections in an animal model of autism spectrum disorder. Cereb Cortex 2024; 34:146-160. [PMID: 38696608 PMCID: PMC11484449 DOI: 10.1093/cercor/bhad501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/21/2023] [Accepted: 12/06/2023] [Indexed: 05/04/2024] Open
Abstract
Autism spectrum disorder is a neurodevelopmental disability that includes sensory disturbances. Hearing is frequently affected and ranges from deafness to hypersensitivity. In utero exposure to the antiepileptic valproic acid is associated with increased risk of autism spectrum disorder in humans and timed valproic acid exposure is a biologically relevant and validated animal model of autism spectrum disorder. Valproic acid-exposed rats have fewer neurons in their auditory brainstem and thalamus, fewer calbindin-positive neurons, reduced ascending projections to the midbrain and thalamus, elevated thresholds, and delayed auditory brainstem responses. Additionally, in the auditory cortex, valproic acid exposure results in abnormal responses, decreased phase-locking, elevated thresholds, and abnormal tonotopic maps. We therefore hypothesized that in utero, valproic acid exposure would result in fewer neurons in auditory cortex, neuronal dysmorphology, fewer calbindin-positive neurons, and reduced connectivity. We approached this hypothesis using morphometric analyses, immunohistochemistry, and retrograde tract tracing. We found thinner cortical layers but no changes in the density of neurons, smaller pyramidal and non-pyramidal neurons in several regions, fewer neurons immunoreactive for calbindin-positive, and fewer cortical neurons projecting to the inferior colliculus. These results support the widespread impact of the auditory system in autism spectrum disorder and valproic acid-exposed animals and emphasize the utility of simple, noninvasive auditory screening for autism spectrum disorder.
Collapse
Affiliation(s)
- Kara Kosmer
- RWJBH Monmouth Medical CenterLong Branch, NJ 07740, United States
| | - Randy Kulesza
- Department of Anatomy, Lake Erie College of Osteopathic Medicine, Erie, PA 16509, United States
| |
Collapse
|
27
|
Honybun E, Cockle E, Malpas CB, O'Brien TJ, Vajda FJ, Perucca P, Rayner G. Neurodevelopmental and Functional Outcomes Following In Utero Exposure to Antiseizure Medication: A Systematic Review. Neurology 2024; 102:e209175. [PMID: 38531021 DOI: 10.1212/wnl.0000000000209175] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 12/04/2023] [Indexed: 03/28/2024] Open
Abstract
BACKGROUND AND OBJECTIVES To undertake a systematic review of the available literature to examine the relationship between prenatal antiseizure medication (ASM) exposure and adverse postnatal neurodevelopmental outcomes, focusing on social, emotional, behavioral, and adaptive domains of human function, and the frequency of neurodevelopmental and psychiatric disorders in ASM-exposed offspring. METHODS Electronic searches of MEDLINE, PsychINFO, and EMBASE were conducted and limited to studies published between 1990 and 2023 in English. Studies were eligible if they prospectively or retrospectively reported neurodevelopmental outcomes of ASM-exposed offspring. The Newcastle-Ottawa scale was used to conduct methodologic quality assessments of included studies, and a narrative synthesis integrated the review findings. RESULTS Forty-three studies were included. Valproate has been consistently associated with a 2- to 4-fold increased risk of autism spectrum disorder (ASD), 2- to 5-fold increased risk of intellectual disability (ID), and poor adaptive functioning. Growing evidence indicates that topiramate is associated with a 2-fold increased risk of ASD and 3- to 4-fold increased risk of ID. The risks of adverse neurodevelopmental outcomes for valproate and topiramate seem to be dose dependent. Phenobarbital has been suggested to be associated with deleterious neurodevelopmental effects, but data are limited. Levetiracetam has recently been linked with an increased risk of attention deficit hyperactivity disorder and anxiety disorders in a single study. Carbamazepine has been associated with variable neurodevelopmental outcomes. Lamotrigine seems to be "safe" in terms of postnatal neurodevelopment. Data for oxcarbazepine, phenytoin, and clonazepam are limited but seem to have little-to-no risk of adverse outcomes. Evidence for the remaining ASMs, including gabapentin, pregabalin, lacosamide, zonisamide, clobazam, perampanel, ethosuximide, or brivaracetam, is lacking. Several methodologic limitations impeded data synthesis, including heterogeneity in outcome measures and small samples of monotherapy exposures. DISCUSSION The findings of this review support the conclusion that valproate and topiramate use during pregnancy is associated with a significantly increased risk of neurodevelopmental effects on the fetus. Apart from lamotrigine, which seems to be free of adverse neurodevelopmental effects, data for the other ASMs are mixed or inadequate to draw definite conclusions. Further research into the neurodevelopmental effects of prenatal exposure to ASMs, including most newer agents, is much needed.
Collapse
Affiliation(s)
- Eliza Honybun
- From the Melbourne School of Psychological Sciences (E.H., C.B.M., G.R.), The University of Melbourne; Epilepsy Research Centre (E.H., P.P., G.R.), Department of Medicine, Austin Hospital, The University of Melbourne; Department of Neuroscience (E.C., C.B.M., T.J.O.B., F.J.V., P.P., G.R.), Central Clinical School, Monash University; Department of Neurology (E.C., C.B.M., T.J.O.B., P.P., G.R.), The Alfred Hospital; Department of Neurology (C.B.M., T.J.O.B., F.J.V., P.P.), Royal Melbourne Hospital; Department of Medicine (C.B.M., T.J.O.B., F.J.V.), Royal Melbourne Hospital, The University of Melbourne; Bladin-Berkovic Comprehensive Epilepsy Program (P.P.), Department of Neurology, Austin Health; and Department of Clinical Neuropsychology (G.R.), Austin Health, Melbourne, Australia
| | - Emily Cockle
- From the Melbourne School of Psychological Sciences (E.H., C.B.M., G.R.), The University of Melbourne; Epilepsy Research Centre (E.H., P.P., G.R.), Department of Medicine, Austin Hospital, The University of Melbourne; Department of Neuroscience (E.C., C.B.M., T.J.O.B., F.J.V., P.P., G.R.), Central Clinical School, Monash University; Department of Neurology (E.C., C.B.M., T.J.O.B., P.P., G.R.), The Alfred Hospital; Department of Neurology (C.B.M., T.J.O.B., F.J.V., P.P.), Royal Melbourne Hospital; Department of Medicine (C.B.M., T.J.O.B., F.J.V.), Royal Melbourne Hospital, The University of Melbourne; Bladin-Berkovic Comprehensive Epilepsy Program (P.P.), Department of Neurology, Austin Health; and Department of Clinical Neuropsychology (G.R.), Austin Health, Melbourne, Australia
| | - Charles B Malpas
- From the Melbourne School of Psychological Sciences (E.H., C.B.M., G.R.), The University of Melbourne; Epilepsy Research Centre (E.H., P.P., G.R.), Department of Medicine, Austin Hospital, The University of Melbourne; Department of Neuroscience (E.C., C.B.M., T.J.O.B., F.J.V., P.P., G.R.), Central Clinical School, Monash University; Department of Neurology (E.C., C.B.M., T.J.O.B., P.P., G.R.), The Alfred Hospital; Department of Neurology (C.B.M., T.J.O.B., F.J.V., P.P.), Royal Melbourne Hospital; Department of Medicine (C.B.M., T.J.O.B., F.J.V.), Royal Melbourne Hospital, The University of Melbourne; Bladin-Berkovic Comprehensive Epilepsy Program (P.P.), Department of Neurology, Austin Health; and Department of Clinical Neuropsychology (G.R.), Austin Health, Melbourne, Australia
| | - Terence J O'Brien
- From the Melbourne School of Psychological Sciences (E.H., C.B.M., G.R.), The University of Melbourne; Epilepsy Research Centre (E.H., P.P., G.R.), Department of Medicine, Austin Hospital, The University of Melbourne; Department of Neuroscience (E.C., C.B.M., T.J.O.B., F.J.V., P.P., G.R.), Central Clinical School, Monash University; Department of Neurology (E.C., C.B.M., T.J.O.B., P.P., G.R.), The Alfred Hospital; Department of Neurology (C.B.M., T.J.O.B., F.J.V., P.P.), Royal Melbourne Hospital; Department of Medicine (C.B.M., T.J.O.B., F.J.V.), Royal Melbourne Hospital, The University of Melbourne; Bladin-Berkovic Comprehensive Epilepsy Program (P.P.), Department of Neurology, Austin Health; and Department of Clinical Neuropsychology (G.R.), Austin Health, Melbourne, Australia
| | - Frank J Vajda
- From the Melbourne School of Psychological Sciences (E.H., C.B.M., G.R.), The University of Melbourne; Epilepsy Research Centre (E.H., P.P., G.R.), Department of Medicine, Austin Hospital, The University of Melbourne; Department of Neuroscience (E.C., C.B.M., T.J.O.B., F.J.V., P.P., G.R.), Central Clinical School, Monash University; Department of Neurology (E.C., C.B.M., T.J.O.B., P.P., G.R.), The Alfred Hospital; Department of Neurology (C.B.M., T.J.O.B., F.J.V., P.P.), Royal Melbourne Hospital; Department of Medicine (C.B.M., T.J.O.B., F.J.V.), Royal Melbourne Hospital, The University of Melbourne; Bladin-Berkovic Comprehensive Epilepsy Program (P.P.), Department of Neurology, Austin Health; and Department of Clinical Neuropsychology (G.R.), Austin Health, Melbourne, Australia
| | - Piero Perucca
- From the Melbourne School of Psychological Sciences (E.H., C.B.M., G.R.), The University of Melbourne; Epilepsy Research Centre (E.H., P.P., G.R.), Department of Medicine, Austin Hospital, The University of Melbourne; Department of Neuroscience (E.C., C.B.M., T.J.O.B., F.J.V., P.P., G.R.), Central Clinical School, Monash University; Department of Neurology (E.C., C.B.M., T.J.O.B., P.P., G.R.), The Alfred Hospital; Department of Neurology (C.B.M., T.J.O.B., F.J.V., P.P.), Royal Melbourne Hospital; Department of Medicine (C.B.M., T.J.O.B., F.J.V.), Royal Melbourne Hospital, The University of Melbourne; Bladin-Berkovic Comprehensive Epilepsy Program (P.P.), Department of Neurology, Austin Health; and Department of Clinical Neuropsychology (G.R.), Austin Health, Melbourne, Australia
| | - Genevieve Rayner
- From the Melbourne School of Psychological Sciences (E.H., C.B.M., G.R.), The University of Melbourne; Epilepsy Research Centre (E.H., P.P., G.R.), Department of Medicine, Austin Hospital, The University of Melbourne; Department of Neuroscience (E.C., C.B.M., T.J.O.B., F.J.V., P.P., G.R.), Central Clinical School, Monash University; Department of Neurology (E.C., C.B.M., T.J.O.B., P.P., G.R.), The Alfred Hospital; Department of Neurology (C.B.M., T.J.O.B., F.J.V., P.P.), Royal Melbourne Hospital; Department of Medicine (C.B.M., T.J.O.B., F.J.V.), Royal Melbourne Hospital, The University of Melbourne; Bladin-Berkovic Comprehensive Epilepsy Program (P.P.), Department of Neurology, Austin Health; and Department of Clinical Neuropsychology (G.R.), Austin Health, Melbourne, Australia
| |
Collapse
|
28
|
Mazzone PP, Hogg KM, Weir CJ, Stephen J, Bhattacharya S, Richer S, Chin RFM. Comparison of neurodevelopmental, educational and adult socioeconomic outcomes in offspring of women with and without epilepsy: A systematic review and meta-analysis. Seizure 2024; 117:213-221. [PMID: 38484631 DOI: 10.1016/j.seizure.2024.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 02/21/2024] [Accepted: 02/21/2024] [Indexed: 05/01/2024] Open
Abstract
BACKGROUND Adequate pre-pregnancy counselling and education planning are essential to improve outcomes for offspring of women with epilepsy (OWWE). The current systematic review and meta-analysis aimed to compare outcomes for OWWE and offspring of women without epilepsy (OWWoE). METHODS We conducted a systematic review and meta-analysis. We searched MEDLINE, EMBASE, CINAHL, PsycINFO (database inception-1st January 2023), OpenGrey, GoogleScholar, and hand-searched journals and reference lists of included studies to identify eligible studies. We placed no language restrictions and included observational studies concerning OWWE and OWWoE. We followed the PRIMSA checklist for abstracting data. The Newcastle-Ottawa Scale for risk of bias assessment was conducted independently by two authors with mediation by a third. We report pooled unadjusted odds ratios (OR) or mean differences (MD) with 95% confidence intervals (95CI) from random (I2>50%) or fixed (I2<50%) effects meta-analyses. Outcomes of interest included offspring autism, attention deficit/hyperactive disorder, intellectual disability, epilepsy, developmental disorder, intelligence, educational, and adulthood socioeconomic outcomes. RESULTS Of 10,928 articles identified, we included 21 in meta-analyses. OWWE had increased odds of autism (2 articles, 4,502,098 offspring) OR [95CI] 1·67 [1·54, 1·82], attention-deficit/hyperactivity disorder (3 articles, 957,581 offspring) 1·59 [1·44, 1·76], intellectual disability (2 articles, 4,501,786 children) 2·37 [2·13, 2·65], having special educational needs (3 articles, 1,308,919 children) 2·60 [1·07, 6·34]. OWWE had worse mean scores for full-scale intelligence (5 articles, 989 children) -6·05 [-10·31, -1·79]. No studies were identified that investigated adulthood socioeconomic outcomes. CONCLUSIONS Increased odds of poor outcomes are higher with greater anti-seizure medication burden including neurodevelopmental and educational outcomes. In fact, these two outcomes seem to be worse in OWWE compared to OWWoE, even if there was no ASM exposure during pregnancy, but further work is needed to take into account potential confounding factors.
Collapse
Affiliation(s)
- Paolo Pierino Mazzone
- Muir Maxwell Epilepsy Centre, University of Edinburgh, Edinburgh, United Kingdom; Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom; Child Life and Health, Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom.
| | | | - Christopher J Weir
- Muir Maxwell Epilepsy Centre, University of Edinburgh, Edinburgh, United Kingdom; Edinburgh Clinical Trials Unit, Usher Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Jacqueline Stephen
- Edinburgh Clinical Trials Unit, Usher Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Sohinee Bhattacharya
- Aberdeen Centre for Women's Health Research, The Institute of Applied Health Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Simone Richer
- The University of Edinburgh, Medical School, Edinburgh, United Kingdom
| | - Richard F M Chin
- Muir Maxwell Epilepsy Centre, University of Edinburgh, Edinburgh, United Kingdom; Royal Hospital for Children and Young People, Edinburgh, United Kingdom; Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom; Child Life and Health, Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
29
|
Salari Z, Moslemizadeh A, Tezerji SS, Sabet N, Parizi AS, Khaksari M, Sheibani V, Jafari E, Shafieipour S, Bashiri H. Sex-dependent alterations of inflammatory factors, oxidative stress, and histopathology of the brain-gut axis in a VPA-induced autistic-like model of rats. Birth Defects Res 2024; 116:e2310. [PMID: 38563145 DOI: 10.1002/bdr2.2310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 12/23/2023] [Accepted: 01/21/2024] [Indexed: 04/04/2024]
Abstract
INTRODUCTION In this study, we aimed to investigate the inflammatory factors, oxidative stress, and histopathological consequences of the brain-gut axis in male and female rats prenatally exposed to VPA. METHODS Pregnant Wistar rats were randomly divided into two groups. The animals received saline, and valproic acid (VPA) (600 mg/kg, i.p.) on embryonic day 12.5 (E12.5). All offspring were weaned on postnatal day 21, and the experiments were done in male and female rats on day 60. The brain and intestine tissues were extracted to assess histopathology, inflammation, and oxidative stress. RESULTS An increase of interleukin-1β (IL-1β) and interleukin-6 (IL-6) and a decrease of interleukin-10 (IL-10) were observed in the two sexes and two tissues of the autistic rats. In the VPA-exposed animals, malondialdehyde (MDA) and protein carbonyl (PC) increased in the brain of both sexes and the intestines of only the males. The total antioxidant capacity (TAC), superoxide dismutase (SOD), and catalase (CAT) significantly decreased in both tissues of male and female autistic groups. Histopathological evaluation showed that the %apoptosis of the cortex in the autistic male and female groups was more than in controls whereas this parameter in the CA1 and CA3 was significant only in the male rats. In the intestine, histopathologic changes were seen only in the male autistic animals. CONCLUSION The inflammatory and antioxidant factors were in line in the brain-gut axis in male and female rats prenatally exposed to VPA. Histopathological consequences were more significant in the VPA-exposed male animals.
Collapse
Affiliation(s)
- Zahra Salari
- Gastroenterology and Hepatology Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Sara Sheibani Tezerji
- Department of Behavioural and Molecular Neurobiology, Regensburg Center for Neuroscience, University of Regensburg, Regensburg, Germany
| | - Nazanin Sabet
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Ali Saeidpour Parizi
- Gastroenterology and Hepatology Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Khaksari
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Vahid Sheibani
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
- Department of Physiology and Pharmacology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Elham Jafari
- Pathology and Stem Cells Research Center, Department of Pathology, Afzalipour School of Medicine, Kerman University of Medical Science, Kerman, Iran
| | - Sara Shafieipour
- Gastroenterology and Hepatology Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Hamideh Bashiri
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
- Department of Physiology and Pharmacology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
30
|
Hernández-Díaz S, Straub L, Bateman BT, Zhu Y, Mogun H, Wisner KL, Gray KJ, Lester B, McDougle CJ, DiCesare E, Pennell PB, Huybrechts KF. Risk of Autism after Prenatal Topiramate, Valproate, or Lamotrigine Exposure. N Engl J Med 2024; 390:1069-1079. [PMID: 38507750 PMCID: PMC11047762 DOI: 10.1056/nejmoa2309359] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
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.).
Collapse
Affiliation(s)
- Sonia Hernández-Díaz
- From the Department of Epidemiology, Harvard T.H. Chan School of Public Health (S.H.-D.), the Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School (L.S., Y.Z., H.M., E.D., K.F.H.), the Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Brigham and Women's Hospital (K.J.G.), and the Department of Psychiatry, Harvard Medical School (C.J.M.), Boston, and the Lurie Center for Autism, Massachusetts General Hospital, Lexington (C.J.M.) - all in Massachusetts; the Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA (B.T.B.); the Asher Center for the Study and Treatment of Depressive Disorders, Department of Psychiatry and Behavioral Sciences, and the Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago (K.L.W.); the Center for the Study of Children at Risk, Departments of Psychiatry and Pediatrics, Alpert Medical School of Brown University, and Women and Infants Hospital, Providence, RI (B.L.); and the Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh (P.B.P.)
| | - Loreen Straub
- From the Department of Epidemiology, Harvard T.H. Chan School of Public Health (S.H.-D.), the Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School (L.S., Y.Z., H.M., E.D., K.F.H.), the Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Brigham and Women's Hospital (K.J.G.), and the Department of Psychiatry, Harvard Medical School (C.J.M.), Boston, and the Lurie Center for Autism, Massachusetts General Hospital, Lexington (C.J.M.) - all in Massachusetts; the Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA (B.T.B.); the Asher Center for the Study and Treatment of Depressive Disorders, Department of Psychiatry and Behavioral Sciences, and the Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago (K.L.W.); the Center for the Study of Children at Risk, Departments of Psychiatry and Pediatrics, Alpert Medical School of Brown University, and Women and Infants Hospital, Providence, RI (B.L.); and the Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh (P.B.P.)
| | - Brian T Bateman
- From the Department of Epidemiology, Harvard T.H. Chan School of Public Health (S.H.-D.), the Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School (L.S., Y.Z., H.M., E.D., K.F.H.), the Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Brigham and Women's Hospital (K.J.G.), and the Department of Psychiatry, Harvard Medical School (C.J.M.), Boston, and the Lurie Center for Autism, Massachusetts General Hospital, Lexington (C.J.M.) - all in Massachusetts; the Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA (B.T.B.); the Asher Center for the Study and Treatment of Depressive Disorders, Department of Psychiatry and Behavioral Sciences, and the Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago (K.L.W.); the Center for the Study of Children at Risk, Departments of Psychiatry and Pediatrics, Alpert Medical School of Brown University, and Women and Infants Hospital, Providence, RI (B.L.); and the Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh (P.B.P.)
| | - Yanmin Zhu
- From the Department of Epidemiology, Harvard T.H. Chan School of Public Health (S.H.-D.), the Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School (L.S., Y.Z., H.M., E.D., K.F.H.), the Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Brigham and Women's Hospital (K.J.G.), and the Department of Psychiatry, Harvard Medical School (C.J.M.), Boston, and the Lurie Center for Autism, Massachusetts General Hospital, Lexington (C.J.M.) - all in Massachusetts; the Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA (B.T.B.); the Asher Center for the Study and Treatment of Depressive Disorders, Department of Psychiatry and Behavioral Sciences, and the Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago (K.L.W.); the Center for the Study of Children at Risk, Departments of Psychiatry and Pediatrics, Alpert Medical School of Brown University, and Women and Infants Hospital, Providence, RI (B.L.); and the Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh (P.B.P.)
| | - Helen Mogun
- From the Department of Epidemiology, Harvard T.H. Chan School of Public Health (S.H.-D.), the Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School (L.S., Y.Z., H.M., E.D., K.F.H.), the Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Brigham and Women's Hospital (K.J.G.), and the Department of Psychiatry, Harvard Medical School (C.J.M.), Boston, and the Lurie Center for Autism, Massachusetts General Hospital, Lexington (C.J.M.) - all in Massachusetts; the Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA (B.T.B.); the Asher Center for the Study and Treatment of Depressive Disorders, Department of Psychiatry and Behavioral Sciences, and the Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago (K.L.W.); the Center for the Study of Children at Risk, Departments of Psychiatry and Pediatrics, Alpert Medical School of Brown University, and Women and Infants Hospital, Providence, RI (B.L.); and the Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh (P.B.P.)
| | - Katherine L Wisner
- From the Department of Epidemiology, Harvard T.H. Chan School of Public Health (S.H.-D.), the Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School (L.S., Y.Z., H.M., E.D., K.F.H.), the Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Brigham and Women's Hospital (K.J.G.), and the Department of Psychiatry, Harvard Medical School (C.J.M.), Boston, and the Lurie Center for Autism, Massachusetts General Hospital, Lexington (C.J.M.) - all in Massachusetts; the Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA (B.T.B.); the Asher Center for the Study and Treatment of Depressive Disorders, Department of Psychiatry and Behavioral Sciences, and the Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago (K.L.W.); the Center for the Study of Children at Risk, Departments of Psychiatry and Pediatrics, Alpert Medical School of Brown University, and Women and Infants Hospital, Providence, RI (B.L.); and the Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh (P.B.P.)
| | - Kathryn J Gray
- From the Department of Epidemiology, Harvard T.H. Chan School of Public Health (S.H.-D.), the Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School (L.S., Y.Z., H.M., E.D., K.F.H.), the Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Brigham and Women's Hospital (K.J.G.), and the Department of Psychiatry, Harvard Medical School (C.J.M.), Boston, and the Lurie Center for Autism, Massachusetts General Hospital, Lexington (C.J.M.) - all in Massachusetts; the Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA (B.T.B.); the Asher Center for the Study and Treatment of Depressive Disorders, Department of Psychiatry and Behavioral Sciences, and the Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago (K.L.W.); the Center for the Study of Children at Risk, Departments of Psychiatry and Pediatrics, Alpert Medical School of Brown University, and Women and Infants Hospital, Providence, RI (B.L.); and the Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh (P.B.P.)
| | - Barry Lester
- From the Department of Epidemiology, Harvard T.H. Chan School of Public Health (S.H.-D.), the Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School (L.S., Y.Z., H.M., E.D., K.F.H.), the Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Brigham and Women's Hospital (K.J.G.), and the Department of Psychiatry, Harvard Medical School (C.J.M.), Boston, and the Lurie Center for Autism, Massachusetts General Hospital, Lexington (C.J.M.) - all in Massachusetts; the Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA (B.T.B.); the Asher Center for the Study and Treatment of Depressive Disorders, Department of Psychiatry and Behavioral Sciences, and the Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago (K.L.W.); the Center for the Study of Children at Risk, Departments of Psychiatry and Pediatrics, Alpert Medical School of Brown University, and Women and Infants Hospital, Providence, RI (B.L.); and the Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh (P.B.P.)
| | - Christopher J McDougle
- From the Department of Epidemiology, Harvard T.H. Chan School of Public Health (S.H.-D.), the Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School (L.S., Y.Z., H.M., E.D., K.F.H.), the Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Brigham and Women's Hospital (K.J.G.), and the Department of Psychiatry, Harvard Medical School (C.J.M.), Boston, and the Lurie Center for Autism, Massachusetts General Hospital, Lexington (C.J.M.) - all in Massachusetts; the Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA (B.T.B.); the Asher Center for the Study and Treatment of Depressive Disorders, Department of Psychiatry and Behavioral Sciences, and the Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago (K.L.W.); the Center for the Study of Children at Risk, Departments of Psychiatry and Pediatrics, Alpert Medical School of Brown University, and Women and Infants Hospital, Providence, RI (B.L.); and the Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh (P.B.P.)
| | - Elyse DiCesare
- From the Department of Epidemiology, Harvard T.H. Chan School of Public Health (S.H.-D.), the Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School (L.S., Y.Z., H.M., E.D., K.F.H.), the Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Brigham and Women's Hospital (K.J.G.), and the Department of Psychiatry, Harvard Medical School (C.J.M.), Boston, and the Lurie Center for Autism, Massachusetts General Hospital, Lexington (C.J.M.) - all in Massachusetts; the Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA (B.T.B.); the Asher Center for the Study and Treatment of Depressive Disorders, Department of Psychiatry and Behavioral Sciences, and the Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago (K.L.W.); the Center for the Study of Children at Risk, Departments of Psychiatry and Pediatrics, Alpert Medical School of Brown University, and Women and Infants Hospital, Providence, RI (B.L.); and the Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh (P.B.P.)
| | - Page B Pennell
- From the Department of Epidemiology, Harvard T.H. Chan School of Public Health (S.H.-D.), the Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School (L.S., Y.Z., H.M., E.D., K.F.H.), the Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Brigham and Women's Hospital (K.J.G.), and the Department of Psychiatry, Harvard Medical School (C.J.M.), Boston, and the Lurie Center for Autism, Massachusetts General Hospital, Lexington (C.J.M.) - all in Massachusetts; the Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA (B.T.B.); the Asher Center for the Study and Treatment of Depressive Disorders, Department of Psychiatry and Behavioral Sciences, and the Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago (K.L.W.); the Center for the Study of Children at Risk, Departments of Psychiatry and Pediatrics, Alpert Medical School of Brown University, and Women and Infants Hospital, Providence, RI (B.L.); and the Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh (P.B.P.)
| | - Krista F Huybrechts
- From the Department of Epidemiology, Harvard T.H. Chan School of Public Health (S.H.-D.), the Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School (L.S., Y.Z., H.M., E.D., K.F.H.), the Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Brigham and Women's Hospital (K.J.G.), and the Department of Psychiatry, Harvard Medical School (C.J.M.), Boston, and the Lurie Center for Autism, Massachusetts General Hospital, Lexington (C.J.M.) - all in Massachusetts; the Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA (B.T.B.); the Asher Center for the Study and Treatment of Depressive Disorders, Department of Psychiatry and Behavioral Sciences, and the Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago (K.L.W.); the Center for the Study of Children at Risk, Departments of Psychiatry and Pediatrics, Alpert Medical School of Brown University, and Women and Infants Hospital, Providence, RI (B.L.); and the Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh (P.B.P.)
| |
Collapse
|
31
|
Hung LY, Margolis KG. Autism spectrum disorders and the gastrointestinal tract: insights into mechanisms and clinical relevance. Nat Rev Gastroenterol Hepatol 2024; 21:142-163. [PMID: 38114585 DOI: 10.1038/s41575-023-00857-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/11/2023] [Indexed: 12/21/2023]
Abstract
Autism spectrum disorders (ASDs) are recognized as central neurodevelopmental disorders diagnosed by impairments in social interactions, communication and repetitive behaviours. The recognition of ASD as a central nervous system (CNS)-mediated neurobehavioural disorder has led most of the research in ASD to be focused on the CNS. However, gastrointestinal function is also likely to be affected owing to the neural mechanistic nature of ASD and the nervous system in the gastrointestinal tract (enteric nervous system). Thus, it is unsurprising that gastrointestinal disorders, particularly constipation, diarrhoea and abdominal pain, are highly comorbid in individuals with ASD. Gastrointestinal problems have also been repeatedly associated with increased severity of the core symptoms diagnostic of ASD and other centrally mediated comorbid conditions, including psychiatric issues, irritability, rigid-compulsive behaviours and aggression. Despite the high prevalence of gastrointestinal dysfunction in ASD and its associated behavioural comorbidities, the specific links between these two conditions have not been clearly delineated, and current data linking ASD to gastrointestinal dysfunction have not been extensively reviewed. This Review outlines the established and emerging clinical and preclinical evidence that emphasizes the gut as a novel mechanistic and potential therapeutic target for individuals with ASD.
Collapse
Affiliation(s)
- Lin Y Hung
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, USA
| | - Kara Gross Margolis
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, USA.
- Department of Cell Biology, NYU Grossman School of Medicine and Langone Medical Center, New York, NY, USA.
- Department of Pediatrics, NYU Grossman School of Medicine and Langone Medical Center, New York, NY, USA.
| |
Collapse
|
32
|
Peron A, Picot C, Jurek L, Nourredine M, Ripoche E, Ajiji P, Cucherat M, Cottin J. Neurodevelopmental outcomes after prenatal exposure to lamotrigine monotherapy in women with epilepsy: a systematic review and meta-analysis. BMC Pregnancy Childbirth 2024; 24:103. [PMID: 38308208 PMCID: PMC10835851 DOI: 10.1186/s12884-023-06242-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 12/31/2023] [Indexed: 02/04/2024] Open
Abstract
BACKGROUND Lamotrigine has become one of the most commonly prescribed antiseizure medications (ASM) in epileptic women during pregnancy and therefore requires regular updates regarding its safety. The aim of this study was to estimate the association between in utero exposure to lamotrigine monotherapy and the occurrence of neurodevelopmental outcomes. METHODS All comparative studies assessing the occurrence of neurodevelopmental outcomes after epilepsy-indicated lamotrigine monotherapy exposure during pregnancy were searched. First, references were identified through a snowballing approach, then, through electronic databases (Medline and Embase) from 2015 to June 2022. One investigator evaluated study eligibility and extracted data and a second independent investigator reviewed the meta-analysis (MA). A systematic review and random-effects model approach were performed using a collaborative WEB-based meta-analysis platform (metaPreg.org) with a registered protocol (osf.io/u4gva). RESULTS Overall, 18 studies were included. For outcomes reported by at least 4 studies, the pooled odds ratios and 95% confidence interval obtained with the number of exposed (N1) and unexposed children (N0) included were: neurodevelopmental disorders as a whole 0.84 [0.66;1.06] (N1 = 5,271; N0 = 22,230); language disorders or delay 1.16 [0.67;2.00] (N1 = 313; N0 = 506); diagnosis or risk of ASD 0.97 [0.61;1.53] (N1 = at least 5,262; N0 = 33,313); diagnosis or risk of ADHD 1.14 [0.75;1.72] (N1 = at least 113; N0 = 11,530) and psychomotor developmental disorders or delay 2.68 [1.29-5.56] (N1 = 163; N0 = 220). The MA of cognitive outcomes included less than 4 studies and retrieved a significant association for infants exposed to lamotrigine younger than 3 years old but not in the older age groups. CONCLUSION Prenatal exposure to lamotrigine monotherapy is not found to be statistically associated with neurodevelopmental disorders as a whole, language disorders or delay, diagnosis or risk of ASD and diagnosis or risk of ADHD. However, the MA found an increased risk of psychomotor developmental disorders or delay and cognitive developmental delay in less than 3 years old children. Nevertheless, these findings were based exclusively on observational studies presenting biases and on a limited number of included children. More studies should assess neurodevelopmental outcomes in children prenatally exposed to lamotrigine.
Collapse
Affiliation(s)
- Audrey Peron
- Service Hospitalo-Universitaire de Pharmacotoxicologie de Lyon, Hospices Civils de Lyon, Bât. A-162, avenue Lacassagne, Lyon Cedex 03, 69424, France
| | - Cyndie Picot
- Service Hospitalo-Universitaire de Pharmacotoxicologie de Lyon, Hospices Civils de Lyon, Bât. A-162, avenue Lacassagne, Lyon Cedex 03, 69424, France
| | - Lucie Jurek
- Pôle de psychiatrie de l'Enfant et l'Adolescent, Centre Hospitalier Le Vinatier, Bron, France
| | - Mikaïl Nourredine
- Service Hospitalo-Universitaire de Pharmacotoxicologie de Lyon, Hospices Civils de Lyon, Bât. A-162, avenue Lacassagne, Lyon Cedex 03, 69424, France
- Service Universitaire d'Addictologie de Lyon, Centre Hospitalier Le Vinatier, Bron, France
| | - Emmanuelle Ripoche
- Adverse Events and Incidents Department-Surveillance Division, Agence nationale de sécurité du médicament et des produits de santé (ANSM), Saint Denis, France
| | - Priscilla Ajiji
- Adverse Events and Incidents Department-Surveillance Division, Agence nationale de sécurité du médicament et des produits de santé (ANSM), Saint Denis, France
- Faculté de Santé, Université Paris-Est Créteil, Créteil, EA 7379, France
| | - Michel Cucherat
- Service Hospitalo-Universitaire de Pharmacotoxicologie de Lyon, Hospices Civils de Lyon, Bât. A-162, avenue Lacassagne, Lyon Cedex 03, 69424, France
| | - Judith Cottin
- Service Hospitalo-Universitaire de Pharmacotoxicologie de Lyon, Hospices Civils de Lyon, Bât. A-162, avenue Lacassagne, Lyon Cedex 03, 69424, France.
| |
Collapse
|
33
|
Taheri F, Joushi S, Esmaeilpour K, Ebrahimi MN, Taherizadeh Z, Taheri P, Sheibani V. Transmission of behavioral and cognitive impairments across generations in rats subjected to prenatal valproic acid exposure. Birth Defects Res 2024; 116:e2309. [PMID: 38343145 DOI: 10.1002/bdr2.2309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/10/2024] [Accepted: 01/18/2024] [Indexed: 02/15/2024]
Abstract
BACKGROUND Autism spectrum disorder (ASD) represents an inheritable neurodevelopmental condition characterized by social communication deficits and repetitive behaviors. Numerous studies have underscored the significant roles played by genetic and environmental factors in the etiology of ASD, and these factors are known to perpetuate behavioral impairments across generations. OBJECTIVES The primary objective of this study was to assess the behavioral and cognitive attributes in the second filial (F2) generation of male and female rats, with a particular focus on those whose parents had been exposed to valproic acid (VPA) during embryonic development. METHODS In this study, a cohort of 32 male and 32 female rats from the second filial (F2) generation, referred to as Mother.ASD, Father.ASD, or Both.ASD, was examined. These designations indicate whether the mother, father, or both parents had experienced embryonic exposure to valproic acid (600 mg/kg, i.p.). During adolescence, the F2 pups underwent behavioral and cognitive assessments, including open field testing, marble burying, social interaction evaluations, and Morris water maze tasks. RESULTS Our data revealed that while both the Mother.ASD and Father.ASD groups, regardless of sex, exhibited elevated anxiety-like behavior in the open field test. Only the Mother.ASD group displayed repetitive behaviors and deficits in social memory. Additionally, spatial memory impairments were observed in both sexes. These findings highlight the transmission of autistic-like behaviors in the offspring of Mother.ASD rats from both sexes. Nevertheless, future research endeavors should be more targeted in identifying the specific genes responsible for this transmission. CONCLUSION In summary, our findings underscore the transmission of autistic-like behaviors, including anxiety-like behavior, repetitive actions, impairments in social interactions, and deficits in memory, to the offspring of the Mother.ASD group, irrespective of their sex.
Collapse
Affiliation(s)
- Farahnaz Taheri
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Sara Joushi
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Khadijeh Esmaeilpour
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
- Public health school, University of Waterloo, Waterloo, Ontario, Canada
| | - Mohammad Navid Ebrahimi
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Zahra Taherizadeh
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Parichehr Taheri
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Vahid Sheibani
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
34
|
Jairaj C, Seneviratne G, Bergink V, Sommer IE, Dazzan P. Postpartum Psychosis: A Proposed Treatment Algorithm. FOCUS (AMERICAN PSYCHIATRIC PUBLISHING) 2024; 22:131-142. [PMID: 38694161 PMCID: PMC11058922 DOI: 10.1176/appi.focus.23021033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2024]
Abstract
Background Postpartum psychosis (PPP) is a psychiatric emergency that generally warrants acute inpatient care. PPP is marked by the sudden onset of affective and psychotic symptoms with a rapid deterioration in mental state. Evidence suggests that PPP is a discrete disorder on the bipolar disorder spectrum with a distinct treatment profile and prognosis. Methods We conducted a PubMed database search for various terms involving PPP and its treatment and included peer-reviewed articles published in English. Objective To provide a treatment algorithm for the management of PPP based on available evidence. Results Pharmacological therapy is the mainstay of PPP management in the acute phase. Evidence points to a combination of antipsychotics and lithium in the acute treatment of PPP. Electroconvulsive therapy can offer a rapid treatment response where required. Lithium appears to have the best evidence for relapse prevention and prophylaxis in PPP. Psychoeducation is essential and psychosocial interventions used in bipolar disorder may be effective in PPP. Conclusion Early detection and prompt treatment with antipsychotics and lithium, followed by maintenance treatment with lithium, is associated with a favourable prognosis in PPP.Reprinted from J Psychopharmacol 2023; 37:960-970, with permission from Sage Journals. Copyright © 2023.
Collapse
Affiliation(s)
- Chaitra Jairaj
- South London and Maudsley NHS Foundation Trust, London, UK (Jairaj, Seneviratne); Trinity College Dublin, Dublin, Ireland (Jairaj); National Maternity Hospital, Dublin, Ireland (Jairaj); Royal College of Psychiatrists, London, UK (Seneviratne); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA (Bergink); Department of Obstetrics, Gynecology, and Reproductive Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA (Bergink); Department of Psychiatry, Erasmus Medical Centre Rotterdam, Rotterdam, The Netherlands (Bergink); Department of Psychiatry, Rijksuniversiteit Groningen (RUG), University Medical Centre Groningen (UMCG), Groningen, The Netherlands (Sommer); Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK (Dazzan); National Institute for Health Research (NIHR) Mental Health Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London, London, UK (Dazzan)
| | - Gertrude Seneviratne
- South London and Maudsley NHS Foundation Trust, London, UK (Jairaj, Seneviratne); Trinity College Dublin, Dublin, Ireland (Jairaj); National Maternity Hospital, Dublin, Ireland (Jairaj); Royal College of Psychiatrists, London, UK (Seneviratne); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA (Bergink); Department of Obstetrics, Gynecology, and Reproductive Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA (Bergink); Department of Psychiatry, Erasmus Medical Centre Rotterdam, Rotterdam, The Netherlands (Bergink); Department of Psychiatry, Rijksuniversiteit Groningen (RUG), University Medical Centre Groningen (UMCG), Groningen, The Netherlands (Sommer); Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK (Dazzan); National Institute for Health Research (NIHR) Mental Health Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London, London, UK (Dazzan)
| | - Veerle Bergink
- South London and Maudsley NHS Foundation Trust, London, UK (Jairaj, Seneviratne); Trinity College Dublin, Dublin, Ireland (Jairaj); National Maternity Hospital, Dublin, Ireland (Jairaj); Royal College of Psychiatrists, London, UK (Seneviratne); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA (Bergink); Department of Obstetrics, Gynecology, and Reproductive Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA (Bergink); Department of Psychiatry, Erasmus Medical Centre Rotterdam, Rotterdam, The Netherlands (Bergink); Department of Psychiatry, Rijksuniversiteit Groningen (RUG), University Medical Centre Groningen (UMCG), Groningen, The Netherlands (Sommer); Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK (Dazzan); National Institute for Health Research (NIHR) Mental Health Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London, London, UK (Dazzan)
| | - Iris E Sommer
- South London and Maudsley NHS Foundation Trust, London, UK (Jairaj, Seneviratne); Trinity College Dublin, Dublin, Ireland (Jairaj); National Maternity Hospital, Dublin, Ireland (Jairaj); Royal College of Psychiatrists, London, UK (Seneviratne); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA (Bergink); Department of Obstetrics, Gynecology, and Reproductive Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA (Bergink); Department of Psychiatry, Erasmus Medical Centre Rotterdam, Rotterdam, The Netherlands (Bergink); Department of Psychiatry, Rijksuniversiteit Groningen (RUG), University Medical Centre Groningen (UMCG), Groningen, The Netherlands (Sommer); Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK (Dazzan); National Institute for Health Research (NIHR) Mental Health Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London, London, UK (Dazzan)
| | - Paola Dazzan
- South London and Maudsley NHS Foundation Trust, London, UK (Jairaj, Seneviratne); Trinity College Dublin, Dublin, Ireland (Jairaj); National Maternity Hospital, Dublin, Ireland (Jairaj); Royal College of Psychiatrists, London, UK (Seneviratne); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA (Bergink); Department of Obstetrics, Gynecology, and Reproductive Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA (Bergink); Department of Psychiatry, Erasmus Medical Centre Rotterdam, Rotterdam, The Netherlands (Bergink); Department of Psychiatry, Rijksuniversiteit Groningen (RUG), University Medical Centre Groningen (UMCG), Groningen, The Netherlands (Sommer); Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK (Dazzan); National Institute for Health Research (NIHR) Mental Health Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London, London, UK (Dazzan)
| |
Collapse
|
35
|
Zarate-Lopez D, Torres-Chávez AL, Gálvez-Contreras AY, Gonzalez-Perez O. Three Decades of Valproate: A Current Model for Studying Autism Spectrum Disorder. Curr Neuropharmacol 2024; 22:260-289. [PMID: 37873949 PMCID: PMC10788883 DOI: 10.2174/1570159x22666231003121513] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 08/30/2023] [Accepted: 08/30/2023] [Indexed: 10/25/2023] Open
Abstract
Autism Spectrum Disorder (ASD) is a neurodevelopmental disorder with increased prevalence and incidence in recent decades. Its etiology remains largely unclear, but it seems to involve a strong genetic component and environmental factors that, in turn, induce epigenetic changes during embryonic and postnatal brain development. In recent decades, clinical studies have shown that inutero exposure to valproic acid (VPA), a commonly prescribed antiepileptic drug, is an environmental factor associated with an increased risk of ASD. Subsequently, prenatal VPA exposure in rodents has been established as a reliable translational model to study the pathophysiology of ASD, which has helped demonstrate neurobiological changes in rodents, non-human primates, and brain organoids from human pluripotent stem cells. This evidence supports the notion that prenatal VPA exposure is a valid and current model to replicate an idiopathic ASD-like disorder in experimental animals. This review summarizes and describes the current features reported with this animal model of autism and the main neurobiological findings and correlates that help elucidate the pathophysiology of ASD. Finally, we discuss the general framework of the VPA model in comparison to other environmental and genetic ASD models.
Collapse
Affiliation(s)
- David Zarate-Lopez
- Laboratory of Neuroscience, School of Psychology, University of Colima, Colima 28040, México
- Physiological Science Ph.D. Program, School of Medicine, University of Colima, Colima 28040, Mexico
| | - Ana Laura Torres-Chávez
- Laboratory of Neuroscience, School of Psychology, University of Colima, Colima 28040, México
- Physiological Science Ph.D. Program, School of Medicine, University of Colima, Colima 28040, Mexico
| | - Alma Yadira Gálvez-Contreras
- Department of Neuroscience, Centro Universitario de Ciencias de la Salud, University of Guadalajara, Guadalajara 44340, México
| | - Oscar Gonzalez-Perez
- Laboratory of Neuroscience, School of Psychology, University of Colima, Colima 28040, México
| |
Collapse
|
36
|
Yenkoyan K, Mkhitaryan M, Bjørklund G. Environmental Risk Factors in Autism Spectrum Disorder: A Narrative Review. Curr Med Chem 2024; 31:2345-2360. [PMID: 38204225 DOI: 10.2174/0109298673252471231121045529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 10/14/2023] [Accepted: 10/19/2023] [Indexed: 01/12/2024]
Abstract
Existing evidence indicates that environmental factors might contribute up to 50% of the variance in autism spectrum disorder (ASD) risk. This structured narrative review offers a comprehensive synthesis of current knowledge on environmental risk factors in ASD, including evaluation of conflicting evidence, exploration of underlying mechanisms, and suggestions for future research directions. Analysis of diverse epidemiological investigations indicates that certain environmental factors, including advanced parental age, preterm birth, delivery complications, and exposure to toxic metals, drugs, air pollutants, and endocrine-disrupting chemicals, are linked to an increased ASD risk through various mechanisms such as oxidative stress, inflammation, hypoxia, and its consequences, changes in neurotransmitters, disruption of signaling pathways and some others. On the other hand, pregnancy-related factors such as maternal diabetes, maternal obesity, and caesarian section show a weaker association with ASD risk. At the same time, other environmental factors, such as vaccination, maternal smoking, or alcohol consumption, are not linked to the risk of ASD. Regarding nutritional elements data are inconclusive. These findings highlight the significance of environmental factors in ASD etiology and emphasize that more focused research is needed to target the risk factors of ASD. Environmental interventions targeting modifiable risk factors might offer promising avenues for ASD prevention and treatment.
Collapse
Affiliation(s)
| | - Meri Mkhitaryan
- Neuroscience Laboratory, Cobrain Center, YSMU, Yerevan, 0025, Armenia
| | - Geir Bjørklund
- Department of Research, Council for Nutritional and Environmental Medicine, Mo i Rana, Norway
| |
Collapse
|
37
|
Ornoy A, Echefu B, Becker M. Valproic Acid in Pregnancy Revisited: Neurobehavioral, Biochemical and Molecular Changes Affecting the Embryo and Fetus in Humans and in Animals: A Narrative Review. Int J Mol Sci 2023; 25:390. [PMID: 38203562 PMCID: PMC10779436 DOI: 10.3390/ijms25010390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 12/22/2023] [Accepted: 12/23/2023] [Indexed: 01/12/2024] Open
Abstract
Valproic acid (VPA) is a very effective anticonvulsant and mood stabilizer with relatively few side effects. Being an epigenetic modulator, it undergoes clinical trials for the treatment of advanced prostatic and breast cancer. However, in pregnancy, it seems to be the most teratogenic antiepileptic drug. Among the proven effects are congenital malformations in about 10%. The more common congenital malformations are neural tube defects, cardiac anomalies, urogenital malformations including hypospadias, skeletal malformations and orofacial clefts. These effects are dose related; daily doses below 600 mg have a limited teratogenic potential. VPA, when added to other anti-seizure medications, increases the malformations rate. It induces malformations even when taken for indications other than epilepsy, adding to the data that epilepsy is not responsible for the teratogenic effects. VPA increases the rate of neurodevelopmental problems causing reduced cognitive abilities and language impairment. It also increases the prevalence of specific neurodevelopmental syndromes like autism (ASD) and Attention Deficit Hyperactivity Disorder (ADHD). High doses of folic acid administered prior to and during pregnancy might alleviate some of the teratogenic effect of VPA and other AEDs. Several teratogenic mechanisms are proposed for VPA, but the most important mechanisms seem to be its effects on the metabolism of folate, SAMe and histones, thus affecting DNA methylation. VPA crosses the human placenta and was found at higher concentrations in fetal blood. Its concentrations in milk are low, therefore nursing is permitted. Animal studies generally recapitulate human data.
Collapse
Affiliation(s)
- Asher Ornoy
- Department of Morphological Sciences and Teratology, Adelson School of Medicine, Ariel University, Ariel 40700, Israel; (B.E.); (M.B.)
- Department of Medical Neurobiology, Hebrew University Hadassah Medical School, Jerusalem 9112102, Israel
| | - Boniface Echefu
- Department of Morphological Sciences and Teratology, Adelson School of Medicine, Ariel University, Ariel 40700, Israel; (B.E.); (M.B.)
| | - Maria Becker
- Department of Morphological Sciences and Teratology, Adelson School of Medicine, Ariel University, Ariel 40700, Israel; (B.E.); (M.B.)
| |
Collapse
|
38
|
Li Y, Ma L, Deng Y, Du Z, Guo B, Yue J, Liu X, Zhang Y. The Notch1/Hes1 signaling pathway affects autophagy by adjusting DNA methyltransferases expression in a valproic acid-induced autism spectrum disorder model. Neuropharmacology 2023; 239:109682. [PMID: 37543138 DOI: 10.1016/j.neuropharm.2023.109682] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 07/23/2023] [Accepted: 08/02/2023] [Indexed: 08/07/2023]
Abstract
As a pervasive neurodevelopmental disease, autism spectrum disorder (ASD) is caused by both hereditary and environmental elements. Research has demonstrated the functions of the Notch pathway and DNA methylation in the etiology of ASD. DNA methyltransferases DNMT3 and DNMT1 are responsible for methylation establishment and maintenance, respectively. In this study, we aimed to explore the association of DNA methyltransferases with the Notch pathway in ASD. Our results showed Notch1 and Hes1 were upregulated, while DNMT3A and DNMT3B were downregulated at the protein level in the prefrontal cortex (PFC), hippocampus (HC) and cerebellum (CB) of VPA-induced ASD rats compared with Control (Con) group. However, the protein levels of DNMT3A and DNMT3B were augmented after treatment with 3,5-difluorophenacetyl-L-alanyl-S-phenylglycine-2-butyl ester (DAPT), suggesting that abnormal Notch pathway activation may affect the expression of DNMT3A and DNMT3B. Besides, our previous findings revealed that the Notch pathway may participate in development of ASD by influencing autophagy. Therefore, we hypothesized the Notch pathway adjusts autophagy and contributes to ASD by affecting DNA methyltransferases. Our current results showed that after receiving the DNA methyltransferase inhibitor 5-Aza-2'-deoxycytidine (5-Aza-2'dc), the VPA + DAPT+5-Aza-2'dc (V + D + Aza) group exhibited reduced social interaction ability and increased stereotyped behaviors, and decreased expression of DNMT3A, DNMT3B and autophagy-related proteins, but did not show changes in Notch1 and Hes1 protein levels. Our results indicated that the Notch1/Hes1 pathway may adjust DNMT3A and DNMT3B expression and subsequently affect autophagy in the occurrence of ASD, providing new insight into the pathogenesis of ASD.
Collapse
Affiliation(s)
- Yanfang Li
- Department of Human Anatomy & Histoembryology, Xinxiang Medical University, Xinxiang, Henan, 453003, China; Xinxiang Key Laboratory of Molecular Neurology, Xinxiang, Henan, 453003, China
| | - Liping Ma
- Department of Human Anatomy & Histoembryology, Xinxiang Medical University, Xinxiang, Henan, 453003, China; Xinxiang Key Laboratory of Molecular Neurology, Xinxiang, Henan, 453003, China
| | - Yanan Deng
- Department of Human Anatomy & Histoembryology, Xinxiang Medical University, Xinxiang, Henan, 453003, China; Xinxiang Key Laboratory of Molecular Neurology, Xinxiang, Henan, 453003, China
| | - Ziwei Du
- Department of Human Anatomy & Histoembryology, Xinxiang Medical University, Xinxiang, Henan, 453003, China; Xinxiang Key Laboratory of Molecular Neurology, Xinxiang, Henan, 453003, China
| | - Bingqian Guo
- Department of Human Anatomy & Histoembryology, Xinxiang Medical University, Xinxiang, Henan, 453003, China; Xinxiang Key Laboratory of Molecular Neurology, Xinxiang, Henan, 453003, China
| | - Jianing Yue
- Department of Human Anatomy & Histoembryology, Xinxiang Medical University, Xinxiang, Henan, 453003, China; Xinxiang Key Laboratory of Molecular Neurology, Xinxiang, Henan, 453003, China
| | - Xianxian Liu
- Department of Human Anatomy & Histoembryology, Xinxiang Medical University, Xinxiang, Henan, 453003, China; Xinxiang Key Laboratory of Molecular Neurology, Xinxiang, Henan, 453003, China
| | - Yinghua Zhang
- Department of Human Anatomy & Histoembryology, Xinxiang Medical University, Xinxiang, Henan, 453003, China; Xinxiang Key Laboratory of Molecular Neurology, Xinxiang, Henan, 453003, China.
| |
Collapse
|
39
|
Bluett-Duncan M, Astill D, Charbak R, Clayton-Smith J, Cole S, Cook PA, Cozens J, Keely K, Morris J, Mukherjee R, Murphy E, Turnpenny P, Williams J, Wood AG, Yates LM, Bromley RL. Neurodevelopmental outcomes in children and adults with Fetal Valproate Spectrum Disorder: A contribution from the ConcePTION project. Neurotoxicol Teratol 2023; 100:107292. [PMID: 37666366 DOI: 10.1016/j.ntt.2023.107292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 07/17/2023] [Accepted: 09/01/2023] [Indexed: 09/06/2023]
Abstract
AIM To describe the neurodevelopmental phenotype of older children and adults with a diagnosis of Fetal Valproate Spectrum Disorder (FVSD). METHODS In this cross-sectional study, 90 caregivers were recruited and completed a series of questionnaires regarding the neurodevelopmental outcomes of 146 individuals aged 7-37 years (M = 18.1), including individuals with a formal diagnosis of FVSD (n = 99), individuals exposed to Valproate but without an FVSD diagnosis (n = 24), and individuals not exposed to Valproate (N = 23). The mean dose of valproate exposure for individuals with an FVSD diagnosis was 1470 mg/day. RESULTS Individuals with a diagnosis of FVSD showed significantly higher levels of moderate (43.4%) and severe (14.4%) cognitive impairment than other groups (p = 0.003), high levels of required formal educational support (77.6%), and poorer academic competence than individuals not exposed to Valproate (p = 0.001). Overall psychosocial problems (p = 0.02), internalising problems (p = 0.05) and attention problems (p = 0.001), but not externalising problems, were elevated in individuals with a diagnosis of FVSD. Rates of neurodevelopmental disorders, particularly autistic spectrum disorders (62.9%) and sensory problems (80.6%) are particularly central to the FVSD phenotype. There was no evidence of a statistical dose-dependent effect, possibly due to the high mean dose of exposure having a uniformly negative impact across the sample. Individuals with FVSD had required a significant number of health and child development services. INTERPRETATION Children and young adults with a diagnosis of FVSD are at an increased risk of a range of altered neurodevelopmental outcomes, highlighting the need for a multidisciplinary approach to clinical management across the lifespan.
Collapse
Affiliation(s)
- M Bluett-Duncan
- Division of Neuroscience, School of Biological Sciences, The University of Manchester, UK.
| | - D Astill
- Foetal Anti-Convulsant Syndrome New Zealand, Aukland, New Zealand
| | - R Charbak
- Division of Neuroscience, School of Biological Sciences, The University of Manchester, UK
| | - J Clayton-Smith
- Division of Evolution, Systems and Genomics, University of Manchester, UK
| | - S Cole
- Pharma Consent, London, UK
| | - P A Cook
- School of Health and Society, University of Salford, UK
| | - J Cozens
- Organisation for Anti-Convulsant Syndrome, UK
| | - K Keely
- Organisation for Anti-Convulsant Syndrome, Ireland
| | - J Morris
- Foetal Anti-Convulsant Syndrome New Zealand, Aukland, New Zealand
| | - R Mukherjee
- Department of Health and Social Science, University of Salford, Allerton Building, Salford, England, UK
| | - E Murphy
- Independent Fetal Anti-Convulsant Trust, Preston, UK
| | - P Turnpenny
- Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | - J Williams
- Independent Fetal Anti-Convulsant Trust, Preston, UK
| | - A G Wood
- Clinical Sciences, Murdoch Children's Research Institute, & School of Psychology, Deakin University, Melbourne, Australia; Institute of Health and Neurodevelopment, Aston University, UK
| | - L M Yates
- Northern Genetics Service, Newcastle upon Tyne Hospitals NHS Foundation Trust, UK; KRISP, University of KwaZulu-Natal, South Africa
| | - R L Bromley
- Division of Neuroscience, School of Biological Sciences, The University of Manchester, UK; Royal Manchester Children's Hospital, Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| |
Collapse
|
40
|
Abstract
BACKGROUND Postpartum psychosis (PPP) is a psychiatric emergency that generally warrants acute inpatient care. PPP is marked by the sudden onset of affective and psychotic symptoms with a rapid deterioration in mental state. Evidence suggests that PPP is a discrete disorder on the bipolar disorder spectrum with a distinct treatment profile and prognosis. METHODS We conducted a PubMed database search for various terms involving PPP and its treatment and included peer-reviewed articles published in English. OBJECTIVE To provide a treatment algorithm for the management of PPP based on available evidence. RESULTS Pharmacological therapy is the mainstay of PPP management in the acute phase. Evidence points to a combination of antipsychotics and lithium in the acute treatment of PPP. Electroconvulsive therapy can offer a rapid treatment response where required. Lithium appears to have the best evidence for relapse prevention and prophylaxis in PPP. Psychoeducation is essential and psychosocial interventions used in bipolar disorder may be effective in PPP. CONCLUSION Early detection and prompt treatment with antipsychotics and lithium, followed by maintenance treatment with lithium, is associated with a favourable prognosis in PPP.
Collapse
Affiliation(s)
- Chaitra Jairaj
- South London and Maudsley NHS Foundation Trust, London, UK
- Trinity College Dublin, Dublin, Ireland
- National Maternity Hospital, Dublin, Ireland
| | - Gertrude Seneviratne
- South London and Maudsley NHS Foundation Trust, London, UK
- Royal College of Psychiatrists, London, UK
| | - Veerle Bergink
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Obstetrics, Gynecology, and Reproductive Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Erasmus Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Iris E Sommer
- Department of Psychiatry, Rijksuniversiteit Groningen (RUG), University Medical Centre Groningen (UMCG), Groningen, The Netherlands
| | - Paola Dazzan
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
- National Institute for Health Research (NIHR) Mental Health Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King’s College London, London, UK
| |
Collapse
|
41
|
Bromley RL, Bullen P, Campbell E, Craig J, Ingham A, Irwin B, Jackson C, Kelly T, Morrow J, Rushton S, García-Fiñana M, Hughes DM, Winterbottom J, Wood A, Yates LM, Clayton-Smith J. Neurodevelopment of babies born to mothers with epilepsy: A prospective observational cohort study. Epilepsia 2023; 64:2454-2471. [PMID: 37403560 DOI: 10.1111/epi.17709] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/27/2023] [Accepted: 06/29/2023] [Indexed: 07/06/2023]
Abstract
OBJECTIVE Despite widespread monotherapy use of lamotrigine or levetiracetam during pregnancy, prospectively collected, blinded child development data are still limited. The NaME (Neurodevelopment of Babies Born to Mothers With Epilepsy) Study prospectively recruited a new cohort of women with epilepsy and their offspring for longitudinal follow-up. METHODS Pregnant women of <21 weeks gestation (n = 401) were recruited from 21 hospitals in the UK. Data collection occurred during pregnancy (recruitment, trimester 3) and at 12 and 24 months of age. The primary outcome was blinded assessment of infant cognitive, language, and motor development on the Bayley Scales of Infant and Toddler Development (3rd edition) at 24 months of age with supplementary parent reporting on the Vinelands Adaptive Behavior Scales (2nd edition). RESULTS There were 394 live births, with 277 children (70%) completing the Bayley assessment at 24 months. There was no evidence of an association of prenatal exposure to monotherapy lamotrigine (-.74, SE = 2.9, 95% confidence interval [CI] = -6.5 to 5.0, p = .80) or levetiracetam (-1.57, SE = 3.1, 95% CI = -4.6 to 7.7, p = .62) with poorer infant cognition, following adjustment for other maternal and child factors in comparison to nonexposed children. Similar results were observed for language and motor scores. There was no evidence of an association between increasing doses of either lamotrigine or levetiracetam. Nor was there evidence that higher dose folic acid supplementation (≥5 mg/day) or convulsive seizure exposure was associated with child development scores. Continued infant exposure to antiseizure medications through breast milk was not associated with poorer outcomes, but the number of women breastfeeding beyond 3 months was low. SIGNIFICANCE These data are reassuring for infant development following in utero exposure to monotherapy lamotrigine or levetiracetam, but child development is dynamic, and future follow-up is required to rule out later emerging effects.
Collapse
Affiliation(s)
- Rebecca L Bromley
- Division of Neuroscience, University of Manchester, Manchester, UK
- Royal Manchester Children's Hospital, Manchester University Hospitals NHS Foundation Trust and Manchester Academic Health Sciences Centre, Manchester, UK
| | - Philip Bullen
- Obstetric and Fetal Medicine, St. Mary's Hospital, Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Ellen Campbell
- Department of Neurology, Belfast Health and Social Care Trust, Belfast, UK
| | - John Craig
- Department of Neurology, Belfast Health and Social Care Trust, Belfast, UK
| | - Amy Ingham
- Manchester Centre for Genomic Medicine, St. Mary's Hospital, University of Manchester, Manchester, UK
| | - Beth Irwin
- Department of Neurology, Belfast Health and Social Care Trust, Belfast, UK
| | - Cerain Jackson
- Department of Neuropsychology, Walton Centre for Neurology and Neurosurgery NHS Foundation Trust, Liverpool, UK
| | - Teresa Kelly
- Obstetric and Fetal Medicine, St. Mary's Hospital, Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - James Morrow
- Department of Neurology, Belfast Health and Social Care Trust, Belfast, UK
| | - Sarah Rushton
- Manchester Centre for Genomic Medicine, St. Mary's Hospital, University of Manchester, Manchester, UK
| | - Marta García-Fiñana
- Department of Health Data Science, Institute of Population Health, University of Liverpool, Liverpool, UK
| | - David M Hughes
- Department of Health Data Science, Institute of Population Health, University of Liverpool, Liverpool, UK
| | - Janine Winterbottom
- Department of Neurology, Walton Centre for Neurology and Neurosurgery NHS Foundation Trust, Liverpool, UK
| | - Amanda Wood
- School of Psychology, Deakin University, Burwood, Victoria, Australia
- Aston Institute for Health and Neurodevelopment, Aston University, Birmingham, UK
| | - Laura M Yates
- Department for Clinical Genetics, Nothern Genetics Service, Newcastle, UK
| | - Jill Clayton-Smith
- Manchester Centre for Genomic Medicine, St. Mary's Hospital, University of Manchester, Manchester, UK
- Division of Evolution and Genomic Sciences, School of Biological Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
42
|
Malhotra AS, Kulesza R. Abnormal auditory brainstem responses in an animal model of autism spectrum disorder. Hear Res 2023; 436:108816. [PMID: 37285705 DOI: 10.1016/j.heares.2023.108816] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 04/15/2023] [Accepted: 05/24/2023] [Indexed: 06/09/2023]
Abstract
Auditory dysfunction is a common feature of autism spectrum disorder (ASD) and ranges from deafness to hypersensitivity. The auditory brainstem response (ABR) permits study of the amplitude and latency of synchronized electrical activity along the ascending auditory pathway in response to clicks and pure tone stimuli. Indeed, numerous studies have shown that subjects with ASD have ABR abnormalities. In utero exposure to the antiepileptic drug valproic acid (VPA) is associated with human cases of ASD and is used as an animal model of ASD. Previous studies have shown that VPA-exposed animals have significantly fewer neurons in the auditory brainstem and thalamus, reduced ascending projections to the auditory midbrain and thalamus and increased neuronal activation in response to pure tone stimuli. Accordingly, we hypothesized that VPA-exposed animals would have abnormal ABRs throughout their lifespans. We approached this hypothesis in two cohorts. First, we examined ABRs from both ears on postnatal day 22 (P22). Then, we examined monaural ABRs in animals at P28, 60, 120, 180, 240, 300 and 360. Our results suggest that at P22, VPA-exposed animals have elevated thresholds and increased peak latencies. However, by P60 these differences largely normalize with differences appearing only near hearing threshold. Additionally, our analysis revealed that maturation of ABR waves occurred at different trajectories in control and VPA-exposed animals. These results, together with our previous work, suggest that VPA exposure not only impacts total neuron number and connectivity, but also auditory evoked responses. Finally, our longitudinal analysis suggests that delayed maturation of auditory brainstem circuits may impact ABRs throughout the lifespan of the animal.
Collapse
Affiliation(s)
- Arjun S Malhotra
- Department of Anatomy Lake Erie College of Osteopathic Medicine, Erie, Pennsylvania, USA; Millcreek Community Hospital LECOM Health, Department of Orthopedic Surgery, Erie, Pennsylvania, USA
| | - Randy Kulesza
- Department of Anatomy Lake Erie College of Osteopathic Medicine, Erie, Pennsylvania, USA.
| |
Collapse
|
43
|
Leung CS, Rosenzweig SJ, Yoon B, Marinelli NA, Hollingsworth EW, Maguire AM, Cowen MH, Schmidt M, Imitola J, Gamsiz Uzun ED, Lizarraga SB. Dysregulation of the chromatin environment leads to differential alternative splicing as a mechanism of disease in a human model of autism spectrum disorder. Hum Mol Genet 2023; 32:1634-1646. [PMID: 36621967 PMCID: PMC10162432 DOI: 10.1093/hmg/ddad002] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/24/2022] [Accepted: 01/03/2023] [Indexed: 01/10/2023] Open
Abstract
Autism spectrum disorder (ASD) affects 1 in 44 children. Chromatin regulatory proteins are overrepresented among genes that contain high risk variants in ASD. Disruption of the chromatin environment leads to widespread dysregulation of gene expression, which is traditionally thought of as a mechanism of disease pathogenesis associated with ASD. Alternatively, alterations in chromatin dynamics could also lead to dysregulation of alternative splicing, which is understudied as a mechanism of ASD pathogenesis. The anticonvulsant valproic acid (VPA) is a well-known environmental risk factor for ASD that acts as a class I histone deacetylase inhibitor. However, the precise molecular mechanisms underlying defects in human neuronal development associated with exposure to VPA are understudied. To dissect how VPA exposure and subsequent chromatin hyperacetylation influence molecular signatures involved in ASD pathogenesis, we conducted RNA sequencing (RNA-seq) in human cortical neurons that were treated with VPA. We observed that differentially expressed genes (DEGs) were enriched for mRNA splicing, mRNA processing, histone modification and metabolism related gene sets. Furthermore, we observed widespread increases in the number and the type of alternative splicing events. Analysis of differential transcript usage (DTU) showed that exposure to VPA induces extensive alterations in transcript isoform usage across neurodevelopmentally important genes. Finally, we find that DEGs and genes that display DTU overlap with known ASD-risk genes. Altogether, these findings suggest that, in addition to differential gene expression, changes in alternative splicing correlated with alterations in the chromatin environment could act as an additional mechanism of disease in ASD.
Collapse
Affiliation(s)
- Calvin S Leung
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA
- Center for Translational Neuroscience, Carney Institute for Brain Science and Brown Institute for Translational Science (BITS), Brown University, Providence, RI 02912, USA
| | - Shoshana J Rosenzweig
- Center for Computational Molecular Biology, Brown University, Providence, RI 02906, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School of Brown University, Providence, RI 02912, USA
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Academic Medical Center, Providence, RI 02903, USA
| | - Brian Yoon
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Nicholas A Marinelli
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Ethan W Hollingsworth
- UCONN Health Comprehensive Multiple Sclerosis Center, Department of Neurology, University of Connecticut School of Medicine, Farmington, CT 06030, USA
- Division of Multiple Sclerosis and Translational Neuroimmunology, Department of Neurology, University of Connecticut School of Medicine, Farmington, CT 06030, USA
| | - Abbie M Maguire
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA
- Center for Translational Neuroscience, Carney Institute for Brain Science and Brown Institute for Translational Science (BITS), Brown University, Providence, RI 02912, USA
| | - Mara H Cowen
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Michael Schmidt
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA
- Center for Translational Neuroscience, Carney Institute for Brain Science and Brown Institute for Translational Science (BITS), Brown University, Providence, RI 02912, USA
| | - Jaime Imitola
- UCONN Health Comprehensive Multiple Sclerosis Center, Department of Neurology, University of Connecticut School of Medicine, Farmington, CT 06030, USA
- Division of Multiple Sclerosis and Translational Neuroimmunology, Department of Neurology, University of Connecticut School of Medicine, Farmington, CT 06030, USA
| | - Ece D Gamsiz Uzun
- Center for Computational Molecular Biology, Brown University, Providence, RI 02906, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School of Brown University, Providence, RI 02912, USA
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Academic Medical Center, Providence, RI 02903, USA
| | - Sofia B Lizarraga
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA
- Center for Translational Neuroscience, Carney Institute for Brain Science and Brown Institute for Translational Science (BITS), Brown University, Providence, RI 02912, USA
| |
Collapse
|
44
|
Mitsuhashi T, Hattori S, Fujimura K, Shibata S, Miyakawa T, Takahashi T. In utero Exposure to Valproic Acid throughout Pregnancy Causes Phenotypes of Autism in Offspring Mice. Dev Neurosci 2023; 45:223-233. [PMID: 37044070 DOI: 10.1159/000530452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 03/24/2023] [Indexed: 04/14/2023] Open
Abstract
Valproic acid (VPA) is an antiepileptic drug that inhibits the epileptic activity of neurons mainly by inhibiting sodium channels and GABA transaminase. VPA is also known to inhibit histone deacetylases, which epigenetically modify the cell proliferation/differentiation characteristics of stem/progenitor cells within developing tissues. Recent clinical studies in humans have indicated that VPA exposure in utero increases the risk of autistic features and intellectual disabilities in offspring; we have previously reported that low-dose VPA exposure in utero throughout pregnancy increases the production of projection neurons from neuronal stem/progenitor cells that are distributed in the superficial neocortical layers of the fetal brain. In the present study, we found that in utero VPA-exposed mice exhibited abnormal social interaction, changes in cognitive function, hypersensitivity to pain/heat, and impaired locomotor activity, all of which are characteristic symptoms of autism spectrum disorder in humans. Taken together, our findings indicate that VPA exposure in utero throughout pregnancy alters higher brain function and predisposes individuals to phenotypes that resemble autism and intellectual disability. Furthermore, these symptoms are likely to be due to neocortical dysgenesis that was caused by an increased number of projection neurons in specific layers of the neocortex.
Collapse
Affiliation(s)
| | - Satoko Hattori
- Division of Systems Medical Science, Center for Medical Science, Fujita Health University, Aichi, Japan
| | - Kimino Fujimura
- Departments of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Shinsuke Shibata
- Departments of Physiology, Keio University School of Medicine, Tokyo, Japan
- Division of Microscopic Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Tsuyoshi Miyakawa
- Division of Systems Medical Science, Center for Medical Science, Fujita Health University, Aichi, Japan
| | - Takao Takahashi
- Departments of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
45
|
Abstract
Epilepsy affects up to 15 million of people assigned female at birth of childbearing age globally. Up to 65% of these people with epilepsy and gestational capacity have an unplanned pregnancy. Seizure control during pregnancy is important for both the childbearer's and fetus' safety. There are multiple antiseizure medications (ASMs) that can be used to control epilepsy; however, each medication has its own teratogenic risk profile, which must be considered. The majority of these ASMs will require frequent plasma concentration monitoring during pregnancy with corresponding dosage adjustments. Dosages can be reduced back to prepregnancy levels within 3 weeks postpartum. Breastfeeding on ASMs is recommended.
Collapse
|
46
|
Knight R, Craig J, Irwin B, Wittkowski A, Bromley RL. Adaptive behaviour in children exposed to topiramate in the womb: An observational cohort study. Seizure 2023; 105:56-64. [PMID: 36731257 DOI: 10.1016/j.seizure.2023.01.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 01/10/2023] [Accepted: 01/12/2023] [Indexed: 01/14/2023] Open
Abstract
OBJECTIVE Many women with epilepsy need to continue anti-seizure medications (ASMs) throughout pregnancy. The current study investigated adaptive behaviour outcomes in children exposed to topiramate in the womb. METHOD An observational, cross-sectional study was designed, recruiting mother-child-pairs from the UK Epilepsy and Pregnancy Register (UKEPR). Health, developmental histories and Vineland Adaptive Behaviour Scale-Third Edition (VABS-III) assessments were administered via telephone by a blinded researcher, supplemented with prospectively collected pregnancy and medication information. Topiramate monotherapy exposed children were compared to VABS-III normative data as recruitment was disrupted by the COVID-19 pandemic. RESULTS Thirty-four women with epilepsy from 135 (25%) initially agreed to participate in the study, of whom 26 women completed telephone interviews about their children (n = 28). Children ranged from 2.5 to 17 years of age at the time of assessment. Six topiramate-exposed children were born small for gestational age, and there were significant associations between birthweight, dose and VABS-III scores. Significantly lower scores were observed in topiramate-exposed children (n = 21) with a significant dose-response relationship established after adjustment for parental educational level. Daily mean dosage was 280.21 mg, with high dosages of topiramate associated with a 12-point reduction in VABS-III scores. Additionally, four topiramate-exposed children (19.05%) had diagnoses of Autism Spectrum Disorder, which was significantly higher than UK prevalence rates (1.1%). CONCLUSIONS The findings of poorer adaptive behaviour, higher incidence of ASD and associations with birth weight are of concern and require further validation and replication using larger prospectively-recruited samples and comparator cohorts. Implications for research and clinical practice are discussed.
Collapse
Affiliation(s)
- R Knight
- Division of Psychology and Mental Health, The University of Manchester, UK; Greater Manchester Mental Health NHS Foundation Trust, UK
| | - J Craig
- Department of Neurology, Belfast Health and Social Care Trust, Belfast, UK
| | - B Irwin
- Department of Neurology, Belfast Health and Social Care Trust, Belfast, UK
| | - A Wittkowski
- Division of Psychology and Mental Health, The University of Manchester, UK; Greater Manchester Mental Health NHS Foundation Trust, UK
| | - R L Bromley
- Division of Neuroscience and Experimental Psychology Science, The University of Manchester, UK; Royal Manchester Children's Hospital, Manchester Academic Health Sciences, Manchester, UK.
| |
Collapse
|
47
|
Cochrane Epilepsy Group, Maguire MJ, Fairclough S, Nevitt SJ. Antiepileptic drugs for treating seizures in people with brain tumours. THE COCHRANE DATABASE OF SYSTEMATIC REVIEWS 2023. [PMCID: PMC9890922 DOI: 10.1002/14651858.cd015467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
This is a protocol for a Cochrane Review (intervention). The objectives are as follows: To compare the efficacy and tolerability of antiepileptic drugs (AEDs) taken as monotherapy or add‐on therapy for seizures in people with brain tumours.
Collapse
Affiliation(s)
| | | | | | - Sarah J Nevitt
- Department of Health Data ScienceUniversity of LiverpoolLiverpoolUK
| |
Collapse
|
48
|
Dixon SC, Calder BJ, Lilya SM, Davies BM, Martin A, Peterson M, Hansen JM, Suli A. Valproic acid affects neurogenesis during early optic tectum development in zebrafish. Biol Open 2023; 12:286129. [PMID: 36537579 PMCID: PMC9916031 DOI: 10.1242/bio.059567] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 12/09/2022] [Indexed: 02/01/2023] Open
Abstract
The mammalian superior colliculus and its non-mammalian homolog, the optic tectum (OT), are midbrain structures that integrate multimodal sensory inputs and guide non-voluntary movements in response to prevalent stimuli. Recent studies have implicated this structure as a possible site affected in autism spectrum disorder (ASD). Interestingly, fetal exposure to valproic acid (VPA) has also been associated with an increased risk of ASD in humans and animal models. Therefore, we took the approach of determining the effects of VPA treatment on zebrafish OT development as a first step in identifying the mechanisms that allow its formation. We describe normal OT development during the first 5 days of development and show that in VPA-treated embryos, neuronal specification and neuropil formation was delayed. VPA treatment was most detrimental during the first 3 days of development and did not appear to be linked to oxidative stress. In conclusion, our work provides a foundation for research into mechanisms driving OT development, as well as the relationship between the OT, VPA, and ASD. This article has an associated First Person interview with one of the co-first authors of the paper.
Collapse
Affiliation(s)
- Sierra C. Dixon
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA
| | - Bailey J. Calder
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA
| | - Shane M. Lilya
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA
| | - Brandon M. Davies
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA
| | - Annalie Martin
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA
| | - Maggie Peterson
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA
| | - Jason M. Hansen
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA
| | - Arminda Suli
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA,Author for correspondence ()
| |
Collapse
|
49
|
Boktor JC, Adame MD, Rose DR, Schumann CM, Murray KD, Bauman MD, Careaga M, Mazmanian SK, Ashwood P, Needham BD. Global metabolic profiles in a non-human primate model of maternal immune activation: implications for neurodevelopmental disorders. Mol Psychiatry 2022; 27:4959-4973. [PMID: 36028571 PMCID: PMC9772216 DOI: 10.1038/s41380-022-01752-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 08/02/2022] [Accepted: 08/12/2022] [Indexed: 01/14/2023]
Abstract
Epidemiological evidence implicates severe maternal infections as risk factors for neurodevelopmental disorders, such as ASD and schizophrenia. Accordingly, animal models mimicking infection during pregnancy, including the maternal immune activation (MIA) model, result in offspring with neurobiological, behavioral, and metabolic phenotypes relevant to human neurodevelopmental disorders. Most of these studies have been performed in rodents. We sought to better understand the molecular signatures characterizing the MIA model in an organism more closely related to humans, rhesus monkeys (Macaca mulatta), by evaluating changes in global metabolic profiles in MIA-exposed offspring. Herein, we present the global metabolome in six peripheral tissues (plasma, cerebrospinal fluid, three regions of intestinal mucosa scrapings, and feces) from 13 MIA and 10 control offspring that were confirmed to display atypical neurodevelopment, elevated immune profiles, and neuropathology. Differences in lipid, amino acid, and nucleotide metabolism discriminated these MIA and control samples, with correlations of specific metabolites to behavior scores as well as to cytokine levels in plasma, intestinal, and brain tissues. We also observed modest changes in fecal and intestinal microbial profiles, and identify differential metabolomic profiles within males and females. These findings support a connection between maternal immune activation and the metabolism, microbiota, and behavioral traits of offspring, and may further the translational applications of the MIA model and the advancement of biomarkers for neurodevelopmental disorders such as ASD or schizophrenia.
Collapse
Affiliation(s)
- Joseph C Boktor
- Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Mark D Adame
- Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Destanie R Rose
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, 95616, USA
- The M.I.N.D. Institute, University of California, Davis, Sacramento, CA, 95817, USA
| | - Cynthia M Schumann
- The M.I.N.D. Institute, University of California, Davis, Sacramento, CA, 95817, USA
| | - Karl D Murray
- The M.I.N.D. Institute, University of California, Davis, Sacramento, CA, 95817, USA
| | - Melissa D Bauman
- The M.I.N.D. Institute, University of California, Davis, Sacramento, CA, 95817, USA
| | - Milo Careaga
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, 95616, USA
- The M.I.N.D. Institute, University of California, Davis, Sacramento, CA, 95817, USA
| | - Sarkis K Mazmanian
- Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, 95616, USA.
- The M.I.N.D. Institute, University of California, Davis, Sacramento, CA, 95817, USA.
| | - Brittany D Needham
- Department of Anatomy, Cell Biology & Physiology, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
50
|
Cipriani C, Giudice M, Petrone V, Fanelli M, Minutolo A, Miele MT, Toschi N, Maracchioni C, Siracusano M, Benvenuto A, Coniglio A, Curatolo P, Mazzone L, Sandro G, Garaci E, Sinibaldi-Vallebona P, Matteucci C, Balestrieri E. Modulation of human endogenous retroviruses and cytokines expression in peripheral blood mononuclear cells from autistic children and their parents. Retrovirology 2022; 19:26. [PMID: 36451209 PMCID: PMC9709758 DOI: 10.1186/s12977-022-00603-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 05/09/2022] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND Putative pathogenic effects mediated by human endogenous retroviruses (HERVs) in neurological and psychiatric disorders in humans have been extensively described. HERVs may alter the development of the brain by means of several mechanisms, including modulation of gene expression, alteration of DNA stability, and activation of immune system. We recently demonstrated that autistic children and their mothers share high expression levels of some HERVs and cytokines in peripheral blood mononuclear cells (PBMCs) ex vivo, suggesting a close mother-child association in Autism Spectrum Disorder (ASD). RESULTS In the present study, PBMCs from autistic children and their parents were exposed to stimulating factors (Interleukin-2/Phytohaemagglutinin) or drugs, as Valproic acid and Efavirenz. The results show that HERVs and cytokines expression can be modulated in vitro by different stimuli in PBMCs from autistic children and their mothers, while no significant changes were found in PBMCs ASD fathers or in controls individuals. In particular, in vitro exposure to interleukin-2/Phytohaemagglutinin or valproic acid induces the expression of several HERVs and cytokines while Efavirenz inhibits them. CONCLUSION Herein we show that autistic children and their mothers share an intrinsic responsiveness to in vitro microenvironmental changes in expressing HERVs and pro-inflammatory cytokines. Remarkably, the antiretroviral drug Efavirenz restores the expression of specific HERV families to values similar to those of the controls, also reducing the expression of proinflammatory cytokines but keeping the regulatory ones high. Our findings open new perspectives to study the role of HERVs in the biological mechanisms underlying Autism.
Collapse
Affiliation(s)
- Chiara Cipriani
- grid.6530.00000 0001 2300 0941Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Martina Giudice
- grid.6530.00000 0001 2300 0941Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Vita Petrone
- grid.6530.00000 0001 2300 0941Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Marialaura Fanelli
- grid.6530.00000 0001 2300 0941Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Antonella Minutolo
- grid.6530.00000 0001 2300 0941Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Martino T. Miele
- grid.6530.00000 0001 2300 0941Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Nicola Toschi
- grid.6530.00000 0001 2300 0941Department of Biomedicine and Prevention, Tor Vergata University of Rome, 00133 Rome, Italy ,grid.38142.3c000000041936754XMartinos Center for Biomedical Imaging and Harvard Medical School, Boston, USA
| | - Christian Maracchioni
- grid.6530.00000 0001 2300 0941Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Martina Siracusano
- grid.6530.00000 0001 2300 0941Department of Biomedicine and Prevention, Tor Vergata University of Rome, 00133 Rome, Italy
| | - Arianna Benvenuto
- grid.413009.fChild Neurology and Psychiatry Unit, System Medicine Department, Tor Vergata University Hospital of Rome, 00133 Rome, Italy
| | - Antonella Coniglio
- grid.413009.fChild Neurology and Psychiatry Unit, System Medicine Department, Tor Vergata University Hospital of Rome, 00133 Rome, Italy
| | - Paolo Curatolo
- grid.413009.fChild Neurology and Psychiatry Unit, System Medicine Department, Tor Vergata University Hospital of Rome, 00133 Rome, Italy
| | - Luigi Mazzone
- grid.413009.fChild Neurology and Psychiatry Unit, System Medicine Department, Tor Vergata University Hospital of Rome, 00133 Rome, Italy
| | - Grelli Sandro
- grid.6530.00000 0001 2300 0941Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy ,Virology Unit, Policlinic of Tor Vergata, 00133 Rome, Italy
| | - Enrico Garaci
- University San Raffaele, Rome, Italy ,grid.18887.3e0000000417581884IRCCS San Raffaele Pisana, 00133 Rome, Italy
| | - Paola Sinibaldi-Vallebona
- grid.6530.00000 0001 2300 0941Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy ,grid.5326.20000 0001 1940 4177Institute of Translational Pharmacology, National Research Council, 00133 Rome, Italy
| | - Claudia Matteucci
- grid.6530.00000 0001 2300 0941Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Emanuela Balestrieri
- grid.6530.00000 0001 2300 0941Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| |
Collapse
|