1
|
Wallace KJ, Dupeyron S, Li M, Kelly AM. Early life social complexity shapes adult neural processing in the communal spiny mouse Acomys cahirinus. Psychopharmacology (Berl) 2025; 242:1025-1040. [PMID: 38055059 DOI: 10.1007/s00213-023-06513-5] [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: 08/24/2023] [Accepted: 11/22/2023] [Indexed: 12/07/2023]
Abstract
RATIONALE Early life social rearing has profound consequences on offspring behavior and resilience. Yet, most studies examining early life development in rodents use species whose young are born immobile and do not produce complex social behavior until later in development. Furthermore, models of rearing under increased social complexity, rather than deprivation, are needed to provide alternative insight into the development of social neural circuitry. OBJECTIVES To understand precocial offspring social development, we manipulated early life social complexity in the communal spiny mouse Acomys cahirinus and assessed long-term consequences on offspring social behavior, exploration, and neural responses to novel social stimuli. METHODS Spiny mouse pups were raised in the presence or absence of a non-kin breeding group. Upon adulthood, subjects underwent social interaction tests, an open field test, and a novel object test. Subjects were then exposed to a novel conspecific and novel group and neural responses were quantified via immunohistochemical staining in brain regions associated with social behavior. RESULTS Early life social experience did not influence behavior in the test battery, but it did influence social processing. In animals exposed to non-kin during development, adult lateral septal neural responses toward a novel conspecific were weaker and hypothalamic neural responses toward a mixed-sex group were stronger. CONCLUSIONS Communal species may exhibit robust behavioral resilience to the early life social environment. But the early life environment can affect how novel social information is processed in the brain during adulthood, with long-term consequences that are likely to shape their behavioral trajectory.
Collapse
Affiliation(s)
| | | | - Mutian Li
- Department of Psychology, Emory University, Atlanta, USA
| | - Aubrey M Kelly
- Department of Psychology, Emory University, Atlanta, USA
| |
Collapse
|
2
|
Jiang Z, Zhou Y, Zhou Y, Yang D, Li J, Li Y, Fan Q, Lin J. Exploring the bidirectional causal relationship between Autism Spectrum Disorder and Schizophrenia using Mendelian randomization. Medicine (Baltimore) 2025; 104:e42119. [PMID: 40228250 PMCID: PMC11999464 DOI: 10.1097/md.0000000000042119] [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: 08/22/2024] [Accepted: 03/25/2025] [Indexed: 04/16/2025] Open
Abstract
Autism Spectrum Disorder (ASD), characterized mainly by stereotyped behaviors and social impairments, affects about one in 100 children worldwide. Schizophrenia (SCZ), a chronic mental illness, affects 1% of the global population. The pathogenesis and specific treatment strategies for ASD and SCZ remain unclear. Previous research has suggested similarities in SCZ and ASD etiology and symptoms. However, no definitive correlation has been confirmed. Therefore, we conducted a Mendelian randomization study to assess the relationship between SCZ and ASD, providing new insights into their etiology and treatment. We used the two-sample Mendelian randomization (TSMR) approach to investigate the bidirectional causal association between SCZ and ASD, employing summary-level genome-wide association studies (GWAS) data. ASD summary data from the IEU GWAS database and SCZ summary data from the Psychiatric Genomics Consortium (PGC) were used as exposure and outcome variables, respectively. Statistical analysis was performed using the TwoSampleMR package in R version 4.3.2, with sensitivity analysis conducted to verify the result's reliability. Based on the results of the MR analysis, we retrieved and analyzed the relevant genetic information from the GWAS Catalog. TSMR analysis revealed higher ASD risk in SCZ (IVW: OR: 1.19, 95% CI: 1.12-1.26, P < .001). Bidirectional MR analysis confirmed a causal relationship between ASD and SCZ (IVW: scz2018clozuk (Clozapine UK), OR: 1.12, 95% CI: 1.04-1.21, P = .003; scz2019asi, OR: 1.14, 95% CI: 1.05-1.23, P = .002). Our study demonstrated a bidirectional relationship between SCZ and ASD in the European population, suggesting that each may induce the onset of the other.
Collapse
Affiliation(s)
- Ziqing Jiang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yiying Zhou
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yingxin Zhou
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Dongmei Yang
- Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Jingjun Li
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yongchun Li
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qin Fan
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Jintao Lin
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
3
|
Manjur SM, Diaz LRM, Lee IO, Skuse DH, Thompson DA, Marmolejos-Ramos F, Constable PA, Posada-Quintero HF. Detecting Autism Spectrum Disorder and Attention Deficit Hyperactivity Disorder Using Multimodal Time-Frequency Analysis with Machine Learning Using the Electroretinogram from Two Flash Strengths. J Autism Dev Disord 2025; 55:1365-1378. [PMID: 38393437 DOI: 10.1007/s10803-024-06290-w] [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] [Accepted: 02/13/2024] [Indexed: 02/25/2024]
Abstract
PURPOSE Autism spectrum disorder (ASD) and attention deficit hyperactivity disorder (ADHD) are conditions that similarly alter cognitive functioning ability and challenge the social interaction, attention, and communication skills of affected individuals. Yet these are distinct neurological conditions that can exhibit diverse characteristics which require different management strategies. It is desirable to develop tools to assist with early distinction so that appropriate early interventions and support may be tailored to an individual's specific requirements. The current diagnostic procedures for ASD and ADHD require a multidisciplinary approach and can be lengthy. This study investigated the potential of electroretinogram (ERG), an eye test measuring retinal responses to light, for rapid screening of ASD and ADHD. METHODS Previous studies identified differences in ERG amplitude between ASD and ADHD, but this study explored time-frequency analysis (TFS) to capture dynamic changes in the signal. ERG data from 286 subjects (146 control, 94 ASD, 46 ADHD) was analyzed using two TFS techniques. RESULTS Key features were selected, and machine learning models were trained to classify individuals based on their ERG response. The best model achieved 70% overall accuracy in distinguishing control, ASD, and ADHD groups. CONCLUSION The ERG to the stronger flash strength provided better separation and the high frequency dynamics (80-300 Hz) were more informative features than lower frequency components. To further improve classification a greater number of different flash strengths may be required along with a discrimination comparison to participants who meet both ASD and ADHD classifications and carry both diagnoses.
Collapse
Affiliation(s)
| | | | - Irene O Lee
- Behavioral and Brain Sciences Unit, Population Policy and Practice Program, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - David H Skuse
- Behavioral and Brain Sciences Unit, Population Policy and Practice Program, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Dorothy A Thompson
- Tony Kriss Visual Electrophysiology Unit, Clinical and Academic Department of Ophthalmology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
- UCL Great Ormond Street Institute for Child Health, University College London, London, UK
| | | | - Paul A Constable
- College of Nursing and Health Sciences, Flinders University, Caring Futures Institute, Adelaide, Australia
| | - Hugo F Posada-Quintero
- Department of Biomedical Engineering, University of Connecticut, 06269, Storrs, CT, USA.
| |
Collapse
|
4
|
King C, Plakke B. Maternal choline supplementation in neurodevelopmental disorders: mechanistic insights from animal models and future directions. Nutr Neurosci 2025; 28:405-424. [PMID: 39046330 DOI: 10.1080/1028415x.2024.2377084] [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] [Indexed: 07/25/2024]
Abstract
OBJECTIVES To synthesize evidence from animal models of neurodevelopmental disorders (NDD) using maternal choline supplementation, to characterize current knowledge on the mechanisms of choline's protective effects against NDD, and to identify gaps in knowledge for future study. METHODS A literature review was conducted in PubMed to identify studies using prenatal choline supplementation interventions in rodent models of neurodevelopmental disorders. 24 studies were identified, and behavioral and biological findings were extracted from each. Studies examining both genetic and environmental risk factors were included. RESULTS Maternal choline supplementation during gestation is protective against both genetic and environmental NDD risk factors. Maternal choline supplementation improves both cognitive and affective outcomes throughout the lifespan in NDD models. Prenatal choline improved these outcomes through its participation in processes like neurogenesis, epigenetic regulation, and anti-inflammatory signaling. DISCUSSION Maternal choline supplementation improves behavioral and neurobiological outcomes in animal models of NDD, paralleling findings in humans. Animal models provide a unique opportunity to study the mechanisms by which gestational choline improves neurodevelopmental outcomes. This is especially important since nearly 90% of pregnant people in the United States are deficient in choline intake. However, much is still unknown about the mechanisms through which choline and its derivatives act. Further research into this topic, especially mechanistic studies in animal models, is critical to modernize maternal choline intake guidelines and to develop interventions to increase maternal choline intake in vulnerable populations.
Collapse
Affiliation(s)
- Cole King
- Psychological Sciences, Kansas State University, Manhattan, KS, USA
| | - Bethany Plakke
- Psychological Sciences, Kansas State University, Manhattan, KS, USA
| |
Collapse
|
5
|
Ashitomi H, Nakagawa T, Nakagawa M, Hosoi T. Cullin-RING Ubiquitin Ligases in Neurodevelopment and Neurodevelopmental Disorders. Biomedicines 2025; 13:810. [PMID: 40299365 PMCID: PMC12024872 DOI: 10.3390/biomedicines13040810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 04/30/2025] Open
Abstract
Ubiquitination is a dynamic and tightly regulated post-translational modification essential for modulating protein stability, trafficking, and function to preserve cellular homeostasis. This process is orchestrated through a hierarchical enzymatic cascade involving three key enzymes: the E1 ubiquitin-activating enzyme, the E2 ubiquitin-conjugating enzyme, and the E3 ubiquitin ligase. The final step of ubiquitination is catalyzed by the E3 ubiquitin ligase, which facilitates the transfer of ubiquitin from the E2 enzyme to the substrate, thereby dictating which proteins undergo ubiquitination. Emerging evidence underscores the critical roles of ubiquitin ligases in neurodevelopment, regulating fundamental processes such as neuronal polarization, axonal outgrowth, synaptogenesis, and synaptic function. Mutations in genes encoding ubiquitin ligases and the consequent dysregulation of these pathways have been increasingly implicated in a spectrum of neurodevelopmental disorders, including autism spectrum disorder, intellectual disability, and attention-deficit/hyperactivity disorder. This review synthesizes current knowledge on the molecular mechanisms underlying neurodevelopment regulated by Cullin-RING ubiquitin ligases-the largest subclass of ubiquitin ligases-and their involvement in the pathophysiology of neurodevelopmental disorders. A deeper understanding of these mechanisms holds significant promise for informing novel therapeutic strategies, ultimately advancing clinical outcomes for individuals affected by neurodevelopmental disorders.
Collapse
Affiliation(s)
- Honoka Ashitomi
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Sanyo-Onoda 756-0084, Japan; (H.A.)
| | - Tadashi Nakagawa
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Sanyo-Onoda 756-0084, Japan; (H.A.)
- Division of Cell Proliferation, United Centers for Advanced Research and Translational Medicine, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan
| | - Makiko Nakagawa
- Institute of Gene Research, Yamaguchi University Science Research Center, Ube 755-8505, Japan
- Advanced Technology Institute, Life Science Division, Yamaguchi University, Ube 755-8611, Japan
| | - Toru Hosoi
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Sanyo-Onoda 756-0084, Japan; (H.A.)
| |
Collapse
|
6
|
Wiśniewska K, Wolski J, Anikiej-Wiczenbach P, Żabińska M, Węgrzyn G, Pierzynowska K. Behavioural disorders and sleep problems in Sanfilippo syndrome: overlaps with some other conditions and importance indications. Eur Child Adolesc Psychiatry 2025:10.1007/s00787-025-02661-5. [PMID: 40087177 DOI: 10.1007/s00787-025-02661-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 02/07/2025] [Indexed: 03/17/2025]
Abstract
Sanfilippo syndrome (MPS III) is one of the types of mucopolysaccharidoses (MPS), a group of inherited metabolic diseases in which the accumulation of glycosaminoglycans (GAGs) results from deficiency of different lysosomal enzymes. The hallmarks of MPS III are relatively minor somatic abnormalities with severe and progressive central nervous system (CNS) symptoms. An analysis of the literature showed that the biggest problems for carers of people with MPS III are behavioural disorders and sleep disorders. Despite extensive discussions on improving the quality of life of patients, little attention was paid to the families/carers of patients. The families/carers are providing appropriate medical and palliative care to the patient every day due to their loss of mobility, self-care skills, tube feeding, airway clearance and other supports continue to have an adverse effect on the quality of life of families/carers. However, a literature review of possible solutions showed that effective methods (both pharmacological and non-pharmacological) exist. The needs of carers of MPS III patients should receive as much attention as the search for new treatments. There are many options for dealing with such problems. The key issue is to identify the source of the problem and choose the most effective therapy. Alleviating behavioural disorders, pain complaints and sleep problems will have a positive impact not only on the quality of life of carers/families, but also on the patients themselves.
Collapse
Affiliation(s)
- Karolina Wiśniewska
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, 80-308, Gdansk, Poland
| | - Jakub Wolski
- Psychiatry Ward, 7, Navy Hospital in Gdańsk, Polanki 117, 80-305, Gdańsk, Poland
| | | | - Magdalena Żabińska
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, 80-308, Gdansk, Poland
| | - Grzegorz Węgrzyn
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, 80-308, Gdansk, Poland
| | - Karolina Pierzynowska
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, 80-308, Gdansk, Poland.
| |
Collapse
|
7
|
Fattal J, McAdams DP, Mittal VA. Interpersonal synchronization: An overlooked factor in development, social cognition, and psychopathology. Neurosci Biobehav Rev 2025; 170:106037. [PMID: 39929382 DOI: 10.1016/j.neubiorev.2025.106037] [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: 10/31/2024] [Revised: 12/31/2024] [Accepted: 01/31/2025] [Indexed: 02/18/2025]
Abstract
Intact social functioning relies on a combination of explicit and implicit behavioral, attentional, and interpersonal processes referred to as "social cognition". Characterizing these interpersonal processes forms a critical underpinning to understanding and treating psychopathology, particularly in disorders where deficits in social functioning do not emerge as a secondary symptom but rather as an essential feature of the disorder. Two of such disorders are autism spectrum disorders (ASD) and schizophrenia spectrum disorders (SZ). However, despite the substantial overlap in the features of social dysfunction between ASD and SZ, including social cognitive deficits in theory of mind, perspective-taking, and empathy, there is a limited understanding of the mechanisms underlying those shared deficits, and how to treat them. We suggest that disruptions of interpersonal functioning emerge over the course of development, and that interpersonal synchronization, a phenomenon in which behavioral and physiological cues align between interacting partners, forms a critical component of social cognition that underlies the disruption in social functioning in ASD and SZ. We present a conceptual review of typical and atypical development of social processes and highlight the role of interpersonal synchronization across the course of development. Then, we review the existing evidence suggesting impairments in both the intentional and spontaneous synchronization of interpersonal processes in ASD and SZ, as well as studies suggesting that interpersonal synchronization and clinical symptoms may be improved through body-oriented interventions within these disorders. Finally, we suggest potential mechanisms that may underpin typical and atypical development of interpersonal synchronization.
Collapse
Affiliation(s)
- Jessica Fattal
- Northwestern University, Department of Psychology, Swift Hall, 2029 Sheridan Road, Evanston, IL 60208, USA.
| | - Dan P McAdams
- Northwestern University, Department of Psychology, Swift Hall, 2029 Sheridan Road, Evanston, IL 60208, USA
| | - Vijay A Mittal
- Northwestern University, Department of Psychology, Swift Hall, 2029 Sheridan Road, Evanston, IL 60208, USA
| |
Collapse
|
8
|
Sigar P, Kathrein N, Gragas E, Kupis L, Uddin LQ, Nomi JS. Age-related changes in brain signal variability in autism spectrum disorder. Mol Autism 2025; 16:8. [PMID: 39923093 PMCID: PMC11806755 DOI: 10.1186/s13229-024-00631-3] [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: 02/07/2024] [Accepted: 11/25/2024] [Indexed: 02/10/2025] Open
Abstract
BACKGROUND Brain signal variability (BSV) is an important understudied aspect of brain function linked to cognitive flexibility and adaptive behavior. Autism spectrum disorder (ASD) is a neurodevelopmental condition characterized by social communication difficulties and restricted and repetitive behaviors (RRBs). While atypical brain function has been identified in individuals with ASD using fMRI task-activation and functional connectivity approaches, little is known about age-related relationships with resting-state BSV and repetitive behaviors in ASD. METHODS We conducted a cross-sectional examination of resting-state BSV and its relationship with age and RRBs in a cohort of individuals with Autism Brain Imaging Data Exchange (n = 351) and typically developing (TD) individuals (n = 402) aged 5-50 years obtained from the Autism Brain Imaging Data Exchange. RRBs were assessed using the Autism Diagnostic Interview-Revised (ADI-RRB) scale. BSV was quantified using the root-mean-square successive difference (rMSSD) of the resting-state fMRI time series. We examined categorical group differences in rMSSD between ASD and TD groups, controlling for both linear and quadratic age. To identify dimensional relationships between age, group, and rMSSD, we utilized interaction regressors for group x age and group x quadratic age. Within a subset of individuals with ASD (269 subjects), we explored the relationship between rMSSD and ADI-RRB scores, both with and without age considerations. The relationship between rMSSD and ADI-RRB scores was further analyzed while accounting for linear and quadratic age. Additionally, we investigated the relationship between BSV, age, and ADI-RRB scores using interaction regressors for age x RRB and quadratic age x RRB. RESULTS When controlling for linear age effects, we observed significant group differences between individuals with ASD and TD individuals in the default-mode network (DMN) and visual network, with decreased BSV in ASD. Similarly, controlling for quadratic age effects revealed significant group differences in the DMN and visual network. In both cases, individuals with ASD showed decreased BSV compared with TD individuals in these brain regions. The group × age interaction demonstrated significant group differences in the DMN, and visual network brain areas, indicating that rMSSD was greater in older individuals compared with younger individuals in the ASD group, while rMSSD was greater in younger individuals compared with older individuals in the TD group. The group × quadratic age interaction showed significant differences in the brain regions included in DMN, with an inverted U-shaped rMSSD-age relationship in ASD (higher rMSSD in younger individuals that slightly increased into middle age before decreasing) and a U-shaped rMSSD-age relationship in TD (higher rMSSD in younger and older individuals compared with middle-aged individuals). When controlling for linear and quadratic age effects, we found a significant positive association between rMSSD and ADI-RRB scores in brain regions within the DMN, salience, and visual network. While no significant results were observed for the linear age × RRB interaction, a significant association between quadratic age and ADI-RRB scores emerged in the DMN, dorsal attention network, and sensorimotor network. Individuals with high ADI-RRB scores exhibited an inverted U-shaped relationship between rMSSD and age, with lower rMSSD levels observed in both younger and older individuals, and higher rMSSD in middle-aged individuals. Those with mid-range ADI-RRB scores displayed a weak inverted U-shaped rMSSD-age association. In contrast, individuals with low ADI-RRB scores showed a U-shaped rMSSD-age association, with higher rMSSD levels in younger and older individuals, but a lower rMSSD in middle-aged individuals. CONCLUSION These findings highlight age-related atypical BSV patterns in ASD and their association with repetitive behaviors, contributing to the growing literature on understanding alterations in functional brain maturation in ASD.
Collapse
Affiliation(s)
- Priyanka Sigar
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, 90024, USA.
| | - Nicholas Kathrein
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, 90024, USA
| | - Elijah Gragas
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, 90024, USA
| | - Lauren Kupis
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, 90024, USA
| | - Lucina Q Uddin
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, 90024, USA
- Department of Psychology, University of California Los Angeles, Los Angeles, CA, 90024, USA
| | - Jason S Nomi
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, 90024, USA.
| |
Collapse
|
9
|
Gao X, Xu G, Fu N, Ben Q, Wang L, Bu X. The effectiveness of music therapy in improving behavioral symptoms among children with autism spectrum disorders: a systematic review and meta-analysis. Front Psychiatry 2025; 15:1511920. [PMID: 39896995 PMCID: PMC11783185 DOI: 10.3389/fpsyt.2024.1511920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 12/18/2024] [Indexed: 02/04/2025] Open
Abstract
Objectives This comprehensive review and meta-analysis aimed to thoroughly identify the effectiveness of music therapy (MT) in improving behavioral symptoms in children with autism spectrum disorders (ASD) by analyzing the data from all relevant randomized controlled trials (RCTs) related to this field. Methods From inception until September 18, 2024, PubMed, Web of Science, the Cochrane Library, SinoMed, and Embase were searched. Two reviewers extracted the data separately, and any controversies between the authors' assessments were resolved by conversation or speaking with another author. The behavioral symptoms scale score before and after the intervention was taken from the included trials and used to reflect the therapeutic effect of music therapy in children with autism. Results 2607 records across all retrieved databases were discovered, thirteen of which were included in a meta-analysis with 1160 participants. According to the meta-analysis, children with autism showed a substantial improvement in their behavior symptoms when receiving music treatment (standardized mean difference [SMD] = -0.66, 95% confidence interval [CI]: -0.93 to -0.39, p < 0.001). With I 2 = 78% and P < 0.001, we did discover a medium level of heterogeneity among the included studies. Conclusions MT has a positive impact on improving behavioral symptoms in children with autism. However, given the significant heterogeneity and limitations in this study, RCTs with rigorous methodological quality are still required to confirm the curative benefits of MT in autistic children precisely. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/, identifier CRD42024597939.
Collapse
Affiliation(s)
- Xiuyan Gao
- School of Nursing, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Guangjun Xu
- School of Health Management, Liaoyang Vocational College of Technology, Liaoyang, Liaoning, China
| | - Ningning Fu
- School of Nursing, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Qi Ben
- School of Nursing, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Lin Wang
- School of Nursing, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Xiumei Bu
- School of Nursing, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| |
Collapse
|
10
|
Jończyk J, Przybylska K, Staszewski M, Godyń J, Werner T, Stefaniak-Napieralska M, Stark H, Walczyński K, Bajda M. Virtual Screening Approaches to Identify Promising Multitarget-Directed Ligands for the Treatment of Autism Spectrum Disorder. Molecules 2024; 29:5271. [PMID: 39598660 PMCID: PMC11596355 DOI: 10.3390/molecules29225271] [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: 09/30/2024] [Revised: 10/30/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024] Open
Abstract
Autism spectrum disorder is a complex neurodevelopmental disorder. The available medical treatment options for autism spectrum disorder are very limited. While the etiology and pathophysiology of autism spectrum disorder are still not fully understood, recent studies have suggested that wide alterations in the GABAergic, glutamatergic, cholinergic, and serotonergic systems play a key role in its development and progression. Histamine neurotransmission is known to have complex interactions with other neurotransmitters that fit perfectly into the complex etiology of this disease. Multitarget-directed compounds with an affinity for the histamine H3 receptor indicate an interesting profile of activity against autism spectrum disorder in animal models. Here, we present the results of our research on the properties of (4-piperazin-1-ylbutyl)guanidine derivatives acting on histamine H3 receptors as potential multitarget ligands. Through the virtual screening approach, we identified promising ligands among 32 non-imidazole histamine H3 receptor antagonists/inverse agonists with potential additional activity against the dopamine D2 receptor and/or cholinesterases. The virtual screening protocol integrated predictions from SwissTargetPrediction, SEA, and PPB2 tools, along with molecular docking simulations conducted using GOLD 5.3 and Glide 7.5 software. Among the selected ligands, compounds 25 and 30 blocked radioligand binding to the D2 receptor at over 50% at a screening concentration of 1 µM. Further experiments allowed us to determine the pKi value at the D2 receptor of 6.22 and 6.12 for compounds 25 and 30, respectively. Our findings suggest that some of the tested compounds could be promising multitarget-directed ligands for the further research and development of more effective treatments for autism spectrum disorder.
Collapse
Affiliation(s)
- Jakub Jończyk
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, 30-688 Kraków, Poland; (J.J.); (J.G.)
- Sano—Centre for Computational Medicine, 30-054 Kraków, Poland
| | - Klaudia Przybylska
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, 30-688 Kraków, Poland; (J.J.); (J.G.)
| | - Marek Staszewski
- Department of Synthesis and Technology of Drugs, Faculty of Pharmacy, Medical University of Lodz, 90-151 Lodz, Poland; (M.S.); (M.S.-N.); (K.W.)
| | - Justyna Godyń
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, 30-688 Kraków, Poland; (J.J.); (J.G.)
| | - Tobias Werner
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University, 40225 Düsseldorf, Germany; (T.W.); (H.S.)
| | - Monika Stefaniak-Napieralska
- Department of Synthesis and Technology of Drugs, Faculty of Pharmacy, Medical University of Lodz, 90-151 Lodz, Poland; (M.S.); (M.S.-N.); (K.W.)
| | - Holger Stark
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University, 40225 Düsseldorf, Germany; (T.W.); (H.S.)
| | - Krzysztof Walczyński
- Department of Synthesis and Technology of Drugs, Faculty of Pharmacy, Medical University of Lodz, 90-151 Lodz, Poland; (M.S.); (M.S.-N.); (K.W.)
| | - Marek Bajda
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, 30-688 Kraków, Poland; (J.J.); (J.G.)
| |
Collapse
|
11
|
Marangelo C, Vernocchi P, Del Chierico F, Scanu M, Marsiglia R, Petrolo E, Fucà E, Guerrera S, Valeri G, Vicari S, Putignani L. Stratification of Gut Microbiota Profiling Based on Autism Neuropsychological Assessments. Microorganisms 2024; 12:2041. [PMID: 39458350 PMCID: PMC11510388 DOI: 10.3390/microorganisms12102041] [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: 09/13/2024] [Revised: 10/04/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder. Investigations of gut microbiota (GM) play an important role in deciphering disease severity and symptoms. Overall, we stratified 70 ASD patients by neuropsychological assessment, based on Calibrated Severity Scores (CSSs) of the Autism Diagnostic Observation Schedule-Second edition (ADOS-2), Child Behavior Checklist (CBCL) and intelligent quotient/developmental quotient (IQ/DQ) parameters. Hence, metataxonomy and PICRUSt-based KEGG predictions of fecal GM were assessed for each clinical subset. Here, 60% of ASD patients showed mild to moderate autism, while the remaining 40% showed severe symptoms; 23% showed no clinical symptoms, 21% had a risk of behavior problems and 56% had clinical symptoms based on the CBCL, which assesses internalizing problems; further, 52% had no clinical symptoms, 21% showed risk, and 26% had clinical symptoms classified by CBCL externalizing problems. Considering the total CBCL index, 34% showed no clinical symptoms, 13% showed risk, and 52% had clinical symptoms. Here, 70% of ASD patients showed cognitive impairment/developmental delay (CI/DD). The GM of ASDs with severe autism was characterized by an increase in Veillonella, a decrease in Monoglobus pectinilyticus and a higher microbial dysbiosis index (MDI) when compared to mild-moderate ASDs. Patients at risk for behavior problems and showing clinical symptoms were characterized by a GM with an increase of Clostridium, Eggerthella, Blautia, Intestinibacter, Coprococcus, Ruminococcus, Onthenecus and Bariatricus, respectively. Peptidoglycan biosynthesis and biofilm formation KEGGs characterized patients with clinical symptoms, while potential microbiota-activated PPAR-γ-signaling was seen in CI/DD patients. This evidence derived from GM profiling may be used to further improve ASD understanding, leasing to a better comprehension of the neurological phenotype.
Collapse
Affiliation(s)
- Chiara Marangelo
- Research Unit of Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (C.M.); (P.V.); (F.D.C.); (M.S.); (R.M.)
| | - Pamela Vernocchi
- Research Unit of Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (C.M.); (P.V.); (F.D.C.); (M.S.); (R.M.)
| | - Federica Del Chierico
- Research Unit of Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (C.M.); (P.V.); (F.D.C.); (M.S.); (R.M.)
| | - Matteo Scanu
- Research Unit of Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (C.M.); (P.V.); (F.D.C.); (M.S.); (R.M.)
| | - Riccardo Marsiglia
- Research Unit of Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (C.M.); (P.V.); (F.D.C.); (M.S.); (R.M.)
| | - Emanuela Petrolo
- Child and Adolescent Neuropsychiatry Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (E.P.); (E.F.); (S.G.); (G.V.); or (S.V.)
| | - Elisa Fucà
- Child and Adolescent Neuropsychiatry Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (E.P.); (E.F.); (S.G.); (G.V.); or (S.V.)
| | - Silvia Guerrera
- Child and Adolescent Neuropsychiatry Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (E.P.); (E.F.); (S.G.); (G.V.); or (S.V.)
| | - Giovanni Valeri
- Child and Adolescent Neuropsychiatry Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (E.P.); (E.F.); (S.G.); (G.V.); or (S.V.)
| | - Stefano Vicari
- Child and Adolescent Neuropsychiatry Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (E.P.); (E.F.); (S.G.); (G.V.); or (S.V.)
- Life Sciences and Public Health Department, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Lorenza Putignani
- Unit of Microbiomics and Research Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy
| |
Collapse
|
12
|
Gouda B, Sinha SN, Sangaraju R, Huynh T, Patangay S, Venkata Mullapudi S, Mungamuri SK, Patil PB, Periketi MC. Extraction, Phytochemical profile, and neuroprotective activity of Phyllanthus emblica fruit extract against sodium valproate-induced postnatal autism in BALB/c mice. Heliyon 2024; 10:e34992. [PMID: 39157403 PMCID: PMC11327600 DOI: 10.1016/j.heliyon.2024.e34992] [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: 11/28/2023] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 08/20/2024] Open
Abstract
The aim of the present study was to evaluate the effect of the ethyl acetate fraction of amla (EAFA) extract on valproic acid (VPA)-induced postnatal autism in BALB/c mice. Our study revealed that mice treated with VPA on postnatal day 14 (PND14) showed significant abnormal behaviours such as social interaction, social affiliation, anxiety, and motor coordination compared to the control group, while EAFA extract treatment (100 mg/kg) ameliorated these symptoms. Our study highlights the protective effect of EAFA extract on improving behavioural alterations, significantly restoring anti-oxidative enzymes such as GST and GR, and reducing MDA and NO levels. Furthermore, the EAFA-treated group significantly lowered the proinflammatory markers (IL-1β and TNF-α) and the expression of up-regulated 5-HT1D, 5-HT2A, and D2 receptor proteins. Based on histopathological studies, the percentage of neuronal injury in the EAFA-treated group as well as cellular structural changes were reduced using SEM analysis. In conclusion, the present study suggests that treatment with EAFA extract ameliorates VPA-induced autism due to its anti-oxidant and neuroprotective activity.
Collapse
Affiliation(s)
- Balaji Gouda
- Division of Food Safety, Indian Council of Medical Research-National Institute of Nutrition, Jamai-Osmania, Tarnaka, Hyderabad, Telangana- 500007, India
- Department of Pharmacy, University College of Technology, Osmania University, Hyderabad-500027, India
| | - Sukesh Narayan Sinha
- Division of Food Safety, Indian Council of Medical Research-National Institute of Nutrition, Jamai-Osmania, Tarnaka, Hyderabad, Telangana- 500007, India
| | - Rajendra Sangaraju
- Division of Food Safety, Indian Council of Medical Research-National Institute of Nutrition, Jamai-Osmania, Tarnaka, Hyderabad, Telangana- 500007, India
- Head of Biology, Department of Biosciences and Food Technology, STEM College, RMIT University, PO Box 71, Bundoora, VIC 3083, Australia
| | - Tien Huynh
- Head of Biology, Department of Biosciences and Food Technology, STEM College, RMIT University, PO Box 71, Bundoora, VIC 3083, Australia
| | - Shashikala Patangay
- Department of Pharmacy, University College of Technology, Osmania University, Hyderabad-500027, India
| | - Surekha Venkata Mullapudi
- Division of Pathology and Microbiology, Indian Council of Medical Research-National Institute of Nutrition, Jamai-Osmania, Tarnaka, Hyderabad, Telangana- 500007, India
| | - Sathish Kumar Mungamuri
- Division of Food Safety, Indian Council of Medical Research-National Institute of Nutrition, Jamai-Osmania, Tarnaka, Hyderabad, Telangana- 500007, India
| | - Pradeep B. Patil
- Animal Facility Division, Indian Council of Medical Research-National Institute of Nutrition, Jamai-Osmania, Tarnaka, Hyderabad, Telangana- 500007, India
| | - Madhusudhana Chary Periketi
- SEM Facility, Cell Biology Division, Indian Council of Medical Research-National Institute of Nutrition, Jamai-Osmania, Tarnaka, Hyderabad, Telangana- 500007, India
| |
Collapse
|
13
|
Blum K, Bowirrat A, Sunder K, Thanos PK, Hanna C, Gold MS, Dennen CA, Elman I, Murphy KT, Makale MT. Dopamine Dysregulation in Reward and Autism Spectrum Disorder. Brain Sci 2024; 14:733. [PMID: 39061473 PMCID: PMC11274922 DOI: 10.3390/brainsci14070733] [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: 05/31/2024] [Revised: 07/09/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Autism spectrum disorder (ASD) is primarily characterized by core deficits in social skills, communication, and cognition and by repetitive stereotyped behaviors. These manifestations are variable between individuals, and ASD pathogenesis is complex, with over a thousand implicated genes, many epigenetic factors, and multiple environmental influences. The mesolimbic dopamine (DA) mediated brain reward system is held to play a key role, but the rapidly expanding literature reveals intricate, nuanced signaling involving a wide array of mesolimbic loci, neurotransmitters and receptor subtypes, and neuronal variants. How altered DA signaling may constitute a downstream convergence of the manifold causal origins of ASD is not well understood. A clear working framework of ASD pathogenesis may help delineate common stages and potential diagnostic and interventional opportunities. Hence, we summarize the known natural history of ASD in the context of emerging data and perspectives to update ASD reward signaling. Then, against this backdrop, we proffer a provisional framework that organizes ASD pathogenesis into successive levels, including (1) genetic and epigenetic changes, (2) disrupted mesolimbic reward signaling pathways, (3) dysregulated neurotransmitter/DA signaling, and finally, (4) altered neurocognitive and social behavior and possible antagonist/agonist based ASD interventions. This subdivision of ASD into a logical progression of potentially addressable parts may help facilitate the rational formulation of diagnostics and targeted treatments.
Collapse
Affiliation(s)
- Kenneth Blum
- Division of Addiction Research & Education, Center for Exercise Sports, Mental Health, Western University of Health Sciences, Pomona, CA 91766, USA
- Sunder Foundation, Palm Springs, CA 92264, USA
- Division of Personalized Neuromodulations, PeakLogic, LLC, Del Mar, CA 92130, USA
| | - Abdalla Bowirrat
- Department of Molecular Biology, Adelson School of Medicine, Ariel University, Ariel 40700, Israel
| | | | - Panayotis K. Thanos
- Department of Pharmacology and Toxicology, State University of New York, SUNY, Buffalo, NY 14215, USA
| | - Colin Hanna
- Department of Pharmacology and Toxicology, State University of New York, SUNY, Buffalo, NY 14215, USA
| | - Mark S. Gold
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Catherine A. Dennen
- Department of Family Medicine, Jefferson Health Northeast, Philadelphia, PA 19145, USA
| | - Igor Elman
- Department of Psychiatry, Harvard University School of Medicine, Cambridge, MA 02215, USA
| | - Kevin T. Murphy
- Division of Personalized Neuromodulations, PeakLogic, LLC, Del Mar, CA 92130, USA
| | - Milan T. Makale
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
14
|
Al-Beltagi M, Saeed NK, Bediwy AS, Elbeltagi R. Metabolomic changes in children with autism. World J Clin Pediatr 2024; 13:92737. [PMID: 38947988 PMCID: PMC11212761 DOI: 10.5409/wjcp.v13.i2.92737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 04/23/2024] [Accepted: 05/06/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) is a neurodevelopmental condition characterized by deficits in social communication and repetitive behaviors. Metabolomic profiling has emerged as a valuable tool for understanding the underlying metabolic dysregulations associated with ASD. AIM To comprehensively explore metabolomic changes in children with ASD, integrating findings from various research articles, reviews, systematic reviews, meta-analyses, case reports, editorials, and a book chapter. METHODS A systematic search was conducted in electronic databases, including PubMed, PubMed Central, Cochrane Library, Embase, Web of Science, CINAHL, Scopus, LISA, and NLM catalog up until January 2024. Inclusion criteria encompassed research articles (83), review articles (145), meta-analyses (6), systematic reviews (6), case reports (2), editorials (2), and a book chapter (1) related to metabolomic changes in children with ASD. Exclusion criteria were applied to ensure the relevance and quality of included studies. RESULTS The systematic review identified specific metabolites and metabolic pathways showing consistent differences in children with ASD compared to typically developing individuals. These metabolic biomarkers may serve as objective measures to support clinical assessments, improve diagnostic accuracy, and inform personalized treatment approaches. Metabolomic profiling also offers insights into the metabolic alterations associated with comorbid conditions commonly observed in individuals with ASD. CONCLUSION Integration of metabolomic changes in children with ASD holds promise for enhancing diagnostic accuracy, guiding personalized treatment approaches, monitoring treatment response, and improving outcomes. Further research is needed to validate findings, establish standardized protocols, and overcome technical challenges in metabolomic analysis. By advancing our understanding of metabolic dysregulations in ASD, clinicians can improve the lives of affected individuals and their families.
Collapse
Affiliation(s)
- Mohammed Al-Beltagi
- Department of Pediatric, Faculty of Medicine, Tanta University, Tanta 31511, Alghrabia, Egypt
- Department of Pediatric, University Medical Center, King Abdulla Medical City, Arabian Gulf University, Manama 26671, Bahrain
- Department of Pediatric, University Medical Center, Dr. Sulaiman Al Habib Medical Group, Manama, Bahrain, Manama 26671, Bahrain
| | - Nermin Kamal Saeed
- Medical Microbiology Section, Department of Pathology, Salmaniya Medical Complex, Ministry of Health, Kingdom of Bahrain, Manama 12, Bahrain
- Medical Microbiology Section, Department of Pathology, Irish Royal College of Surgeon, Bahrain, Busaiteen 15503, Muharraq, Bahrain
| | - Adel Salah Bediwy
- Department of Pulmonology, Faculty of Medicine, Tanta University, Tanta 31527, Alghrabia, Egypt
- Department of Chest Disease, University Medical Center, King Abdulla Medical City, Arabian Gulf University, Manama 26671, Bahrain
- Department of Chest Disease, University Medical Center, Dr. Sulaiman Al Habib Medical Group, Manama, Manama 26671, Bahrain
| | - Reem Elbeltagi
- Department of Medicine, The Royal College of Surgeons in Ireland - Bahrain, Busiateen 15503, Muharraq, Bahrain
| |
Collapse
|
15
|
Dai H, Jiang Y, Liu S, Li D, Zhang X. Dietary flavonoids modulate the gut microbiota: A new perspective on improving autism spectrum disorder through the gut-brain axis. Food Res Int 2024; 186:114404. [PMID: 38729686 DOI: 10.1016/j.foodres.2024.114404] [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: 12/19/2023] [Revised: 04/18/2024] [Accepted: 04/20/2024] [Indexed: 05/12/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder with an unknown etiology. It is associated with various factors and causes great inconvenience to the patient's life. The gut-brain axis (GBA), which serves as a bidirectional information channel for exchanging information between the gut microbiota and the brain, is vital in studying many neurodegenerative diseases. Dietary flavonoids provide anti-inflammatory and antioxidant benefits, as well as regulating the structure and function of the gut microbiota. The occurrence and development of ASD are associated with dysbiosis of the gut microbiota. Modulation of gut microbiota can effectively improve the severity of ASD. This paper reviews the links between gut microbiota, flavonoids, and ASD, focusing on the mechanism of dietary flavonoids in regulating ASD through the GBA.
Collapse
Affiliation(s)
- Haochen Dai
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, PR China
| | - Yuhan Jiang
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, PR China
| | - Shuxun Liu
- College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, PR China.
| | - Dandan Li
- Sinograin Chengdu Storage Research Institute Co., Ltd, Chengdu 610091, PR China
| | - Xin Zhang
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, PR China.
| |
Collapse
|
16
|
Shi Z, Wang S, Chen M, Hu A, Long Q, Lee Y. The effect of music therapy on language communication and social skills in children with autism spectrum disorder: a systematic review and meta-analysis. Front Psychol 2024; 15:1336421. [PMID: 38774719 PMCID: PMC11106491 DOI: 10.3389/fpsyg.2024.1336421] [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: 11/13/2023] [Accepted: 04/12/2024] [Indexed: 05/24/2024] Open
Abstract
Background Studies have shown that music therapy can be used as a therapeutic aid for clinical disorders. To evaluate the effects of music therapy (MT) on language communication and social skills in children with autism spectrum disorder (ASD), a meta-analysis was performed on eligible studies in this field. Methods A systematic search was conducted in eight databases: PubMed, Embase, Web of Science, Cochrane Library databases, the China National Knowledge Infrastructure (CNKI), Wanfang Data, the Chinese Biomedical Literature (CBM) Database, and the VIP Chinese Science and Technology Periodicals Database. The standard mean difference (SMD) values were used to evaluate outcomes, and the pooled proportions and SMD with their 95% confidence intervals (CIs) were also calculated. Results Eighteen randomized controlled trial (RCT) studies were included, with a total of 1,457 children with ASD. This meta-analysis revealed that music therapy improved their language communication [SMD = -1.20; 95%CI -1.45, -0.94; χ2 (17) = 84.17, I2 = 80%, p < 0.001] and social skills [SMD = -1. 13; 95%CI -1.49, -0.78; χ2 (17) = 162.53, I2 = 90%, p < 0.001]. In addition, behavior [SMD = -1.92; 94%CI -2.56, -1.28; χ2 (13) = 235.08, I2 = 95%, p < 0.001], sensory perception [SMD = -1.62; 95%CI -2.17, -1.08; χ2 (16) = 303.80, I2 = 95%, p < 0.001], self-help [SMD = -2. 14; 95%CI -3.17, -1.10; χ2 (6) = 173.07, I2 = 97%, p < 0.001] were all improved. Conclusion Music therapy has a positive effect on the improvement of symptoms in children with ASD. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/.
Collapse
Affiliation(s)
- Zijuan Shi
- North Sichuan Medical College, Nanchong, China
| | - Si Wang
- North Sichuan Medical College, Nanchong, China
| | | | - Aimin Hu
- North Sichuan Medical College, Nanchong, China
| | | | - Yujun Lee
- North Sichuan Medical College, Nanchong, China
- The Graduate School of Xi’an International Studies University, Shaanxi, China
| |
Collapse
|
17
|
Kale G, Addepalli V, Joshi S. A snapshot on introspection of autism spectrum disorder. Mol Biol Rep 2024; 51:610. [PMID: 38704762 DOI: 10.1007/s11033-024-09514-5] [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: 02/09/2024] [Accepted: 04/03/2024] [Indexed: 05/07/2024]
Abstract
Autism spectrum disorder is a neurodevelopmental condition marked by restricted interests and difficulty with social communication. ASD is characterized by heightened neuroinflammation and irregular neuronal connections. ASD is more frequent in male than female with male-female ratio of around 4:1. ASD affects 2.8% or 1 in 36 8-year-olds, based on the CDC's Morbidity and Mortality Weekly Report. Various factors like Environmental, Genetic, Epigenetic and Developmental factors are linked with genesis of ASD. Repetitive behaviors, Impaired communication skills, difficulty with social interaction are some of the clinical features of ASD. Current Pharmacotherapy of ASD limits to management of symptoms only, not cure. The stem cell therapy has a promising potential to be a breakthrough in treating ASD. Various types of stem cells have been successfully tested in children with ASD. AI has a potential to emerge as a tool for early detection of ASD. Robotics can assist the children with ASD to overcome the challenges associated with ASD.
Collapse
Affiliation(s)
- Govind Kale
- Dr. D.Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, 411018, Maharashtra, India
| | - Veeranjaneyulu Addepalli
- Dr. D.Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, 411018, Maharashtra, India.
| | - Sharvari Joshi
- Dr. D.Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, 411018, Maharashtra, India
| |
Collapse
|
18
|
Bove M, Palmieri MA, Santoro M, Agosti LP, Gaetani S, Romano A, Dimonte S, Costantino G, Sikora V, Tucci P, Schiavone S, Morgese MG, Trabace L. Amygdalar neurotransmission alterations in the BTBR mice model of idiopathic autism. Transl Psychiatry 2024; 14:193. [PMID: 38632257 PMCID: PMC11024334 DOI: 10.1038/s41398-024-02905-z] [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: 12/27/2023] [Revised: 04/02/2024] [Accepted: 04/08/2024] [Indexed: 04/19/2024] Open
Abstract
Autism Spectrum Disorders (ASD) are principally diagnosed by three core behavioural symptoms, such as stereotyped repertoire, communication impairments and social dysfunctions. This complex pathology has been linked to abnormalities of corticostriatal and limbic circuits. Despite experimental efforts in elucidating the molecular mechanisms behind these abnormalities, a clear etiopathogenic hypothesis is still lacking. To this aim, preclinical studies can be really helpful to longitudinally study behavioural alterations resembling human symptoms and to investigate the underlying neurobiological correlates. In this regard, the BTBR T+ Itpr3tf/J (BTBR) mice are an inbred mouse strain that exhibits a pattern of behaviours well resembling human ASD-like behavioural features. In this study, the BTBR mice model was used to investigate neurochemical and biomolecular alterations, regarding Nerve Growth Factor (NGF) and Brain-Derived Neurotrophic Factor (BDNF), together with GABAergic, glutamatergic, cholinergic, dopaminergic and noradrenergic neurotransmissions and their metabolites in four different brain areas, i.e. prefrontal cortex, hippocampus, amygdala and hypothalamus. In our results, BTBR strain reported decreased noradrenaline, acetylcholine and GABA levels in prefrontal cortex, while hippocampal measurements showed reduced NGF and BDNF expression levels, together with GABA levels. Concerning hypothalamus, no differences were retrieved. As regarding amygdala, we found reduced dopamine levels, accompanied by increased dopamine metabolites in BTBR mice, together with decreased acetylcholine, NGF and GABA levels and enhanced glutamate content. Taken together, our data showed that the BTBR ASD model, beyond its face validity, is a useful tool to untangle neurotransmission alterations that could be underpinned to the heterogeneous ASD-like behaviours, highlighting the crucial role played by amygdala.
Collapse
Affiliation(s)
- Maria Bove
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, 71122, Foggia, Italy
| | - Maria Adelaide Palmieri
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, 71122, Foggia, Italy
| | - Martina Santoro
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, 00185, Rome, Italy
| | - Lisa Pia Agosti
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, 71122, Foggia, Italy
| | - Silvana Gaetani
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, 00185, Rome, Italy
| | - Adele Romano
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, 00185, Rome, Italy
| | - Stefania Dimonte
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, 71122, Foggia, Italy
| | - Giuseppe Costantino
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, 71122, Foggia, Italy
| | - Vladyslav Sikora
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, 71122, Foggia, Italy
- Department of Pathology, Sumy State University, 40007, Sumy, Ukraine
| | - Paolo Tucci
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, 71122, Foggia, Italy
| | - Stefania Schiavone
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, 71122, Foggia, Italy
| | - Maria Grazia Morgese
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, 71122, Foggia, Italy
| | - Luigia Trabace
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, 71122, Foggia, Italy.
| |
Collapse
|
19
|
Restrepo-Martínez M, Ramirez-Bermudez J, Chacon-Gonzalez J, Ruiz-Garcia R, Malik R, Finger E. Defining repetitive behaviours in frontotemporal dementia. Brain 2024; 147:1149-1165. [PMID: 38134315 DOI: 10.1093/brain/awad431] [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: 09/03/2023] [Revised: 11/08/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
Repetitive behaviours are common manifestations of frontotemporal dementia (FTD). Patients with FTD exhibit various types of repetitive behaviours with unique behavioural and cognitive substrates, including compulsivity, lack of impulse control, stereotypy and hoarding. Other sources of repetitive behaviours, such as restrictive interests and insistence on sameness, may also be seen in FTD. Although repetitive behaviours are highly prevalent and potentially discriminatory in this population, their expression varies widely between patients, and the field lacks consensus about the classification of these behaviours. Terms used to describe repetitive behaviours in FTD are highly heterogeneous and may lack precise definitions. This lack of harmonization of the definitions for distinct forms of repetitive behaviour limits the ability to differentiate between pathological behaviours and impedes understanding of their underlying mechanisms. This review examines established definitions of well-characterized repetitive behaviours in other neuropsychiatric disorders and proposes operational definitions applicable to patients with FTD. Building on extant models of repetitive behaviours in non-human and lesion work and models of social behavioural changes in FTD, we describe the potential neurocognitive bases for the emergence of different types of repetitive behaviours in FTD and their potential perpetuation by a predisposition towards habit formation. Finally, examples of distinct therapeutic approaches for different forms of repetitive behaviours are highlighted, along with future directions to accurately classify, measure and treat these symptoms when they impair quality of life.
Collapse
Affiliation(s)
- Miguel Restrepo-Martínez
- Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada
- Deparment of Cognitive Neurology, Parkwood Institute, London, ON N6C 5J1, Canada
| | - Jesus Ramirez-Bermudez
- Department of Neuropsychiatry, National Institute of Neurology and Neurosurgery Manuel Velasco Suarez, Mexico City, 14269, Mexico
| | - Jacobo Chacon-Gonzalez
- Department of Neuropsychiatry, National Institute of Neurology and Neurosurgery Manuel Velasco Suarez, Mexico City, 14269, Mexico
| | - Ramiro Ruiz-Garcia
- Department of Neuropsychiatry, National Institute of Neurology and Neurosurgery Manuel Velasco Suarez, Mexico City, 14269, Mexico
| | - Rubina Malik
- Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada
- Deparment of Cognitive Neurology, Parkwood Institute, London, ON N6C 5J1, Canada
| | - Elizabeth Finger
- Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada
- Deparment of Cognitive Neurology, Parkwood Institute, London, ON N6C 5J1, Canada
| |
Collapse
|
20
|
Silvestri R, Di Perri MC. Is iron depletion the only cause of motor restlessness in restless sleep presenting in autism spectrum disorder? J Clin Sleep Med 2024; 20:337-338. [PMID: 38174874 PMCID: PMC11019212 DOI: 10.5664/jcsm.11024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 01/03/2024] [Indexed: 01/05/2024]
Affiliation(s)
- Rosalia Silvestri
- Sleep Medicine Center, UOSD of Neurophysiopathology and Movement Disorders, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Maria Caterina Di Perri
- Sleep Medicine Center, UOSD of Neurophysiopathology and Movement Disorders, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| |
Collapse
|
21
|
Shilbayeh SAR, Adeen IS, Alhazmi AS, Aljurayb H, Altokhais RS, Alhowaish N, Aldilaijan KE, Kamal M, Alnakhli AM. The polymorphisms of candidate pharmacokinetic and pharmacodynamic genes and their pharmacogenetic impacts on the effectiveness of risperidone maintenance therapy among Saudi children with autism. Eur J Clin Pharmacol 2024:10.1007/s00228-024-03658-w. [PMID: 38421437 DOI: 10.1007/s00228-024-03658-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: 05/16/2023] [Accepted: 02/22/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND Antipsychotics, including risperidone (RIS), are frequently indicated for various autism spectrum disorder (ASD) manifestations; however, "actionable" PGx testing in psychiatry regarding antipsychotic dosing and selection has limited applications in routine clinical practice because of the lack of standard guidelines, mostly due to the inconsistency and scarcity of genetic variant data. The current study is aimed at examining the association of RIS effectiveness, according to ABC-CV and CGI indexes, with relevant pharmacokinetics (PK) and pharmacodynamics (PD) genes. METHODS Eighty-nine ASD children who received a consistent RIS-based regimen for at least 8 weeks were included. The Axiom PharmacoFocus Array technique was employed to generate accurate star allele-predicted phenotypes of 3 PK genes (CYP3A4, CYP3A5, and CYP2D6). Genotype calls for 5 candidate PD receptor genes (DRD1, DRD2, DRD3, HTR2C, and HTR2A) were obtained and reported as wild type, heterozygous, or homozygous for 11 variants. RESULTS Based on the ABC total score, 42 (47.2%) children were classified as responders, while 47 (52.8%) were classified as nonresponders. Multivariate logistic regression analyses, adjusted for nongenetic factors, suggested nonsignificant impacts of the star allele-predicted phenotypes of all 3 PK genes on improvement in ASD symptoms or CGI scores. However, significant positive or negative associations of certain PD variants involved in dopaminergic and serotonergic pathways were observed with specific ASD core and noncore symptom subdomains. Our significant polymorphism findings, mainly those in DRD2 (rs1800497, rs1799978, and rs2734841), HTR2C (rs3813929), and HTR2A (rs6311), were largely consistent with earlier findings (predictors of RIS effectiveness in adult schizophrenia patients), confirming their validity for identifying ASD children with a greater likelihood of core symptom improvement compared to noncarriers/wild types. Other novel findings of this study, such as significant improvements in DRD3 rs167771 carriers, particularly in ABC total and lethargy/social withdrawal scores, and DRD1 rs1875964 homozygotes and DRD2 rs1079598 wild types in stereotypic behavior, warrant further verification in biochemical and clinical studies to confirm their feasibility for inclusion in a PGx panel. CONCLUSION In conclusion, we provide evidence of potential genetic markers involved in clinical response variability to RIS therapy in ASD children. However, replication in prospective samples with greater ethnic diversity and sample sizes is necessary.
Collapse
Affiliation(s)
- Sireen Abdul Rahim Shilbayeh
- Department of Pharmacy Practice, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia.
| | - Iman Sharaf Adeen
- Department of Pediatric Behavior and Development and Adolescent Medicine, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Ayman Shawqi Alhazmi
- Department of Pediatric Behavior and Development and Adolescent Medicine, King Saud Medical City, Riyadh, Saudi Arabia
| | - Haya Aljurayb
- Molecular Pathology Laboratory, Pathology and Clinical Laboratory Medicine Administration, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Rana Saad Altokhais
- Department of Pediatric Behavior and Development and Adolescent Medicine, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Nourah Alhowaish
- Department of Prevention and Research, King Abdullah International Medical Research Center (KAIMRC), King Abdulaziz Medical City, Ministry of National Guard - Health Affairs, Riyadh, Saudi Arabia
| | - Khawlah Essa Aldilaijan
- Health Sciences Research Center, King Abdullah Bin Abdulaziz University Hospital, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Mostafa Kamal
- Department of Life Science Application Support, Gulf Scientific Corporation, Riyadh, Saudi Arabia
| | - Anwar Mansour Alnakhli
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| |
Collapse
|
22
|
Al-Radadi NS, Al-Bishri WM, Salem NA, ElShebiney SA. Plant-mediated green synthesis of gold nanoparticles using an aqueous extract of Passiflora ligularis, optimization, characterizations, and their neuroprotective effect on propionic acid-induced autism in Wistar rats. Saudi Pharm J 2024; 32:101921. [PMID: 38283153 PMCID: PMC10820356 DOI: 10.1016/j.jsps.2023.101921] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 12/12/2023] [Indexed: 01/30/2024] Open
Abstract
The current study was conducted to examine an innovative method for synthesizing gold nanoparticles (AuNPs) from an aqueous sweet granadilla (Passiflora ligularis Juss) P. ligularis. Furthermore, the synthesized AuNPs were used to explore their potential neuroprotective impact against propionic acid (PPA)-induced autism. A sweet granadilla extract was used to achieve the synthesis of AuNPs. The structural and dimensional dispersion of AuNPs were confirmed by different techniques, including UV-Vis spectrophotometer (UV-Vis), X-ray Diffraction (XRD) Pattern, Energy Dispersive X-ray (EDX), Zeta potential, and High-Resolution Transmission Electron Microscopy (HRTEM) analysis. The AuNPs mediated by P. ligularis adopt a spherical shape morphology and the particle size was distributed in the range of 8.43-13 nm without aggregation. Moreover, in vivo, the anti-autistic effects of AuNPs administration were higher than those of P. ligularis extract per second. In addition, the reduced anxiety and neurobehavioral deficits of AuNPs were observed in autistic rats which halted the brain oxidative stress, reduced inflammatory cytokines, ameliorated neurotransmitters, and neurochemical release, and suppressed apoptotic genes (p < 0.05). The alleviated antiapoptotic gene expression and histopathological analysis confirmed that the treatment of AuNPs showed significant neural pathways that aid in reducing tissue damage and necrosis. The results emphasize that the biomedical activity was increased by using the green source synthesis P. ligularis -AuNPs. Additionally, the formulation of AuNPs demonstrates strong neuroprotective effects against PPA-induced autism that were arbitrated by a range of different mechanisms, such as anti-inflammatory, antioxidant, neuromodulator, and antiapoptotic effects.
Collapse
Affiliation(s)
- Najlaa S. Al-Radadi
- Department of Chemistry, Faculty of Science, Taibah University, P.O. Box 30002, Al-Madinah Al-Munawarah 14177, Saudi Arabia
| | - Widad M. Al-Bishri
- Department of Biochemistry, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Neveen A. Salem
- Department of Biochemistry, College of Science, University of Jeddah, Jeddah, Saudi Arabia
- Department of Narcotics, Ergogenic Aids and Poisons, Medical Research and Clinical Studies Institute, National Research Centre, Cairo, Egypt
| | - Shaimaa A. ElShebiney
- Department of Narcotics, Ergogenic Aids and Poisons, Medical Research and Clinical Studies Institute, National Research Centre, Cairo, Egypt
| |
Collapse
|
23
|
Socha J, Grochecki P, Smaga I, Jastrzębska J, Wronikowska-Denysiuk O, Marszalek-Grabska M, Slowik T, Kotlinski R, Filip M, Lubec G, Kotlinska JH. Social Interaction in Adolescent Rats with Neonatal Ethanol Exposure: Impact of Sex and CE-123, a Selective Dopamine Reuptake Inhibitor. Int J Mol Sci 2024; 25:1041. [PMID: 38256113 PMCID: PMC10816180 DOI: 10.3390/ijms25021041] [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: 12/20/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
Children with fetal alcohol spectrum disorders (FASDs) demonstrate deficits in social functioning that contribute to early withdrawal from school and delinquency, as well as the development of anxiety and depression. Dopamine is involved in reward, motivation, and social behavior. Thus, we evaluated whether neonatal ethanol exposure (in an animal model of FASDs) has an impact on social recognition memory using the three-chamber social novelty discrimination test during early and middle adolescence in male and female rats, and whether the modafinil analog, the novel atypical dopamine reuptake inhibitor CE-123, can modify this effect. Our study shows that male and female rats neonatally exposed to ethanol exhibited sex- and age-dependent deficits in social novelty discrimination in early (male) and middle (female) adolescence. These deficits were specific to the social domain and not simply due to more general deficits in learning and memory because these animals did not exhibit changes in short-term recognition memory in the novel object recognition task. Furthermore, early-adolescent male rats that were neonatally exposed to ethanol did not show changes in the anxiety index but demonstrated an increase in locomotor activity. Chronic treatment with CE-123, however, prevented the appearance of these social deficits. In the hippocampus of adolescent rats, CE-123 increased BDNF and decreased its signal transduction TrkB receptor expression level in ethanol-exposed animals during development, suggesting an increase in neuroplasticity. Thus, selective dopamine reuptake inhibitors, such as CE-123, represent interesting drug candidates for the treatment of deficits in social behavior in adolescent individuals with FASDs.
Collapse
Affiliation(s)
- Justyna Socha
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodzki 4a, 20-093 Lublin, Poland; (J.S.); (P.G.)
| | - Pawel Grochecki
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodzki 4a, 20-093 Lublin, Poland; (J.S.); (P.G.)
| | - Irena Smaga
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Smetna 12, 31-343 Krakow, Poland; (I.S.); (J.J.); (M.F.)
| | - Joanna Jastrzębska
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Smetna 12, 31-343 Krakow, Poland; (I.S.); (J.J.); (M.F.)
| | - Olga Wronikowska-Denysiuk
- Independent Laboratory of Behavioral Studies, Chair of Biomedical Sciences, Medical University of Lublin, Chodzki 4a, 20-093 Lublin, Poland;
| | - Marta Marszalek-Grabska
- Department of Experimental and Clinical Pharmacology, Medical University, Jaczewskiego 8b, 20-090 Lublin, Poland;
| | - Tymoteusz Slowik
- Experimental Medicine Center, Medical University, Jaczewskiego 8, 20-090 Lublin, Poland;
| | - Robert Kotlinski
- Clinical Department of Cardiac Surgery, University of Rzeszow, 35-601 Rzeszow, Poland;
| | - Małgorzata Filip
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Smetna 12, 31-343 Krakow, Poland; (I.S.); (J.J.); (M.F.)
| | - Gert Lubec
- Department of Neuroproteomics, Paracelsus Medical University, 5020 Salzburg, Austria;
| | - Jolanta H. Kotlinska
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodzki 4a, 20-093 Lublin, Poland; (J.S.); (P.G.)
| |
Collapse
|
24
|
Um KB, Kwak S, Cheon SH, Kim J, Hwang SK. AST-001 Improves Social Deficits and Restores Dopamine Neuron Activity in a Mouse Model of Autism. Biomedicines 2023; 11:3283. [PMID: 38137504 PMCID: PMC10741043 DOI: 10.3390/biomedicines11123283] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/05/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder characterized by impaired social communication and social interaction, restricted and repetitive behavior, and interests. The core symptoms of ASD are associated with deficits in mesocorticolimbic dopamine pathways that project from the ventral tegmental area (VTA) to the nucleus accumbens (NAc) and medial prefrontal cortex (mPFC). AST-001 is an investigational product currently in a phase 3 clinical trial for treating the core symptoms of ASD, with L-serine as the API (active pharmaceutical ingredient). Because the causes of ASD are extremely heterogeneous, a single genetic ASD model cannot represent all autism models. In this paper, we used the VPA-exposed model, which is more general and widely used than a single genetic model, but this is also one of the animal models of autism. Herein, we conducted experiments to demonstrate the efficacy of AST-001 as L-Serine that alters the regulation of the firing rate in dopamine neurons by inhibiting small conductance Ca2+-activated K+ channels (SK channels). Through these actions, AST-001 improved sociability and social novelty by rescuing the intrinsic excitabilities of dopamine neurons in VPA-exposed ASD mouse models that showed ASD-related behavioral abnormalities. It is thought that this effect of improving social deficits in VPA-exposed ASD mouse models is due to AST-001 normalizing aberrant SK channel activities that slowed VTA dopamine neuron firing. Overall, these findings suggest that AST-001 may be a potential therapeutic agent for ASD patients, and that its mechanism of action may involve the regulation of dopamine neuron activity and the improvement of social interaction.
Collapse
Affiliation(s)
- Ki Bum Um
- Astrogen Inc., 440, Hyeoksin-daero, Dong-gu, Daegu 41072, Republic of Korea; (K.B.U.); (S.K.)
| | - Soyoung Kwak
- Astrogen Inc., 440, Hyeoksin-daero, Dong-gu, Daegu 41072, Republic of Korea; (K.B.U.); (S.K.)
| | - Sun-Ha Cheon
- Astrogen Inc., 440, Hyeoksin-daero, Dong-gu, Daegu 41072, Republic of Korea; (K.B.U.); (S.K.)
| | - JuHyun Kim
- Astrogen Inc., 440, Hyeoksin-daero, Dong-gu, Daegu 41072, Republic of Korea; (K.B.U.); (S.K.)
| | - Su-Kyeong Hwang
- Astrogen Inc., 440, Hyeoksin-daero, Dong-gu, Daegu 41072, Republic of Korea; (K.B.U.); (S.K.)
- Department of Pediatrics, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| |
Collapse
|
25
|
Mabry S, Wilson EN, Bradshaw JL, Gardner JJ, Fadeyibi O, Vera E, Osikoya O, Cushen SC, Karamichos D, Goulopoulou S, Cunningham RL. Sex and age differences in social and cognitive function in offspring exposed to late gestational hypoxia. Biol Sex Differ 2023; 14:81. [PMID: 37951901 PMCID: PMC10640736 DOI: 10.1186/s13293-023-00557-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 10/09/2023] [Indexed: 11/14/2023] Open
Abstract
BACKGROUND Gestational sleep apnea is a hypoxic sleep disorder that affects 8-26% of pregnancies and increases the risk for central nervous system dysfunction in offspring. Specifically, there are sex differences in the sensitivity of the fetal hippocampus to hypoxic insults, and hippocampal impairments are associated with social dysfunction, repetitive behaviors, anxiety, and cognitive impairment. Yet, it is unclear whether gestational sleep apnea impacts these hippocampal-associated functions and if sex and age modify these effects. To examine the relationship between gestational sleep apnea and hippocampal-associated behaviors, we used chronic intermittent hypoxia (CIH) to model late gestational sleep apnea in pregnant rats. We hypothesized that late gestational CIH would produce sex- and age-specific social, anxiety-like, repetitive, and cognitive impairments in offspring. METHODS Timed pregnant Long-Evans rats were exposed to CIH or room air normoxia from GD 15-19. Behavioral testing of offspring occurred during either puberty or young adulthood. To examine gestational hypoxia-induced behavioral phenotypes, we quantified hippocampal-associated behaviors (social function, repetitive behaviors, anxiety-like behaviors, and spatial memory and learning), hippocampal neuronal activity (glutamatergic NMDA receptors, dopamine transporter, monoamine oxidase-A, early growth response protein 1, and doublecortin), and circulating hormones in offspring. RESULTS Late gestational CIH induced sex- and age-specific differences in social, repetitive, and memory functions in offspring. In female pubertal offspring, CIH impaired social function, increased repetitive behaviors, and elevated circulating corticosterone levels but did not impact memory. In contrast, CIH transiently induced spatial memory dysfunction in pubertal male offspring but did not impact social or repetitive functions. Long-term effects of gestational CIH on social behaviors were only observed in female offspring, wherein CIH induced social disengagement and suppression of circulating corticosterone levels in young adulthood. No effects of gestational CIH were observed in anxiety-like behaviors, hippocampal neuronal activity, or circulating testosterone and estradiol levels, regardless of sex or age of offspring. CONCLUSIONS Our results indicate that hypoxia-associated pregnancy complications during late gestation can increase the risk for behavioral and physiological outcomes in offspring, such as social dysfunction, repetitive behaviors, and cognitive impairment, that are dependent on sex and age.
Collapse
Affiliation(s)
- Steve Mabry
- Department of Pharmaceutical Sciences, School of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107, USA
| | - E Nicole Wilson
- Department of Pharmaceutical Sciences, School of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107, USA
| | - Jessica L Bradshaw
- Department of Pharmaceutical Sciences, School of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107, USA
| | - Jennifer J Gardner
- Department of Pharmaceutical Sciences, School of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107, USA
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Oluwadarasimi Fadeyibi
- Department of Pharmaceutical Sciences, School of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107, USA
| | - Edward Vera
- Department of Pharmaceutical Sciences, School of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107, USA
- Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Oluwatobiloba Osikoya
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Spencer C Cushen
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
- Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Dimitrios Karamichos
- Department of Pharmaceutical Sciences, School of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107, USA
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3430 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
- Department of Pharmacology and Neuroscience, University of North Texas Health Science, Fort Worth, TX, 76107, USA
| | - Styliani Goulopoulou
- Departments of Basic Sciences, Gynecology and Obstetrics, Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Rebecca L Cunningham
- Department of Pharmaceutical Sciences, School of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107, USA.
| |
Collapse
|
26
|
Fernandes L, Kleene R, Congiu L, Freitag S, Kneussel M, Loers G, Schachner M. CHL1 depletion affects dopamine receptor D2-dependent modulation of mouse behavior. Front Behav Neurosci 2023; 17:1288509. [PMID: 38025382 PMCID: PMC10665519 DOI: 10.3389/fnbeh.2023.1288509] [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: 09/04/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction The dopaminergic system plays a key role in the appropriate functioning of the central nervous system, where it is essential for emotional balance, arousal, reward, and motor control. The cell adhesion molecule close homolog of L1 (CHL1) contributes to dopaminergic system development, and CHL1 and the dopamine receptor D2 (D2R) are associated with mental disorders like schizophrenia, addiction, autism spectrum disorder and depression. Methods Here, we investigated how the interplay between CHL1 and D2R affects the behavior of young adult male and female wild-type (CHL+/+) and CHL1-deficient (CHL1-/-) mice, when D2R agonist quinpirole and antagonist sulpiride are applied. Results Low doses of quinpirole (0.02 mg/kg body weight) induced hypolocomotion of CHL1+/+ and CHL1-/- males and females, but led to a delayed response in CHL1-/- mice. Sulpiride (1 mg/kg body weight) affected locomotion of CHL1-/- females and social interaction of CHL1+/+ females as well as social interactions of CHL1-/- and CHL1+/+ males. Quinpirole increased novelty-seeking behavior of CHL1-/- males compared to CHL1+/+ males. Vehicle-treated CHL1-/- males and females showed enhanced working memory and reduced stress-related behavior. Discussion We propose that CHL1 regulates D2R-dependent functions in vivo. Deficiency of CHL1 leads to abnormal locomotor activity and emotionality, and to sex-dependent behavioral differences.
Collapse
Affiliation(s)
- Luciana Fernandes
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Ralf Kleene
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Ludovica Congiu
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Sandra Freitag
- Institut für Molekulare Neurogenetik, Zentrum für Molekulare Neurobiologie Hamburg, ZMNH, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Matthias Kneussel
- Institut für Molekulare Neurogenetik, Zentrum für Molekulare Neurobiologie Hamburg, ZMNH, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Gabriele Loers
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Melitta Schachner
- Department of Cell Biology and Neuroscience, Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, NJ, United States
| |
Collapse
|
27
|
Vilela J, Martiniano H, Marques AR, Santos JX, Asif M, Rasga C, Oliveira G, Vicente AM. Identification of Neurotransmission and Synaptic Biological Processes Disrupted in Autism Spectrum Disorder Using Interaction Networks and Community Detection Analysis. Biomedicines 2023; 11:2971. [PMID: 38001974 PMCID: PMC10668950 DOI: 10.3390/biomedicines11112971] [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: 10/03/2023] [Revised: 10/26/2023] [Accepted: 11/01/2023] [Indexed: 11/26/2023] Open
Abstract
Autism Spectrum Disorder (ASD) is a neurodevelopmental disorder characterized by communication deficits and repetitive behavioral patterns. Hundreds of candidate genes have been implicated in ASD, including neurotransmission and synaptic (NS) genes; however, the genetic architecture of this disease is far from clear. In this study, we seek to clarify the biological processes affected by NS gene variants identified in individuals with ASD and the global networks that link those processes together. For a curated list of 1216 NS candidate genes, identified in multiple databases and the literature, we searched for ultra-rare (UR) loss-of-function (LoF) variants in the whole-exome sequencing dataset from the Autism Sequencing Consortium (N = 3938 cases). Filtering for population frequency was carried out using gnomAD (N = 60,146 controls). NS genes with UR LoF variants were used to construct a network of protein-protein interactions, and the network's biological communities were identified by applying the Leiden algorithm. We further explored the expression enrichment of network genes in specific brain regions. We identified 356 variants in 208 genes, with a preponderance of UR LoF variants in the PDE11A and SYTL3 genes. Expression enrichment analysis highlighted several subcortical structures, particularly the basal ganglia. The interaction network defined seven network communities, clustering synaptic and neurotransmitter pathways with several ubiquitous processes that occur in multiple organs and systems. This approach also uncovered biological pathways that are not usually associated with ASD, such as brain cytochromes P450 and brain mitochondrial metabolism. Overall, the community analysis suggests that ASD involves the disruption of synaptic and neurotransmitter pathways but also ubiquitous, but less frequently implicated, biological processes.
Collapse
Affiliation(s)
- Joana Vilela
- Departamento de Promoção da Saúde e Doenças Não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, Avenida Padre Cruz, 1649-016 Lisboa, Portugal; (J.V.); (H.M.); (A.R.M.); (J.X.S.); (M.A.); (C.R.)
- BioISI-Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Campo Grande, C8, 1749-016 Lisboa, Portugal
| | - Hugo Martiniano
- Departamento de Promoção da Saúde e Doenças Não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, Avenida Padre Cruz, 1649-016 Lisboa, Portugal; (J.V.); (H.M.); (A.R.M.); (J.X.S.); (M.A.); (C.R.)
- BioISI-Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Campo Grande, C8, 1749-016 Lisboa, Portugal
| | - Ana Rita Marques
- Departamento de Promoção da Saúde e Doenças Não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, Avenida Padre Cruz, 1649-016 Lisboa, Portugal; (J.V.); (H.M.); (A.R.M.); (J.X.S.); (M.A.); (C.R.)
- BioISI-Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Campo Grande, C8, 1749-016 Lisboa, Portugal
| | - João Xavier Santos
- Departamento de Promoção da Saúde e Doenças Não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, Avenida Padre Cruz, 1649-016 Lisboa, Portugal; (J.V.); (H.M.); (A.R.M.); (J.X.S.); (M.A.); (C.R.)
- BioISI-Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Campo Grande, C8, 1749-016 Lisboa, Portugal
| | - Muhammad Asif
- Departamento de Promoção da Saúde e Doenças Não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, Avenida Padre Cruz, 1649-016 Lisboa, Portugal; (J.V.); (H.M.); (A.R.M.); (J.X.S.); (M.A.); (C.R.)
- BioISI-Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Campo Grande, C8, 1749-016 Lisboa, Portugal
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad 38000, Pakistan
| | - Célia Rasga
- Departamento de Promoção da Saúde e Doenças Não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, Avenida Padre Cruz, 1649-016 Lisboa, Portugal; (J.V.); (H.M.); (A.R.M.); (J.X.S.); (M.A.); (C.R.)
- BioISI-Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Campo Grande, C8, 1749-016 Lisboa, Portugal
| | - Guiomar Oliveira
- Unidade de Neurodesenvolvimento e Autismo, Serviço do Centro de Desenvolvimento da Criança, Centro de Investigação e Formação Clínica, Hospital Pediátrico, Centro Hospitalar e Universitário de Coimbra (CHUC), 3000-602 Coimbra, Portugal;
- Coimbra Institute for Biomedical Imaging and Translational Research, University Clinic of Pediatrics, Faculty of Medicine, University of Coimbra, 3000-602 Coimbra, Portugal
| | - Astrid Moura Vicente
- Departamento de Promoção da Saúde e Doenças Não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, Avenida Padre Cruz, 1649-016 Lisboa, Portugal; (J.V.); (H.M.); (A.R.M.); (J.X.S.); (M.A.); (C.R.)
- BioISI-Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Campo Grande, C8, 1749-016 Lisboa, Portugal
| |
Collapse
|
28
|
Bodin R, Seewooruttun C, Corona A, Delanaud S, Pelletier A, Villégier AS. Sex-dependent impact of perinatal 5G electromagnetic field exposure in the adolescent rat behavior. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:113704-113717. [PMID: 37851267 DOI: 10.1007/s11356-023-30256-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/29/2023] [Indexed: 10/19/2023]
Abstract
The fifth generation (5G) network is currently being worldwide spread out, raising questions about the potential impact of this new technology, particularly on immature organisms. The current study aimed to investigate the effects of daily 5G electromagnetic field (EMF) perinatal exposure on the neurodevelopment of rats. The exposure level was set to the limit of whole-body public exposure defined by the International Commission on Non-Ionizing Radiation Protection. The mother rat specific absorption rate (SAR) was 0.07 W/kg for 22 h/day at 3500 MHz continuous wave from gestational day (GD) 8 to post-natal day (PND) 21. Clinical observations were performed on weight, length, sex ratio, number of pups per litter, and number of stillborn in sham and EMF-exposed groups (n = 7). The age of pinna ear detachment, incisor eruption, and eye opening were recorded. Behavior was assessed on righting, gripping, and negative geotaxis reflexes at PND 3 or 7 and on stereotyped and horizontal movements in the open field at PND 43. Our results indicated that both male and female pups showed delayed incisor eruption in the EMF-exposed group compared to the sham group (+ 1 day). Regarding activity in the open field, adolescent females showed less stereotyped movements (- 70%), while adolescent males showed more stereotyped movements (+ 50%) compared to the sham-exposed adolescent rats. Thus, the present study suggested that perinatal exposure to 5G at SAR level below reglementary threshold led to perturbations in the descendants seen in juveniles and adolescents.
Collapse
Affiliation(s)
- Raphaël Bodin
- PERITOX Laboratory (UMR_I 01), INERIS, MIV/TEAM, Verneuil-en-Halatte, France
| | | | - Aurélie Corona
- University of Picardie Jules Verne, CURS, Amiens, France
| | | | | | | |
Collapse
|
29
|
de Miguel L, Ballester P, Egoavil C, Sánchez-Ocaña ML, García-Muñoz AM, Cerdá B, Zafrilla P, Ramos E, Peiró AM. Pharmacogenetics May Prevent Psychotropic Adverse Events in Autism Spectrum Disorder: An Observational Pilot Study. Pharmaceuticals (Basel) 2023; 16:1496. [PMID: 37895967 PMCID: PMC10610471 DOI: 10.3390/ph16101496] [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: 08/25/2023] [Revised: 10/15/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
INTRODUCTION Up to 73% of individuals with autism spectrum disorder (ASD) and intellectual disability (ID) currently have prescriptions for psychotropic drugs. This is explained by a higher prevalence of medical and psychiatric chronic comorbidities, which favors polypharmacy, increasing the probability of the appearance of adverse events (AEs). These could be a preventable cause of harm to patients with ASD and an unnecessary waste of healthcare resources. OBJECTIVE To study the impact of pharmacogenetic markers on the prevention of AE appearance in a population with ASD and ID. METHODS This is a cross-sectional, observational study (n = 118, 72 participants completed all information) in the ASD population. Sociodemographic and pharmacological data were gathered. The Udvalg for Kliniske Undersøgelser Scale (UKU Scale) was used to identify AEs related to the use of psychotropic medication. Polymorphisms of DOP2, ABCB1, and COMT were genotyped and correlated with the AE to find candidate genes. Furthermore, a review of all medications assessed in a clinical trial for adults with autism was performed to enrich the search for potential pharmacogenetic markers, keeping in mind the usual medications. RESULTS The majority of the study population were men (75%) with multiple comorbidities and polypharmacy, the most frequently prescribed drugs were antipsychotics (69%); 21% of the participants had four or more AEs related to psychotropic drugs. The most common were "Neurological" and" Psychiatric" (both 41%). Statistical analysis results suggested a significant correlation between the neurological symptoms and the DOP2 genotype, given that they are not equally distributed among its allelic variants. The final review considered 19 manuscripts of medications for adults with ASD, and the confirmed genetic markers for those medications were consulted in databases. CONCLUSION A possible correlation between neurologic AEs and polymorphisms of DOP2 was observed; therefore, studying this gene could contribute to the safety of this population's prescriptions. The following studies are underway to maximize statistical power and have a better representation of the population.
Collapse
Affiliation(s)
- Laura de Miguel
- Pharmacogenetic Unit, Clinical Pharmacology Department, Alicante Institute for Health and Biomedical Research (ISABIAL), General University Hospital of Alicante, c/Pintor Baeza, 12, 03010 Alicante, Spain
- Clinical Pharmacology, Toxicology and Chemical Safety Unit, Institute of Bioengineering, Miguel Hernández University, Avda. de la Universidad s/n, 03202 Elche, Spain
| | - Pura Ballester
- Faculty of Pharmacy and Nutrition, Campus de los Jerónimos, Universidad Católica San Antonio de Murcia (UCAM), Guadalupe, 30107 Murcia, Spain
| | - Cecilia Egoavil
- Pharmacogenetic Unit, Clinical Pharmacology Department, Alicante Institute for Health and Biomedical Research (ISABIAL), General University Hospital of Alicante, c/Pintor Baeza, 12, 03010 Alicante, Spain
- Clinical Pharmacology Unit, Dr. Balmis General University Hospital, 03010 Alicante, Spain
| | - María Luisa Sánchez-Ocaña
- Faculty of Pharmacy and Nutrition, Campus de los Jerónimos, Universidad Católica San Antonio de Murcia (UCAM), Guadalupe, 30107 Murcia, Spain
| | - Ana María García-Muñoz
- Faculty of Pharmacy and Nutrition, Campus de los Jerónimos, Universidad Católica San Antonio de Murcia (UCAM), Guadalupe, 30107 Murcia, Spain
| | - Begoña Cerdá
- Faculty of Pharmacy and Nutrition, Campus de los Jerónimos, Universidad Católica San Antonio de Murcia (UCAM), Guadalupe, 30107 Murcia, Spain
| | - Pilar Zafrilla
- Faculty of Pharmacy and Nutrition, Campus de los Jerónimos, Universidad Católica San Antonio de Murcia (UCAM), Guadalupe, 30107 Murcia, Spain
| | - Enrique Ramos
- Clinical Pharmacology, Toxicology and Chemical Safety Unit, Institute of Bioengineering, Miguel Hernández University, Avda. de la Universidad s/n, 03202 Elche, Spain
| | - Ana M. Peiró
- Pharmacogenetic Unit, Clinical Pharmacology Department, Alicante Institute for Health and Biomedical Research (ISABIAL), General University Hospital of Alicante, c/Pintor Baeza, 12, 03010 Alicante, Spain
- Clinical Pharmacology, Toxicology and Chemical Safety Unit, Institute of Bioengineering, Miguel Hernández University, Avda. de la Universidad s/n, 03202 Elche, Spain
- Clinical Pharmacology Unit, Dr. Balmis General University Hospital, 03010 Alicante, Spain
| |
Collapse
|
30
|
Dorsey SG, Mocci E, Lane MV, Krueger BK. Rapid effects of valproic acid on the fetal brain transcriptome: Implications for brain development and autism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.01.538959. [PMID: 37205520 PMCID: PMC10187231 DOI: 10.1101/2023.05.01.538959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
There is an increased incidence of autism among the children of women who take the anti-epileptic, mood stabilizing drug, valproic acid (VPA) during pregnancy; moreover, exposure to VPA in utero causes autistic-like symptoms in rodents and non-human primates. Analysis of RNAseq data ob-tained from E12.5 fetal mouse brains 3 hours after VPA administration revealed that VPA significant-ly increased or decreased the expression of approximately 7,300 genes. No significant sex differ-ences in VPA-induced gene expression were observed. Expression of genes associated with neu-rodevelopmental disorders (NDDs) such as autism as well as neurogenesis, axon growth and syn-aptogenesis, GABAergic, glutaminergic and dopaminergic synaptic transmission, perineuronal nets, and circadian rhythms was dysregulated by VPA. Moreover, expression of 399 autism risk genes was significantly altered by VPA as was expression of 252 genes that have been reported to play fundamental roles in the development of the nervous system but are not otherwise linked to autism. The goal of this study was to identify mouse genes that are: (a) significantly up- or down-regulated by VPA in the fetal brain and (b) known to be associated with autism and/or to play a role in embryonic neurodevelopmental processes, perturbation of which has the potential to alter brain connectivity in the postnatal and adult brain. The set of genes meeting these criteria pro-vides potential targets for future hypothesis-driven approaches to elucidating the proximal underly-ing causes of defective brain connectivity in NDDs such as autism.
Collapse
|
31
|
Mabry S, Wilson EN, Bradshaw JL, Gardner JJ, Fadeyibi O, Vera E, Osikoya O, Cushen SC, Karamichos D, Goulopoulou S, Cunningham RL. Sex and age differences in social and cognitive function in offspring exposed to late gestational hypoxia. RESEARCH SQUARE 2023:rs.3.rs-2507737. [PMID: 37333114 PMCID: PMC10275064 DOI: 10.21203/rs.3.rs-2507737/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Background Gestational sleep apnea affects 8-26% of pregnancies and can increase the risk for autism spectrum disorder (ASD) in offspring. ASD is a neurodevelopmental disorder associated with social dysfunction, repetitive behaviors, anxiety, and cognitive impairment. To examine the relationship between gestational sleep apnea and ASD-associated behaviors, we used a chronic intermittent hypoxia (CIH) protocol between gestational days (GD) 15-19 in pregnant rats to model late gestational sleep apnea. We hypothesized that late gestational CIH would produce sex- and age-specific social, mood, and cognitive impairments in offspring. Methods Timed pregnant Long-Evans rats were exposed to CIH or room air normoxia from GD 15-19. Behavioral testing of offspring occurred during either puberty or young adulthood. To examine ASD-associated phenotypes, we quantified ASD-associated behaviors (social function, repetitive behaviors, anxiety-like behaviors, and spatial memory and learning), hippocampal activity (glutamatergic NMDA receptors, dopamine transporter, monoamine oxidase-A, EGR-1, and doublecortin), and circulating hormones in offspring. Results Late gestational CIH induced sex- and age-specific differences in social, repetitive and memory functions in offspring. These effects were mostly transient and present during puberty. In female pubertal offspring, CIH impaired social function, increased repetitive behaviors, and increased circulating corticosterone levels, but did not impact memory. In contrast, CIH transiently induced spatial memory dysfunction in pubertal male offspring but did not impact social or repetitive functions. Long-term effects of gestational CIH were only observed in female offspring, wherein CIH induced social disengagement and suppression of circulating corticosterone levels in young adulthood. No effects of gestational CIH were observed on anxiety-like behaviors, hippocampal activity, circulating testosterone levels, or circulating estradiol levels, regardless of sex or age of offspring. Conclusions Our results indicate that hypoxia-associated pregnancy complications during late gestation can increase the risk for ASD-associated behavioral and physiological outcomes, such as pubertal social dysfunction, corticosterone dysregulation, and memory impairments.
Collapse
Affiliation(s)
- Steve Mabry
- UNTHSC: University of North Texas Health Science Center
| | | | | | | | | | - Edward Vera
- UNTHSC: University of North Texas Health Science Center
| | | | | | | | | | | |
Collapse
|
32
|
Nakamura Y, Ishida T, Tanaka SC, Mitsuyama Y, Yokoyama S, Shinzato H, Itai E, Okada G, Kobayashi Y, Kawashima T, Miyata J, Yoshihara Y, Takahashi H, Aoki R, Nakamura M, Ota H, Itahashi T, Morita S, Kawakami S, Abe O, Okada N, Kunimatsu A, Yamashita A, Yamashita O, Imamizu H, Morimoto J, Okamoto Y, Murai T, Hashimoto R, Kasai K, Kawato M, Koike S. Distinctive alterations in the mesocorticolimbic circuits in various psychiatric disorders. Psychiatry Clin Neurosci 2023; 77:345-354. [PMID: 36905180 PMCID: PMC11488596 DOI: 10.1111/pcn.13542] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 03/03/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023]
Abstract
AIM Increasing evidence suggests that psychiatric disorders are linked to alterations in the mesocorticolimbic dopamine-related circuits. However, the common and disease-specific alterations remain to be examined in schizophrenia (SCZ), major depressive disorder (MDD), and autism spectrum disorder (ASD). Thus, this study aimed to examine common and disease-specific features related to mesocorticolimbic circuits. METHODS This study included 555 participants from four institutes with five scanners: 140 individuals with SCZ (45.0% female), 127 individuals with MDD (44.9%), 119 individuals with ASD (15.1%), and 169 healthy controls (HC) (34.9%). All participants underwent resting-state functional magnetic resonance imaging. A parametric empirical Bayes approach was adopted to compare estimated effective connectivity among groups. Intrinsic effective connectivity focusing on the mesocorticolimbic dopamine-related circuits including the ventral tegmental area (VTA), shell and core parts of the nucleus accumbens (NAc), and medial prefrontal cortex (mPFC) were examined using a dynamic causal modeling analysis across these psychiatric disorders. RESULTS The excitatory shell-to-core connectivity was greater in all patients than in the HC group. The inhibitory shell-to-VTA and shell-to-mPFC connectivities were greater in the ASD group than in the HC, MDD, and SCZ groups. Furthermore, the VTA-to-core and VTA-to-shell connectivities were excitatory in the ASD group, while those connections were inhibitory in the HC, MDD, and SCZ groups. CONCLUSION Impaired signaling in the mesocorticolimbic dopamine-related circuits could be an underlying neuropathogenesis of various psychiatric disorders. These findings will improve the understanding of unique neural alternations of each disorder and will facilitate identification of effective therapeutic targets.
Collapse
Affiliation(s)
- Yuko Nakamura
- Center for Evolutionary Cognitive Sciences, Graduate School of Art and SciencesUniversity of TokyoTokyoJapan
- University of Tokyo Institute for Diversity & Adaptation of Human Mind (UTIDAHM)TokyoJapan
| | - Takuya Ishida
- Center for Evolutionary Cognitive Sciences, Graduate School of Art and SciencesUniversity of TokyoTokyoJapan
- Department of NeuropsychiatryGraduate School of Wakayama Medical UniversityWakayamaJapan
| | - Saori C. Tanaka
- Brain Information Communication Research Laboratory GroupAdvanced Telecommunications Research Institutes International (ATR)KyotoJapan
- Information Science, Graduate School of Science and TechnologyNara Institute of Science and TechnologyNaraJapan
| | - Yuki Mitsuyama
- Department of Psychiatry and NeurosciencesHiroshima UniversityHiroshimaJapan
| | - Satoshi Yokoyama
- Department of Psychiatry and NeurosciencesHiroshima UniversityHiroshimaJapan
| | - Hotaka Shinzato
- Department of Psychiatry and NeurosciencesHiroshima UniversityHiroshimaJapan
| | - Eri Itai
- Department of Psychiatry and NeurosciencesHiroshima UniversityHiroshimaJapan
| | - Go Okada
- Department of Psychiatry and NeurosciencesHiroshima UniversityHiroshimaJapan
| | - Yuko Kobayashi
- Department of Psychiatry, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Takahiko Kawashima
- Department of Psychiatry, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Jun Miyata
- Department of Psychiatry, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Yujiro Yoshihara
- Department of Psychiatry, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Hidehiko Takahashi
- Department of Psychiatry and Behavioral SciencesTokyo Medical and Dental UniversityTokyoJapan
| | - Ryuta Aoki
- Medical Institute of Developmental Disabilities ResearchShowa UniversityTokyoJapan
| | - Motoaki Nakamura
- Medical Institute of Developmental Disabilities ResearchShowa UniversityTokyoJapan
| | - Haruhisa Ota
- Medical Institute of Developmental Disabilities ResearchShowa UniversityTokyoJapan
| | - Takashi Itahashi
- Medical Institute of Developmental Disabilities ResearchShowa UniversityTokyoJapan
| | - Susumu Morita
- Department of Neuropsychiatry, Graduate School of MedicineUniversity of TokyoTokyoJapan
| | - Shintaro Kawakami
- Department of Neuropsychiatry, Graduate School of MedicineUniversity of TokyoTokyoJapan
| | - Osamu Abe
- Department of Radiology, Graduate School of Medicinethe University of TokyoTokyoJapan
| | - Naohiro Okada
- The International Research Center for Neurointelligence (WPI‐IRCN), Institutes for Advanced Study (UTIAS)University of TokyoTokyoJapan
| | - Akira Kunimatsu
- Department of RadiologyInternational University of Health and Welfare Mita HospitalTokyoJapan
| | - Ayumu Yamashita
- Brain Information Communication Research Laboratory GroupAdvanced Telecommunications Research Institutes International (ATR)KyotoJapan
- Department of PsychiatryBoston University School of MedicineBostonMassachusettsUSA
| | - Okito Yamashita
- Brain Information Communication Research Laboratory GroupAdvanced Telecommunications Research Institutes International (ATR)KyotoJapan
- Center for Advanced Intelligence ProjectRIKENTokyoJapan
| | - Hiroshi Imamizu
- Brain Information Communication Research Laboratory GroupAdvanced Telecommunications Research Institutes International (ATR)KyotoJapan
- Department of Psychology, Graduate School of Humanities and Sociologythe University of TokyoTokyoJapan
| | - Jun Morimoto
- Brain Information Communication Research Laboratory GroupAdvanced Telecommunications Research Institutes International (ATR)KyotoJapan
- Department of Systems Science, Graduate School of InformaticsKyoto UniversityKyotoJapan
| | - Yasumasa Okamoto
- Department of Psychiatry and NeurosciencesHiroshima UniversityHiroshimaJapan
| | - Toshiya Murai
- Department of Psychiatry, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Ryu‐Ichiro Hashimoto
- Medical Institute of Developmental Disabilities ResearchShowa UniversityTokyoJapan
- Department of Language SciencesTokyo Metropolitan UniversityTokyoJapan
| | - Kiyoto Kasai
- Center for Evolutionary Cognitive Sciences, Graduate School of Art and SciencesUniversity of TokyoTokyoJapan
- University of Tokyo Institute for Diversity & Adaptation of Human Mind (UTIDAHM)TokyoJapan
- Department of Neuropsychiatry, Graduate School of MedicineUniversity of TokyoTokyoJapan
- The International Research Center for Neurointelligence (WPI‐IRCN), Institutes for Advanced Study (UTIAS)University of TokyoTokyoJapan
| | - Mitsuo Kawato
- Brain Information Communication Research Laboratory GroupAdvanced Telecommunications Research Institutes International (ATR)KyotoJapan
| | - Shinsuke Koike
- Center for Evolutionary Cognitive Sciences, Graduate School of Art and SciencesUniversity of TokyoTokyoJapan
- University of Tokyo Institute for Diversity & Adaptation of Human Mind (UTIDAHM)TokyoJapan
- The International Research Center for Neurointelligence (WPI‐IRCN), Institutes for Advanced Study (UTIAS)University of TokyoTokyoJapan
| |
Collapse
|
33
|
Mai AS, Yau CE, Tseng FS, Foo QXJ, Wang DQ, Tan EK. Linking autism spectrum disorders and parkinsonism: clinical and genetic association. Ann Clin Transl Neurol 2023; 10:484-496. [PMID: 36738194 PMCID: PMC10109258 DOI: 10.1002/acn3.51736] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/03/2023] [Accepted: 01/09/2023] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Autism spectrum disorders (ASD) comprise many complex and clinically distinct neurodevelopmental conditions, with increasing evidence linking them to parkinsonism. METHODS We searched Medline and Embase from inception to 21 March 2022 and reviewed the bibliographies of relevant articles. Studies were screened and reviewed comprehensively by two independent authors. RESULTS Of 863 references from our search, we included eight clinical studies, nine genetic studies, and five case reports. Regardless of age group, Parkinson's disease (PD) and parkinsonian syndromes were more frequently observed in patients with ASD, though the evidence for increased rates of parkinsonism is less clear for children and adolescents. Parkinsonian features and hypokinetic behavior were common in Rett syndrome, with prevalence estimates ranging from 40% to 80%. Frequently observed parkinsonian features include bradykinesia, rigidity, hypomimia, and gait freezing. PD gene PARK2 copy number variations appear more frequently in ASD cases than controls. Evidence suggests that RIT2 and CD157/BST1 are implicated in ASD and PD, while the evidence for other PD-related genes (DRD2, GPCR37, the SLC gene family, and SMPD1) is less clear. Rare mutations, such as ATP13A2, CLN3, and WDR45, could result in autistic behavior and concomitant parkinsonism. CONCLUSION The prevalence of parkinsonism in ASD is substantially greater than in the general population or matched controls. Various PD-associated gene loci, especially PARK2, could confer susceptibility to ASD as well. Important future directions include conducting prospective cohort studies to understand how parkinsonian symptoms may progress, genetic studies to reveal relevant gene loci, and pathophysiologic studies to identify potential therapeutic targets.
Collapse
Affiliation(s)
- Aaron Shengting Mai
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chun En Yau
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Fan Shuen Tseng
- Department of Neurology, Singapore General Hospital Campus, National Neuroscience Institute, Singapore, Singapore
| | - Qi Xuan Joel Foo
- Department of Neurology, Singapore General Hospital Campus, National Neuroscience Institute, Singapore, Singapore
| | - Dennis Qing Wang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Southern Medical University, Guangzhou, China
| | - Eng-King Tan
- Department of Neurology, Singapore General Hospital Campus, National Neuroscience Institute, Singapore, Singapore.,Neuroscience and Behavioural Disorders, Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
34
|
Wilson EN, Mabry S, Bradshaw JL, Gardner JJ, Rybalchenko N, Engelland R, Fadeyibi O, Osikoya O, Cushen SC, Goulopoulou S, Cunningham RL. Gestational hypoxia in late pregnancy differentially programs subcortical brain maturation in male and female rat offspring. Biol Sex Differ 2022; 13:54. [PMID: 36175941 PMCID: PMC9524087 DOI: 10.1186/s13293-022-00463-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 09/22/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Hypoxia is associated with pregnancy complications, such as preeclampsia, placental abruption, and gestational sleep apnea. Hypoxic insults during gestation can impact the brain maturation of cortical and subcortical pathways, such as the nigrostriatal pathway. However, the long-term effects of in utero hypoxic stress exposure on brain maturation in offspring are unclear, especially exposure during late gestation. The purpose of this study was to determine the impact of gestational hypoxia in late pregnancy on developmental programming of subcortical brain maturation by focusing on the nigrostriatal pathway. METHODS Timed pregnant Long-Evans rats were exposed to chronic intermittent hypoxia or room air normoxia from gestational day (GD) 15-19 (term 22-23 days). Male and female offspring were assessed during two critical periods: puberty from postnatal day (PND) 40-45 or young adulthood (PND 60-65). Brain maturation was quantified by examining (1) the structural development of the nigrostriatal pathway via analysis of locomotor behaviors and the substantia nigra dopaminergic neuronal cell bodies and (2) the refinement of the nigrostriatal pathway by quantifying ultrasonic vocalizations (USVs). RESULTS The major findings of this study are gestational hypoxia has age- and sex-dependent effects on subcortical brain maturation in offspring by adversely impacting the refinement of the nigrostriatal pathway in the absence of any effects on the structural development of the pathway. During puberty, female offspring were impacted more than male offspring, as evidenced by decreased USV call frequency, chirp USV call duration, and simple call frequency. In contrast, male offspring were impacted more than female offspring during young adulthood, as evidenced by increased latency to first USV, decreased simple USV call intensity, and increased harmonic USV call bandwidth. No effects of gestational hypoxia on the structural development of the nigrostriatal pathway were observed. CONCLUSIONS These novel findings demonstrate hypoxic insults during pregnancy mediate developmental programming of the cortical and subcortical pathways, in which male offspring exhibit long-term adverse effects compared to female offspring. Impairment of cortical and subcortical pathways maturation, such as the nigrostriatal pathway, may increase risk for neuropsychiatric disorders (e.g., mood disorders, cognitive dysfunction, brain connectivity dysfunction).
Collapse
Affiliation(s)
- E Nicole Wilson
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, School of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107, USA
| | - Steve Mabry
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, School of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107, USA
| | - Jessica L Bradshaw
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, School of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107, USA
| | - Jennifer J Gardner
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, School of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107, USA
| | - Nataliya Rybalchenko
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, School of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107, USA
| | - Rachel Engelland
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, School of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107, USA
| | - Oluwadarasimi Fadeyibi
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, School of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107, USA
| | - Oluwatobiloba Osikoya
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Spencer C Cushen
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
- Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Styliani Goulopoulou
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
- Department of Basic Sciences, Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Rebecca L Cunningham
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, School of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107, USA.
| |
Collapse
|
35
|
Koch E, Demontis D. Drug repurposing candidates to treat core symptoms in autism spectrum disorder. Front Pharmacol 2022; 13:995439. [PMID: 36172193 PMCID: PMC9510394 DOI: 10.3389/fphar.2022.995439] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 08/03/2022] [Indexed: 11/17/2022] Open
Abstract
Autism spectrum disorder (ASD) is characterized by high heritability and clinical heterogeneity. The main core symptoms are social communication deficits. There are no medications approved for the treatment of these symptoms, and medications used to treat non-specific symptoms have serious side effects. To identify potential drugs for repurposing to effectively treat ASD core symptoms, we studied ASD risk genes within networks of protein-protein interactions of gene products. We first defined an ASD network from network-based analyses, and identified approved drugs known to interact with proteins within this network. Thereafter, we evaluated if these drugs can change ASD-associated gene expression perturbations in genes in the ASD network. This was done by analyses of drug-induced versus ASD-associated gene expression, where opposite gene expression perturbations in drug versus ASD indicate that the drug could counteract ASD-associated perturbations. Four drugs showing significant (p < 0.05) opposite gene expression perturbations in drug versus ASD were identified: Loperamide, bromocriptine, drospirenone, and progesterone. These drugs act on ASD-related biological systems, indicating that these drugs could effectively treat ASD core symptoms. Based on our bioinformatics analyses of ASD genetics, we shortlist potential drug repurposing candidates that warrant clinical translation to treat core symptoms in ASD.
Collapse
Affiliation(s)
- Elise Koch
- Norwegian Centre for Mental Disorders Research (NORMENT), University of Oslo and Oslo University Hospital, Oslo, Norway
- *Correspondence: Elise Koch,
| | - Ditte Demontis
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Department of Biomedicine (Human Genetics) and Centre for Integrative Sequencing, Aarhus University, Aarhus, Denmark
- Center for Genomics and Personalized Medicine, Aarhus, Denmark
| |
Collapse
|