1
|
Ozler E, Sanlier N. Nutritional Approaches in Autism Spectrum Disorder: A Scoping Review. Curr Nutr Rep 2025; 14:61. [PMID: 40259156 PMCID: PMC12011661 DOI: 10.1007/s13668-025-00655-y] [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] [Accepted: 04/14/2025] [Indexed: 04/23/2025]
Abstract
PURPOSE OF REVIEW This review was conducted to discuss the etiology of autism in the light of current information, to draw attention to the fact that defects in different biological mechanisms cause autism, and to examine the effectiveness of dietary interventions and supplements in relieving ASD symptoms. RECENT FINDINGS Autism spectrum disorder (ASD) is an extremely heterogeneous condition characterized by delays in reciprocal social interaction and communication skills, stereotyped behaviors, and a narrowed range of interests and limited activities. Comorbid conditions such as cognitive impairment, epilepsy, psychiatric diseases, and behavioral symptoms such as impaired social communication, repetitive behaviors, lack of interest in the environment, nutritional disorders, gastrointestinal diseases and abnormal (dysbiotic) states, sleep disorders, and dysmorphism are frequently encountered in individuals with ASD. Although nutrition is one of the environmental factors affecting ASD, it can also be effective in alleviating the behavioral and gastrointestinal symptoms of ASD. Various dietary models (GFCF diet, low glycemic index diet, ketogenic diet, specific carbohydrate diet, Mediterranean diet, GAPS, Feingold, Candida body ecology, allergy elimination diets, etc.) and supplements (vitamin D, polyunsaturated fatty acids, probiotics and prebiotics, phytochemicals) can be used to alleviate symptoms in individuals with ASD. The effectiveness and reliability of dietary interventions in individuals with ASD are a matter of significant debate, and the evidence for these practices is limited. Furthermore, there is no consensus on establishing an ideal nutritional model for individuals with ASD.
Collapse
Affiliation(s)
- Ebru Ozler
- Department of Nutrition and Dietetics, School of Health Sciences, Ankara Medipol University, 06050, Altındağ, Ankara, Turkey
| | - Nevin Sanlier
- Department of Nutrition and Dietetics, School of Health Sciences, Ankara Medipol University, 06050, Altındağ, Ankara, Turkey.
| |
Collapse
|
2
|
Li J, Zhai P, Bi L, Wang Y, Yang X, Yang Y, Li N, Dang W, Feng G, Li P, Liu Y, Zhang Q, Mei X. Associations between amino acid levels and autism spectrum disorder severity. BMC Psychiatry 2025; 25:332. [PMID: 40186136 PMCID: PMC11969702 DOI: 10.1186/s12888-025-06771-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 03/24/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) imposes a significant burden on both patients and society. Amino acid metabolism abnormalities are particularly relevant to ASD pathology due to their crucial role in neurotransmitter synthesis, synaptic function, and overall neurodevelopment. This study aims to explore the association between amino acid metabolic abnormalities and the severity of ASD by analyzing the amino acid concentrations in the blood of children with ASD. METHODS Fasting peripheral blood samples were collected from 344 children with ASD, and amino acid concentrations were measured using liquid chromatography-tandem mass spectrometry (LC-MS/MS) while strictly following quality control measures. The association between amino acid concentrations and ASD severity was evaluated using logistic regression and restricted cubic spline (RCS) analysis. The ROC (receiver operating characteristic) curve, decision curve analysis (DCA), and calibration curve were used to construct and validate predictive models and nomograms, thereby assessing their predictive performance. RESULTS Multivariate logistic regression analysis showed that aspartic acid (OR = 1.037, 95% CI: 1.009-1.068, P = 0.01), glutamic acid (OR = 1.009, 95% CI: 1.001-1.017, P = 0.03), phenylalanine (OR = 1.036, 95% CI: 1.003-1.072, P = 0.04), and leucine/isoleucine (OR = 1.021, 95% CI: 1.006-1.039, P = 0.01) were significantly positively correlated with the severity of ASD. On the other hand, tryptophan (OR = 0.935, 95% CI: 0.903-0.965, P < 0.01) and valine (OR = 0.987, 95% CI: 0.977-0.997, P = 0.01) were significantly negatively correlated with the severity of ASD. RCS analysis further revealed a nonlinear relationship between the concentrations of aspartic acid, proline, and glutamic acid and the risk of ASD. ROC curve analysis showed that the combined model achieved an AUC (area under the curve) of 0.806, indicating high diagnostic accuracy. Calibration and decision curve analysis further validated the predictive effectiveness and clinical utility of the model. CONCLUSIONS This study identifies potential amino acid biomarkers that may contribute to ASD severity assessment. Further research is needed to validate these findings and explore their clinical utility.
Collapse
Affiliation(s)
- Jing Li
- Department of Pediatrics, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, 450000, China
- School of Pediatric Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China
| | - Panpan Zhai
- Department of Pediatrics, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, 450000, China
- School of Pediatric Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China
| | - Liangliang Bi
- Department of Pediatrics, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, 450000, China
- School of Pediatric Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China
| | - Ying Wang
- Department of Pediatrics, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, 450000, China
- School of Pediatric Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China
| | - Xiaoqing Yang
- Department of Pediatrics, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, 450000, China
- School of Pediatric Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China
| | - Yueli Yang
- Department of Pediatrics, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, 450000, China
- School of Pediatric Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China
| | - Nan Li
- Beijing Fuyou Longhui Genetic Disease Clinic, Beijing, Beijing, 100070, China
| | - Weili Dang
- Department of Pediatrics, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, 450000, China.
- School of Pediatric Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China.
| | - Gang Feng
- Department of Pediatrics, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, 450000, China.
- School of Pediatric Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China.
| | - Pei Li
- Beijing Fuyou Longhui Genetic Disease Clinic, Beijing, Beijing, 100070, China
| | - Yuan Liu
- Beijing Fuyou Longhui Genetic Disease Clinic, Beijing, Beijing, 100070, China
| | - Qiushuang Zhang
- Department of Pediatrics, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, 450000, China
- School of Pediatric Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China
| | - Xiaofeng Mei
- Department of Pediatrics, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, 450000, China
- School of Pediatric Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China
| |
Collapse
|
3
|
Meng Y, Hu Y, Xue Y, Zhen Z. Metabolomic Profiling of the Striatum in Shank3 Knockout ASD Rats: Effects of Early Swimming Regulation. Metabolites 2025; 15:134. [PMID: 39997759 PMCID: PMC11857520 DOI: 10.3390/metabo15020134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/27/2025] [Accepted: 02/05/2025] [Indexed: 02/26/2025] Open
Abstract
Objectives: This study aimed to investigate the regulatory impact of early swimming intervention on striatal metabolism in Shank3 gene knockout ASD model rats. Methods:Shank3 gene knockout exon 11-21 male 8-day-old SD rats were used as experimental subjects and randomly divided into the following three groups: a Shank3 knockout control group (KC), a wild-type control group (WC) from the same litter, and a Shank3 knockout swimming group (KS). The rats in the exercise group received early swimming intervention for 8 weeks starting at 8 days old. LC-MS metabolism was employed to detect the changes in metabolites in the striatum. Results: There were 17 differential metabolites (14 down-regulated) between the KC and WC groups, 19 differential metabolites (18 up-regulated) between the KS and KC groups, and 22 differential metabolites (18 up-regulated) between the KS and WC groups. Conclusions: The metabolism of striatum in Shank3 knockout ASD model rats is disrupted, involving metabolites related to synaptic morphology, and the Glu and GABAergic synapses are abnormal. Early swimming intervention regulated the striatal metabolome group of the ASD model rats, with differential metabolites primarily related to nerve development, synaptic membrane structure, and synaptic signal transduction.
Collapse
Affiliation(s)
- Yunchen Meng
- Department of Physical Education and Research, China University of Mining and Technology—Beijing, Beijing 100083, China;
| | - Yiling Hu
- Department of Physical Education and Research, China University of Mining and Technology—Beijing, Beijing 100083, China;
| | - Yaqi Xue
- College of P.E and Sports, Beijing Normal University, Beijing 100875, China;
| | - Zhiping Zhen
- College of P.E and Sports, Beijing Normal University, Beijing 100875, China;
| |
Collapse
|
4
|
Osama A, Anwar AM, Ezzeldin S, Ahmed EA, Mahgoub S, Ibrahim O, Ibrahim SA, Abdelhamid IA, Bakry U, Diab AA, A Sayed A, Magdeldin S. Integrative multi-omics analysis of autism spectrum disorder reveals unique microbial macromolecules interactions. J Adv Res 2025:S2090-1232(25)00055-4. [PMID: 39870302 DOI: 10.1016/j.jare.2025.01.036] [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: 08/31/2024] [Revised: 01/23/2025] [Accepted: 01/23/2025] [Indexed: 01/29/2025] Open
Abstract
INTRODUCTION Gut microbiota alterations have been implicated in Autism Spectrum Disorder (ASD), yet the mechanisms linking these changes to ASD pathophysiology remain unclear. OBJECTIVES This study utilized a multi-omics approach to uncover mechanisms linking gut microbiota to ASD by examining microbial diversity, bacterial metaproteins, associated metabolic pathways and host proteome. METHODS The gut microbiota of 30 children with severe ASD and 30 healthy controls was analyzed. Microbial diversity was assessed using 16S rRNA V3 and V4 sequencing. A novel metaproteomics pipeline identified bacterial proteins, while untargeted metabolomics explored altered metabolic pathways. Finally, multi-omics integration was employed to connect macromolecular changes to neurodevelopmental deficits. RESULTS Children with ASD exhibited significant alterations in gut microbiota, including lower diversity and richness compared to controls. Tyzzerella was uniquely associated with the ASD group. Microbial network analysis revealed rewiring and reduced stability in ASD. Major metaproteins identified were produced by Bifidobacterium and Klebsiella (e.g., xylose isomerase and NADH peroxidase). Metabolomics profiling identified neurotransmitters (e.g., glutamate, DOPAC), lipids, and amino acids capable of crossing the blood-brain barrier, potentially contributing to neurodevelopmental and immune dysregulation. Host proteome analysis revealed altered proteins, including kallikrein (KLK1) and transthyretin (TTR), involved in neuroinflammation and immune regulation. Finally, multi-omics integration supported single-omics findings and reinforced the hypothesis that gut microbiota and their macromolecular products may contribute to ASD-associated symptoms. CONCLUSIONS The integration of multi-omics data provided critical evidence that alteration in gut microbiota and associated macromolecule production may play a role in ASD-related symptoms and co-morbidities. Key bacterial metaproteins and metabolites were identified as potential contributors to neurological and immune dysregulation in ASD, underscoring possible novel targets for therapeutic intervention.
Collapse
Affiliation(s)
- Aya Osama
- Proteomics and Metabolomics Unit, Basic Research Department, Children's Cancer Hospital, 57357 Cairo, (CCHE-57357), Egypt
| | - Ali Mostafa Anwar
- Proteomics and Metabolomics Unit, Basic Research Department, Children's Cancer Hospital, 57357 Cairo, (CCHE-57357), Egypt
| | - Shahd Ezzeldin
- Proteomics and Metabolomics Unit, Basic Research Department, Children's Cancer Hospital, 57357 Cairo, (CCHE-57357), Egypt
| | - Eman Ali Ahmed
- Proteomics and Metabolomics Unit, Basic Research Department, Children's Cancer Hospital, 57357 Cairo, (CCHE-57357), Egypt; Department of Pharmacology, Faculty of Veterinary Medicine, Suez Canal University, 41522 Ismailia, Egypt
| | - Sebaey Mahgoub
- Proteomics and Metabolomics Unit, Basic Research Department, Children's Cancer Hospital, 57357 Cairo, (CCHE-57357), Egypt
| | - Omneya Ibrahim
- Psychiatry and Neurology Department, Faculty of Medicine, Suez Canal University, Egypt
| | | | | | - Usama Bakry
- Egypt Center for Research and Regenerative Medicine (ECRRM), Egypt
| | - Aya A Diab
- Genomic Research Program, Basic Research Department, Children's Cancer Hospital Egypt 57357, 11441 Cairo, Egypt
| | - Ahmed A Sayed
- Genomic Research Program, Basic Research Department, Children's Cancer Hospital Egypt 57357, 11441 Cairo, Egypt; Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Sameh Magdeldin
- Proteomics and Metabolomics Unit, Basic Research Department, Children's Cancer Hospital, 57357 Cairo, (CCHE-57357), Egypt; Department of Physiology, Faculty of Veterinary Medicine, Suez Canal University, 41522 Ismailia, Egypt.
| |
Collapse
|
5
|
Długosz A, Wróblewski M, Błaszak B, Szulc J. The Role of Nutrition, Oxidative Stress, and Trace Elements in the Pathophysiology of Autism Spectrum Disorders. Int J Mol Sci 2025; 26:808. [PMID: 39859522 PMCID: PMC11765825 DOI: 10.3390/ijms26020808] [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: 12/27/2024] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 01/27/2025] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental condition characterized by deficits in social communication and interaction, alongside repetitive behaviors, and atypical sensory-motor patterns. The growing prevalence of ASD has driven substantial advancements in research aimed at understanding its etiology, preventing its onset, and mitigating its impact. This ongoing effort necessitates continuous updates to the body of knowledge and the identification of previously unexplored factors. The present study addresses this need by examining the roles of nutrition, oxidative stress, and trace elements in the pathophysiology of ASD. In this review, an overview is provided of the key dietary recommendations for individuals with ASD, including gluten-free and casein-free (GFCF) diets, ketogenic diets (KDs), and other nutritional interventions. Furthermore, it explores the involvement of oxidative stress in ASD and highlights the significance of trace elements in maintaining neuropsychiatric health. The impact of these factors on molecular and cellular mechanisms was discussed, alongside therapeutic strategies and their efficacy in managing ASD.
Collapse
Affiliation(s)
- Anna Długosz
- Department of Food Industry Technology and Engineering, Faculty of Chemical Technology and Engineering, Bydgoszcz University of Science and Technology, 3 Seminaryjna St., 85-326 Bydgoszcz, Poland; (B.B.); (J.S.)
| | - Marcin Wróblewski
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 24 Karłowicza St., 85-092 Bydgoszcz, Poland;
| | - Błażej Błaszak
- Department of Food Industry Technology and Engineering, Faculty of Chemical Technology and Engineering, Bydgoszcz University of Science and Technology, 3 Seminaryjna St., 85-326 Bydgoszcz, Poland; (B.B.); (J.S.)
| | - Joanna Szulc
- Department of Food Industry Technology and Engineering, Faculty of Chemical Technology and Engineering, Bydgoszcz University of Science and Technology, 3 Seminaryjna St., 85-326 Bydgoszcz, Poland; (B.B.); (J.S.)
| |
Collapse
|
6
|
Majumder P, Chatterjee B, Akter K, Ahsan A, Tan SJ, Huang CC, Chu JF, Shen CKJ. Molecular switch of the dendrite-to-spine transport of TDP-43/FMRP-bound neuronal mRNAs and its impairment in ASD. Cell Mol Biol Lett 2025; 30:6. [PMID: 39815169 PMCID: PMC11737055 DOI: 10.1186/s11658-024-00684-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 12/23/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Regulation of messenger RNA (mRNA) transport and translation in neurons is essential for dendritic plasticity and learning/memory development. The trafficking of mRNAs along the hippocampal neuron dendrites remains translationally silent until they are selectively transported into the spines upon glutamate-induced receptor activation. However, the molecular mechanism(s) behind the spine entry of dendritic mRNAs under metabotropic glutamate receptor (mGluR)-mediated neuroactivation and long-term depression (LTD) as well as the fate of these mRNAs inside the spines are still elusive. METHOD Different molecular and imaging techniques, e.g., immunoprecipitation (IP), RNA-IP, Immunofluorescence (IF)/fluorescence in situ hybridization (FISH), live cell imaging, live cell tracking of RNA using beacon, and mouse model study are used to elucidate a novel mechanism regulating dendritic spine transport of mRNAs in mammalian neurons. RESULTS We demonstrate here that brief mGluR1 activation-mediated dephosphorylation of pFMRP (S499) results in the dissociation of FMRP from TDP-43 and handover of TDP-43/Rac1 mRNA complex from the dendritic transport track on microtubules to myosin V track on the spine actin filaments. Rac1 mRNA thus enters the spines for translational reactivation and increases the mature spine density. In contrast, during mGluR1-mediated neuronal LTD, FMRP (S499) remains phosphorylated and the TDP-43/Rac1 mRNA complex, being associated with kinesin 1-FMRP/cortactin/drebrin, enters the spines owing to Ca2+-dependent microtubule invasion into spines, but without translational reactivation. In a VPA-ASD mouse model, this regulation become anomalous. CONCLUSIONS This study, for the first time, highlights the importance of posttranslational modification of RBPs, such as the neurodevelopmental disease-related protein FMRP, as the molecular switch regulating the dendrite-to-spine transport of specific mRNAs under mGluR1-mediated neurotransmissions. The misregulation of this switch could contribute to the pathogenesis of FMRP-related neurodisorders including the autism spectrum disorder (ASD). It also could indicate a molecular connection between ASD and neurodegenerative disease-related protein TDP-43 and opens up a new perspective of research to elucidate TDP-43 proteinopathy among patients with ASD.
Collapse
Affiliation(s)
- Pritha Majumder
- PhD Program in Medical Neuroscience, Taipei Medical University, Taipei, Taiwan (R.O.C.).
- Institute of Molecular Medicine, College of Medicine, National Chen Kung University, Tainan, Taiwan (R.O.C.).
| | - Biswanath Chatterjee
- PhD Program in Medical Neuroscience, Taipei Medical University, Taipei, Taiwan (R.O.C.)
| | - Khadiza Akter
- PhD Program in Medical Neuroscience, Taipei Medical University, Taipei, Taiwan (R.O.C.)
| | - Asmar Ahsan
- PhD Program in Medical Neuroscience, Taipei Medical University, Taipei, Taiwan (R.O.C.)
| | - Su Jie Tan
- Institute of Molecular Medicine, College of Medicine, National Chen Kung University, Tainan, Taiwan (R.O.C.)
| | - Chi-Chen Huang
- PhD Program in Medical Neuroscience, Taipei Medical University, Taipei, Taiwan (R.O.C.)
| | - Jen-Fei Chu
- PhD Program in Medical Neuroscience, Taipei Medical University, Taipei, Taiwan (R.O.C.).
| | - Che-Kun James Shen
- PhD Program in Medical Neuroscience, Taipei Medical University, Taipei, Taiwan (R.O.C.).
- Institute of Molecular Biology, Academia Sinica, Nangang, Taipei, 115, Taiwan (R.O.C.).
| |
Collapse
|
7
|
Ferraro S, Saielli L, Biganzoli D, Tosi M, Guidi L, Longo R, Severino F, Carelli S, Rossi M, Pisciotta L, Ricci E, Brustia F, Verduci E, Zuccotti G, Mussap M, Cereda C. Amino Acid Patterns in Children with Autistic Spectrum Disorder: A Preliminary Biochemical Evaluation. Nutrients 2025; 17:274. [PMID: 39861405 PMCID: PMC11767892 DOI: 10.3390/nu17020274] [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: 12/04/2024] [Revised: 01/05/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND The metabolism of plasma amino acid (AA) in children with autism spectrum disorder (ASD) has been extensively investigated, yielding inconclusive results. This study aims to characterize the metabolic alterations in AA profiles among early-diagnosed children with ASD and compare the findings with those from non-ASD children. METHODS We analyzed plasma AA profiles, measured by ion exchange chromatography, from 1242 ASD children (median age = 4 years; 81% male). Additionally, we studied AA profiles from 488 children, matched for age and free of ASD (control group). Principal component and cluster analysis were employed to explore potential associations within the ASD group and to identify subgroups. RESULTS We observed lower plasma levels of glutamine in children with ASD compared to non-ASD children (p < 0.001). Six essential, two conditionally essential, and four non-essential AA were found to be increased in children with ASD. The clustering analysis revealed two groups, labeled Neurological (NEU) and Nutritional (NUT), which included a majority of ASD children (94% and 78%, respectively). The NEU group exhibited high levels of taurine, aspartate, glutamic acid, and ornithine, while the NUT group showed elevated levels of branched-chain AA. CONCLUSIONS In children with ASD, we identified some heterogeneous AA patterns that may serve as biochemical signatures of neurological impairment in some individuals, while in others they may indicate nutritional dysregulation.
Collapse
Affiliation(s)
- Simona Ferraro
- Department of Pediatrics, Buzzi Children’s Hospital, 20154 Milan, Italy (C.C.)
| | - Laura Saielli
- Center of Functional Genomics and Rare Diseases, Buzzi Children’s Hospital, 20154 Milan, Italy
| | - Davide Biganzoli
- Department of Pediatrics, Buzzi Children’s Hospital, 20154 Milan, Italy (C.C.)
| | - Martina Tosi
- Department of Pediatrics, Buzzi Children’s Hospital, 20154 Milan, Italy (C.C.)
- Department of Health Sciences, University of Milan, 20146 Milan, Italy
| | - Laura Guidi
- Center of Functional Genomics and Rare Diseases, Buzzi Children’s Hospital, 20154 Milan, Italy
| | - Roberto Longo
- Corporate Information Systems, Buzzi Children’s Hospital, 20154 Milan, Italy
| | - Francesca Severino
- Department of Biomedical and Clinical Sciences, University of Milan, 20157 Milan, Italy;
| | - Stephana Carelli
- Department of Pediatrics, Buzzi Children’s Hospital, 20154 Milan, Italy (C.C.)
- Department of Biomedical and Clinical Sciences, University of Milan, 20157 Milan, Italy;
- Pediatric Clinical Research Center “Romeo ed Enrica Invernizzi”, University of Milan, 20157 Milan, Italy
| | - Maura Rossi
- Child and Adolescent Neuropsychiatry Unit, ASST Fatebenefratelli Sacco, 20157 Milan, Italy (L.P.)
| | - Livia Pisciotta
- Child and Adolescent Neuropsychiatry Unit, ASST Fatebenefratelli Sacco, 20157 Milan, Italy (L.P.)
| | - Emilia Ricci
- Child Neuropsychiatry Unit, Epilepsy Center, San Paolo Hospital, Department of Health Sciences, University of Milan, 20142 Milan, Italy;
| | - Francesca Brustia
- Child Neuropsychiatry Unit, University Hospital Maggiore della Carità, 28100 Novara, Italy
| | - Elvira Verduci
- Department of Health Sciences, University of Milan, 20146 Milan, Italy
- Metabolic Diseases Unit, Buzzi Children’s Hospital, 20154 Milan, Italy
| | - Gianvincenzo Zuccotti
- Department of Pediatrics, Buzzi Children’s Hospital, 20154 Milan, Italy (C.C.)
- Department of Biomedical and Clinical Sciences, University of Milan, 20157 Milan, Italy;
| | - Michele Mussap
- Laboratory Medicine, Hospital Foundation Villa Salus, 30174 Venice, Italy;
- Molecular Unit, Department of Surgical Sciences, University of Cagliari, 09124 Cagliari, Italy
| | - Cristina Cereda
- Department of Pediatrics, Buzzi Children’s Hospital, 20154 Milan, Italy (C.C.)
- Center of Functional Genomics and Rare Diseases, Buzzi Children’s Hospital, 20154 Milan, Italy
| |
Collapse
|
8
|
Lichtarge J, Cappuccio G, Pati S, Dei-Ampeh AK, Sing S, Ma L, Liu Z, Maletic-Savatic M. MetaboLINK is a novel algorithm for unveiling cell-specific metabolic pathways in longitudinal datasets. Front Neurosci 2025; 18:1520982. [PMID: 39872998 PMCID: PMC11769959 DOI: 10.3389/fnins.2024.1520982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 12/20/2024] [Indexed: 01/30/2025] Open
Abstract
Introduction In the rapidly advancing field of 'omics research, there is an increasing demand for sophisticated bioinformatic tools to enable efficient and consistent data analysis. As biological datasets, particularly metabolomics, become larger and more complex, innovative strategies are essential for deciphering the intricate molecular and cellular networks. Methods We introduce a pioneering analytical approach that combines Principal Component Analysis (PCA) with Graphical Lasso (GLASSO). This method is designed to reduce the dimensionality of large datasets while preserving significant variance. For the first time, we applied the PCA-GLASSO algorithm (i.e., MetaboLINK) to metabolomics data derived from Nuclear Magnetic Resonance (NMR) spectroscopy performed on neural cells at various developmental stages, from human embryonic stem cells to neurons. Results The MetaboLINK analysis of longitudinal metabolomics data has revealed distinct pathways related to amino acids, lipids, and energy metabolism, uniquely associated with specific cell progenies. These findings suggest that different metabolic pathways play a critical role at different stages of cellular development, each contributing to diverse cellular functions. Discussion Our study demonstrates the efficacy of the MetaboLINK approach in analyzing NMR-based longitudinal metabolomic datasets, highlighting key metabolic shifts during cellular transitions. We share the methodology and the code to advance general 'omics research, providing a powerful tool for dissecting large datasets in neurobiology and other fields.
Collapse
Affiliation(s)
- Jared Lichtarge
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, United States
| | - Gerarda Cappuccio
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, United States
- Department of Pediatrics-Neurology, Baylor College of Medicine, Houston, TX, United States
| | - Soumya Pati
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, United States
- Department of Pediatrics-Neurology, Baylor College of Medicine, Houston, TX, United States
| | - Alfred Kwabena Dei-Ampeh
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, United States
- Department of Pediatrics-Neurology, Baylor College of Medicine, Houston, TX, United States
| | - Senghong Sing
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, United States
- Department of Pediatrics-Neurology, Baylor College of Medicine, Houston, TX, United States
- College of Natural Sciences and Mathematics, University of Houston, Houston, TX, United States
| | - LiHua Ma
- Shared Equipment Authority, Rice University, Houston, TX, United States
| | - Zhandong Liu
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, United States
- Department of Pediatrics-Neurology, Baylor College of Medicine, Houston, TX, United States
| | - Mirjana Maletic-Savatic
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, United States
- Department of Pediatrics-Neurology, Baylor College of Medicine, Houston, TX, United States
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
9
|
Hosseini S, Ghadimi M, Reyhani N, Khazaei S, Rahmatkhah-Yazdi M, Soleimani-Farsani R, Vaseghi S. BDNF and GSK-3beta expression changes underlie the beneficial effects of crocin on behavioral alterations in a rat model of autism induced by prenatal valproic acid administration. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-024-03777-2. [PMID: 39777538 DOI: 10.1007/s00210-024-03777-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 12/28/2024] [Indexed: 01/11/2025]
Abstract
Autism spectrum disorder (ASD) is a serious neurodevelopmental disorder characterized by impairments in social interaction, language, and communication and induction of stereotypic behavior. In rodents, prenatal administration of valproic acid (often on 12.5 gestational days) is used for the induction of an ASD-like model. In the present study, we aimed to assess the potential therapeutic effects of crocin (a major component of Saffron, a neuroprotective and anti-inflammatory agent) on behavioral dysfunctions with respect to the level of brain-derived neurotrophic factor (BDNF) and glycogen synthase kinase-3 beta (GSK-3beta) in the medial prefrontal cortex. Valproic acid was intraperitoneally injected at the dose of 600 mg/kg on 12.5 gestational days. BDNF and GSK-3beta expression levels were also measured using real-time PCR. Locomotion, anxiety-like behavior, grooming, and sniffing were also measured in the open-field test. The results showed that prenatal valproic acid administration induced hyperactivity, anxiety-like behavior, increased grooming and sniffing (stereotyped behavior), decreased BDNF levels, and increased GSK-3beta levels in the medial prefrontal cortex. However, crocin dose-dependently restored the effects of prenatal valproic acid administration on behavioral functions and gene expressions. In conclusion, we suggested that BDNF and GSK-3beta expression changes in the medial prefrontal cortex may underlie the pathophysiology of ASD. The therapeutic effects of crocin may be also related to counteracting BDNF and GSK-3beta expression changes induced by prenatal valproic acid.
Collapse
Affiliation(s)
- Seyedehfatemeh Hosseini
- Cognitive Neuroscience Lab, Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran
| | - Mozhgan Ghadimi
- Cognitive Neuroscience Lab, Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran
| | - Niloufar Reyhani
- Cognitive Neuroscience Lab, Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran
| | - Sepideh Khazaei
- Cognitive Neuroscience Lab, Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran
| | - Majid Rahmatkhah-Yazdi
- Cognitive Neuroscience Lab, Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran
| | - Reza Soleimani-Farsani
- Cognitive Neuroscience Lab, Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran
| | - Salar Vaseghi
- Cognitive Neuroscience Lab, Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran.
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran.
| |
Collapse
|
10
|
Lin YL, Yao T, Wang YW, Lu JH, Chen YM, Wu YQ, Qian XG, Liu JC, Fang LX, Zheng C, Wu CH, Lin JF. Causal association between mitochondrial function and psychiatric disorders: Insights from a bidirectional two-sample Mendelian randomization study. J Affect Disord 2025; 368:55-66. [PMID: 39265869 DOI: 10.1016/j.jad.2024.09.039] [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: 04/01/2024] [Revised: 09/04/2024] [Accepted: 09/08/2024] [Indexed: 09/14/2024]
Abstract
BACKGROUND Previous observational studies have suggested that there appears to be a close association between mitochondrial function and psychiatric disorders, but whether a causal role exists remains unclear. METHODS We extracted genetic instruments for 67 mitochondrial-related proteins and 10 psychiatric disorders from publicly available genome-wide association studies, and employed five distinct MR methods and false discovery rate correction to detect causal associations between them. Additionally, we conducted a series of sensitivity tests and additional model analysis to ensure the robustness of the results. For potential causal associations, we further performed reverse MR analyses to assess the impact of reverse causality. RESULTS We identified a total of 2 significant causal associations and 24 suggestive causal associations. Specifically, Phenylalanine-tRNA ligase was found to increase the risk of Alzheimer's disease, while Mitochondrial glutamate carrier 2 decreased the risk of autism spectrum disorder. Furthermore, there was no evidence of significant pleiotropy, heterogeneity, or reverse causality. LIMITATIONS This study was limited to individuals of European ancestry, and the conclusions drawn are merely revelatory. CONCLUSION This study provides novel insights into the relationship between mitochondria and psychiatric disorders, as well as the pathogenesis and treatment strategies for psychiatric disorders.
Collapse
Affiliation(s)
- Yun-Lu Lin
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Tao Yao
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Ying-Wei Wang
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Jia-Hao Lu
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Yan-Min Chen
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Yu-Qing Wu
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Xin-Ge Qian
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Jing-Chen Liu
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Luo-Xiang Fang
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Cheng Zheng
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Chun-Hui Wu
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Cardiovascular Development and Translational Medicine, Wenzhou Medical University, Wenzhou 325027, Zhejiang, China; Department of Ultrasonography, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China.
| | - Jia-Feng Lin
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China.
| |
Collapse
|
11
|
Cheong E, Lee CJ. Gliotransmission in physiologic and pathologic conditions. HANDBOOK OF CLINICAL NEUROLOGY 2025; 209:93-116. [PMID: 40122634 DOI: 10.1016/b978-0-443-19104-6.00003-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
This chapter explores the roles of gliotransmission in physiologic and pathologic conditions, including psychiatric and neurologic disorders. Gliotransmission, facilitated by astrocytes through the release of gliotransmitters such as glutamate, d-serine, and GABA, regulates neuronal activity and synaptic transmission. Under physiologic conditions, astrocytic gliotransmission maintains the balance of tonic excitation and inhibition, influencing synaptic plasticity and cognitive functions. In psychiatric disorders, the chapter examines how dysregulated gliotransmission contributes to major depression and schizophrenia. In major depression, changes in astrocytic glutamate and adenosine signaling impact mood regulation and cognitive functions. Schizophrenia involves complex astrocyte-neuron interactions, with dysregulated astrocytic activity affecting synaptic function and contributing to symptoms. The chapter also delves into neurologic disorders. In Alzheimer disease, aberrant GABA release from reactive astrocytes impairs memory and cognitive functions. Parkinson disease features alterations in glutamatergic and GABAergic systems, affecting motor and nonmotor symptoms. Epilepsy involves a disruption in the balance between excitatory and inhibitory neurotransmission, with astrocytic GABA accumulation helping to maintain neuronal stability. Autism spectrum disorder (ASD) is linked to imbalances in glutamatergic and GABAergic neurotransmission, underlying sensory, cognitive, and social impairments. Overall, the chapter underscores the pivotal role of gliotransmission in maintaining neural homeostasis and highlights its potential as a therapeutic target in various disorders.
Collapse
Affiliation(s)
- Eunji Cheong
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea.
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, South Korea.
| |
Collapse
|
12
|
Zhang J, Toulopoulou T, Li Q, Niu L, Peng L, Dai H, Chen K, Wang X, Huang R, Wei X, Zhang R. Charting brain GABA and glutamate levels across psychiatric disorders by quantitative analysis of 121 1H-MRS studies. Psychol Med 2024:1-12. [PMID: 39564744 DOI: 10.1017/s0033291724001673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
BACKGROUND Psychiatric diagnosis is based on categorical diagnostic classification, yet similarities in genetics and clinical features across disorders suggest that these classifications share commonalities in neurobiology, particularly regarding neurotransmitters. Glutamate (Glu) and gamma-aminobutyric acid (GABA), the brain's primary excitatory and inhibitory neurotransmitters, play critical roles in brain function and physiological processes. METHODS We examined the levels of Glu, combined glutamate and glutamine (Glx), and GABA across psychiatric disorders by pooling data from 121 1H-MRS studies and further divided the sample based on Axis I disorders. RESULTS Statistically significant differences in GABA levels were found in the combined psychiatric group compared with healthy controls (Hedge's g = -0.112, p = 0.008). Further analyses based on brain regions showed that brain GABA levels significantly differed across Axis I disorders and controls in the parieto-occipital cortex (Hedge's g = 0.277, p = 0.019). Furthermore, GABA levels were reduced in affective disorders in the occipital cortex (Hedge's g = -0.468, p = 0.043). Reductions in Glx levels were found in neurodevelopmental disorders (Hedge's g = -0.287, p = 0.022). Analysis focusing on brain regions suggested that Glx levels decreased in the frontal cortex (Hedge's g = -0.226, p = 0.025), and the reduction of Glu levels in patients with affective disorders in the frontal cortex is marginally significant (Hedge's g = -0.172, p = 0.052). When analyzing the anterior cingulate cortex and prefrontal cortex separately, reductions were only found in GABA levels in the former (Hedge's g = - 0.191, p = 0.009) across all disorders. CONCLUSIONS Altered glutamatergic and GABAergic metabolites were found across psychiatric disorders, indicating shared dysfunction. We found reduced GABA levels across psychiatric disorders and lower Glu levels in affective disorders. These results highlight the significance of GABA and Glu in psychiatric etiology and partially support rethinking current diagnostic categories.
Collapse
Affiliation(s)
- Jiayuan Zhang
- Laboratory of Cognitive Control and Brain Healthy, Department of Psychology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Timothea Toulopoulou
- Department of Psychology & National Magnetic Resonance Research Center (UMRAM) & Aysel Sabuncu Brain Research Center, Bilkent University, Ankara, Turkey
- Department of Psychiatry, National and Kapodistrian University of Athens, Athens, Greece
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Qian Li
- Laboratory of Cognitive Control and Brain Healthy, Department of Psychology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Lijing Niu
- Laboratory of Cognitive Control and Brain Healthy, Department of Psychology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Lanxin Peng
- Laboratory of Cognitive Control and Brain Healthy, Department of Psychology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Haowei Dai
- Laboratory of Cognitive Control and Brain Healthy, Department of Psychology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Keyin Chen
- Laboratory of Cognitive Control and Brain Healthy, Department of Psychology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xingqin Wang
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital of Southern Medical University, Guangzhou, PR China
| | - Ruiwang Huang
- School of Psychology, South China Normal University, Guangzhou, China
| | - Xinhua Wei
- Department of Radiology, Guangzhou First Affiliated Hospital, Guangzhou, PR China
| | - Ruibin Zhang
- Laboratory of Cognitive Control and Brain Healthy, Department of Psychology, School of Public Health, Southern Medical University, Guangzhou, China
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases
- Department of Psychiatry, Zhujiang Hospital, Southern Medical University, Guangzhou, PR China
| |
Collapse
|
13
|
Grumbach P, Kasper J, Hipp JF, Forsyth A, Valk SL, Muthukumaraswamy S, Eickhoff SB, Schilbach L, Dukart J. Local activity alterations in autism spectrum disorder correlate with neurotransmitter properties and ketamine induced brain changes. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.20.24315801. [PMID: 39502665 PMCID: PMC11537324 DOI: 10.1101/2024.10.20.24315801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/14/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental condition associated with altered resting-state brain function. An increased excitation-inhibition (E/I) ratio is discussed as a potential pathomechanism but in-vivo evidence of disturbed neurotransmission underlying these functional alterations remains scarce. We compared rs-fMRI local activity (LCOR) between ASD (N=405, N=395) and neurotypical controls (N=473, N=474) in two independent cohorts (ABIDE1 and ABIDE2). We then tested how these LCOR alterations co-localize with specific neurotransmitter systems derived from nuclear imaging and compared them with E/I changes induced by GABAergic (midazolam) and glutamatergic medication (ketamine). Across both cohorts, ASD subjects consistently exhibited reduced LCOR, particularly in higher-order default mode network nodes, alongside increases in bilateral temporal regions, the cerebellum, and brainstem. These LCOR alterations negatively co-localized with dopaminergic (D1, D2, DAT), glutamatergic (NMDA, mGluR5), GABAergic (GABAa) and cholinergic neurotransmission (VAChT). The NMDA-antagonist ketamine, but not GABAa-potentiator midazolam, induced LCOR changes which co-localize with D1, NMDA and GABAa receptors, thereby resembling alterations observed in ASD. We find consistent local activity alterations in ASD to be spatially associated with several major neurotransmitter systems. NMDA-antagonist ketamine induced neurochemical changes similar to ASD-related alterations, supporting the notion that pharmacological modulation of the E/I balance in healthy individuals can induce ASD-like functional brain changes. These findings provide novel insights into neurophysiological mechanisms underlying ASD.
Collapse
Affiliation(s)
- Pascal Grumbach
- Institute of Neurosciences and Medicine, Brain & Behaviour (INM-7), Research Centre Juelich; Wilhelm-Johnen-Straße 1, 52425 Juelich, Germany
- Department of Psychiatry and Psychotherapy, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University Duesseldorf; Bergische Landstraße 2, 40629 Duesseldorf, Germany
| | - Jan Kasper
- Institute of Neurosciences and Medicine, Brain & Behaviour (INM-7), Research Centre Juelich; Wilhelm-Johnen-Straße 1, 52425 Juelich, Germany
- Institute of Systems Neuroscience, Medical Faculty & University Hospital Duesseldorf, Heinrich Heine University Duesseldorf; Moorenstraße 5, 40225 Duesseldorf, Germany
| | - Joerg F. Hipp
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center Basel, F. Hoffmann–La Roche Ltd.; Basel, Switzerland
| | - Anna Forsyth
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland; 85 Park Road, Grafton, Auckland, 1023, New Zealand
| | - Sofie L. Valk
- Institute of Neurosciences and Medicine, Brain & Behaviour (INM-7), Research Centre Juelich; Wilhelm-Johnen-Straße 1, 52425 Juelich, Germany
- Institute of Systems Neuroscience, Medical Faculty & University Hospital Duesseldorf, Heinrich Heine University Duesseldorf; Moorenstraße 5, 40225 Duesseldorf, Germany
- Max Planck School of Cognition; Stephanstraße 1A, 04103 Leipzig, Germany
- Max Planck Institute for Human Cognitive and Brain Sciences; Stephanstraße 1A, 04103 Leipzig, Germany
| | - Suresh Muthukumaraswamy
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland; 85 Park Road, Grafton, Auckland, 1023, New Zealand
| | - Simon B. Eickhoff
- Institute of Neurosciences and Medicine, Brain & Behaviour (INM-7), Research Centre Juelich; Wilhelm-Johnen-Straße 1, 52425 Juelich, Germany
- Institute of Systems Neuroscience, Medical Faculty & University Hospital Duesseldorf, Heinrich Heine University Duesseldorf; Moorenstraße 5, 40225 Duesseldorf, Germany
| | - Leonhard Schilbach
- Department of General Psychiatry 2, LVR-Klinikum Duesseldorf; Bergische Landstraße 2, 40629 Duesseldorf, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig Maximilians University Munich; Nußbaumstraße 7, 80336 München
| | - Juergen Dukart
- Institute of Neurosciences and Medicine, Brain & Behaviour (INM-7), Research Centre Juelich; Wilhelm-Johnen-Straße 1, 52425 Juelich, Germany
- Institute of Systems Neuroscience, Medical Faculty & University Hospital Duesseldorf, Heinrich Heine University Duesseldorf; Moorenstraße 5, 40225 Duesseldorf, Germany
| |
Collapse
|
14
|
Gora C, Dudas A, Vaugrente O, Drobecq L, Pecnard E, Lefort G, Pellissier LP. Deciphering autism heterogeneity: a molecular stratification approach in four mouse models. Transl Psychiatry 2024; 14:416. [PMID: 39366951 PMCID: PMC11452541 DOI: 10.1038/s41398-024-03113-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 10/06/2024] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental condition characterized by impairments in social interaction and communication, as well as restrained or stereotyped behaviors. The inherent heterogeneity within the autism spectrum poses challenges for developing effective pharmacological treatments targeting core features. Successful clinical trials require the identification of robust markers to enable patient stratification. In this study, we identified molecular markers within the oxytocin and immediate early gene families across five interconnected brain structures of the social circuit. We used wild-type and four heterogeneous mouse models, each exhibiting unique autism-like behaviors modeling the autism spectrum. While dysregulations in the oxytocin family were model-specific, immediate early genes displayed widespread alterations, reflecting global changes across the four models. Through integrative analysis, we identified Egr1, Foxp1, Homer1a, Oxt, and Oxtr as five robust and discriminant molecular markers that allowed the successful stratification of the four models. Importantly, our stratification demonstrated predictive values when challenged with a fifth mouse model or identifying subgroups of mice potentially responsive to oxytocin treatment. Beyond providing insights into oxytocin and immediate early gene mRNA dynamics, this proof-of-concept study represents a significant step toward the potential stratification of individuals with ASD. This work has implications for the success of clinical trials and the development of personalized medicine in autism.
Collapse
Affiliation(s)
- Caroline Gora
- INRAE, CNRS, Université de Tours, PRC, 37380, Nouzilly, France
| | - Ana Dudas
- INRAE, CNRS, Université de Tours, PRC, 37380, Nouzilly, France
| | | | - Lucile Drobecq
- INRAE, CNRS, Université de Tours, PRC, 37380, Nouzilly, France
| | | | - Gaëlle Lefort
- INRAE, CNRS, Université de Tours, PRC, 37380, Nouzilly, France
| | | |
Collapse
|
15
|
Seneff S, Kyriakopoulos AM, Nigh G. Is autism a PIN1 deficiency syndrome? A proposed etiological role for glyphosate. J Neurochem 2024; 168:2124-2146. [PMID: 38808598 DOI: 10.1111/jnc.16140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 05/30/2024]
Abstract
Autism is a neurodevelopmental disorder, the prevalence of which has increased dramatically in the United States over the past two decades. It is characterized by stereotyped behaviors and impairments in social interaction and communication. In this paper, we present evidence that autism can be viewed as a PIN1 deficiency syndrome. Peptidyl-prolyl cis/trans isomerase, NIMA-Interacting 1 (PIN1) is a peptidyl-prolyl cis/trans isomerase, and it has widespread influences in biological organisms. Broadly speaking, PIN1 deficiency is linked to many neurodegenerative diseases, whereas PIN1 over-expression is linked to cancer. Death-associated protein kinase 1 (DAPK1) strongly inhibits PIN1, and the hormone melatonin inhibits DAPK1. Melatonin deficiency is strongly linked to autism. It has recently been shown that glyphosate exposure to rats inhibits melatonin synthesis as a result of increased glutamate release from glial cells and increased expression of metabotropic glutamate receptors. Glyphosate's inhibition of melatonin leads to a reduction in PIN1 availability in neurons. In this paper, we show that PIN1 deficiency can explain many of the unique morphological features of autism, including increased dendritic spine density, missing or thin corpus callosum, and reduced bone density. We show how PIN1 deficiency disrupts the functioning of powerful high-level signaling molecules, such as nuclear factor erythroid 2-related factor 2 (NRF2) and p53. Dysregulation of both of these proteins has been linked to autism. Severe depletion of glutathione in the brain resulting from chronic exposure to oxidative stressors and extracellular glutamate leads to oxidation of the cysteine residue in PIN1, inactivating the protein and further contributing to PIN1 deficiency. Impaired autophagy leads to increased sensitivity of neurons to ferroptosis. It is imperative that further research be conducted to experimentally validate whether the mechanisms described here take place in response to chronic glyphosate exposure and whether this ultimately leads to autism.
Collapse
Affiliation(s)
- Stephanie Seneff
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | | | - Greg Nigh
- Immersion Health, Portland, Oregon, USA
| |
Collapse
|
16
|
Falcão M, Monteiro P, Jacinto L. Tactile sensory processing deficits in genetic mouse models of autism spectrum disorder. J Neurochem 2024; 168:2105-2123. [PMID: 38837765 DOI: 10.1111/jnc.16135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 05/04/2024] [Accepted: 05/07/2024] [Indexed: 06/07/2024]
Abstract
Altered sensory processing is a common feature in autism spectrum disorder (ASD), as recognized in the Diagnostic and Statistical Manual of Mental Disorders (DSM-5). Although altered responses to tactile stimuli are observed in over 60% of individuals with ASD, the neurobiological basis of this phenomenon is poorly understood. ASD has a strong genetic component and genetic mouse models can provide valuable insights into the mechanisms underlying tactile abnormalities in ASD. This review critically addresses recent findings regarding tactile processing deficits found in mouse models of ASD, with a focus on behavioral, anatomical, and functional alterations. Particular attention was given to cellular and circuit-level functional alterations, both in the peripheral and central nervous systems, with the objective of highlighting possible convergence mechanisms across models. By elucidating the impact of mutations in ASD candidate genes on somatosensory circuits and correlating them with behavioral phenotypes, this review significantly advances our understanding of tactile deficits in ASD. Such insights not only broaden our comprehension but also pave the way for future therapeutic interventions.
Collapse
Affiliation(s)
- Margarida Falcão
- Department of Biomedicine-Experimental Biology Unit, Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal
| | - Patricia Monteiro
- Department of Biomedicine-Experimental Biology Unit, Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal
| | - Luis Jacinto
- Department of Biomedicine-Experimental Biology Unit, Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal
| |
Collapse
|
17
|
Ricarte M, Tagkalidou N, Bellot M, Bedrossiantz J, Prats E, Gomez-Canela C, Garcia-Reyero N, Raldúa D. Short- and Long-Term Neurobehavioral Effects of Developmental Exposure to Valproic Acid in Zebrafish. Int J Mol Sci 2024; 25:7688. [PMID: 39062930 PMCID: PMC11277053 DOI: 10.3390/ijms25147688] [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: 06/20/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by impairments in social interaction and communication, anxiety, hyperactivity, and interest restricted to specific subjects. In addition to the genetic factors, multiple environmental factors have been related to the development of ASD. Animal models can serve as crucial tools for understanding the complexity of ASD. In this study, a chemical model of ASD has been developed in zebrafish by exposing embryos to valproic acid (VPA) from 4 to 48 h post-fertilization, rearing them to the adult stage in fish water. For the first time, an integrative approach combining behavioral analysis and neurotransmitters profile has been used for determining the effects of early-life exposure to VPA both in the larval and adult stages. Larvae from VPA-treated embryos showed hyperactivity and decreased visual and vibrational escape responses, as well as an altered neurotransmitters profile, with increased glutamate and decreased acetylcholine and norepinephrine levels. Adults from VPA-treated embryos exhibited impaired social behavior characterized by larger shoal sizes and a decreased interest for their conspecifics. A neurotransmitter analysis revealed a significant decrease in dopamine and GABA levels in the brain. These results support the potential predictive validity of this model for ASD research.
Collapse
Affiliation(s)
- Marina Ricarte
- Institute for Environmental Assessment and Water Research (IDAEA-CSIC), 08034 Barcelona, Spain; (M.R.); (N.T.); (J.B.)
- Department of Analytical and Applied Chemistry, School of Engineering, Institut Químic de Sarrià, Universitat Ramon Llull, 08017 Barcelona, Spain; (M.B.); (C.G.-C.)
| | - Niki Tagkalidou
- Institute for Environmental Assessment and Water Research (IDAEA-CSIC), 08034 Barcelona, Spain; (M.R.); (N.T.); (J.B.)
| | - Marina Bellot
- Department of Analytical and Applied Chemistry, School of Engineering, Institut Químic de Sarrià, Universitat Ramon Llull, 08017 Barcelona, Spain; (M.B.); (C.G.-C.)
| | - Juliette Bedrossiantz
- Institute for Environmental Assessment and Water Research (IDAEA-CSIC), 08034 Barcelona, Spain; (M.R.); (N.T.); (J.B.)
| | - Eva Prats
- Research and Development Center (CID-CSIC), 08034 Barcelona, Spain;
| | - Cristian Gomez-Canela
- Department of Analytical and Applied Chemistry, School of Engineering, Institut Químic de Sarrià, Universitat Ramon Llull, 08017 Barcelona, Spain; (M.B.); (C.G.-C.)
| | - Natalia Garcia-Reyero
- Institute for Genomics, Biocomputing & Biotechnology (IGBB), Mississippi State University, Starkville, MS 39762, USA;
| | - Demetrio Raldúa
- Institute for Environmental Assessment and Water Research (IDAEA-CSIC), 08034 Barcelona, Spain; (M.R.); (N.T.); (J.B.)
| |
Collapse
|
18
|
Garofalo M, De Simone G, Motta Z, Nuzzo T, De Grandis E, Bruno C, Boeri S, Riccio MP, Pastore L, Bravaccio C, Iasevoli F, Salvatore F, Pollegioni L, Errico F, de Bartolomeis A, Usiello A. Decreased free D-aspartate levels in the blood serum of patients with schizophrenia. Front Psychiatry 2024; 15:1408175. [PMID: 39050919 PMCID: PMC11266155 DOI: 10.3389/fpsyt.2024.1408175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 06/17/2024] [Indexed: 07/27/2024] Open
Abstract
Introduction Schizophrenia (SCZ) and autism spectrum disorder (ASD) are neurodevelopmental diseases characterized by different psychopathological manifestations and divergent clinical trajectories. Various alterations at glutamatergic synapses have been reported in both disorders, including abnormal NMDA and metabotropic receptor signaling. Methods We conducted a bicentric study to assess the blood serum levels of NMDA receptors-related glutamatergic amino acids and their precursors, including L-glutamate, L-glutamine, D-aspartate, L-aspartate, L-asparagine, D-serine, L-serine and glycine, in ASD, SCZ patients and their respective control subjects. Specifically, the SCZ patients were subdivided into treatment-resistant and non-treatment-resistant SCZ patients, based on their responsivity to conventional antipsychotics. Results D-serine and D-aspartate serum reductions were found in SCZ patients compared to controls. Conversely, no significant differences between cases and controls were found in amino acid concentrations in the two ASD cohorts analyzed. Discussion This result further encourages future research to evaluate the predictive role of selected D-amino acids as peripheral markers for SCZ pathophysiology and diagnosis.
Collapse
Affiliation(s)
- Martina Garofalo
- CEINGE Biotecnologie Avanzate “Franco Salvatore”, Naples, Italy
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Università degli Studi della Campania “Luigi Vanvitelli”, Caserta, Italy
| | - Giuseppe De Simone
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Odontostomatology, University Medical School of Naples “Federico II”, Naples, Italy
| | - Zoraide Motta
- ”The Protein Factory 2.0”, Dipartimento di Biotecnologie e Scienze della Vita, Università degli Studi dell’Insubria, Varese, Italy
| | - Tommaso Nuzzo
- CEINGE Biotecnologie Avanzate “Franco Salvatore”, Naples, Italy
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Università degli Studi della Campania “Luigi Vanvitelli”, Caserta, Italy
| | - Elisa De Grandis
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal, and Child Health - DINOGMI, University of Genoa, Genoa, Italy
| | - Claudio Bruno
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal, and Child Health - DINOGMI, University of Genoa, Genoa, Italy
- Center of Translational and Experimental Myology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genoa, Italy
| | - Silvia Boeri
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal, and Child Health - DINOGMI, University of Genoa, Genoa, Italy
| | - Maria Pia Riccio
- Department of Maternal and Child Health, Unità Operativa semplice di Dipartimento (UOSD) of Child and Adolescent Psychiatry, Azienda Ospedaliera Universitaria (AOU) Federico II, Naples, Italy
| | - Lucio Pastore
- CEINGE Biotecnologie Avanzate “Franco Salvatore”, Naples, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, Naples, Italy
| | - Carmela Bravaccio
- Department of Medical and Translational Sciences, Child Neuropsychiatry, Federico II University, Napoli, Italy
| | - Felice Iasevoli
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Odontostomatology, University Medical School of Naples “Federico II”, Naples, Italy
| | - Francesco Salvatore
- CEINGE Biotecnologie Avanzate “Franco Salvatore”, Naples, Italy
- Centro Interuniversitario per Malattie Multigeniche e Multifattoriali e loro Modelli Animali (Federico II, Naples; Tor Vergata, Rome and “G. D’Annunzio”, Chieti-Pescara), Naples, Italy
| | - Loredano Pollegioni
- ”The Protein Factory 2.0”, Dipartimento di Biotecnologie e Scienze della Vita, Università degli Studi dell’Insubria, Varese, Italy
| | - Francesco Errico
- CEINGE Biotecnologie Avanzate “Franco Salvatore”, Naples, Italy
- Dipartimento di Agraria, Università degli Studi di Napoli “Federico II”, Portici, Italy
| | - Andrea de Bartolomeis
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Odontostomatology, University Medical School of Naples “Federico II”, Naples, Italy
| | - Alessandro Usiello
- CEINGE Biotecnologie Avanzate “Franco Salvatore”, Naples, Italy
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Università degli Studi della Campania “Luigi Vanvitelli”, Caserta, Italy
| |
Collapse
|
19
|
Nicosia N, Giovenzana M, Misztak P, Mingardi J, Musazzi L. Glutamate-Mediated Excitotoxicity in the Pathogenesis and Treatment of Neurodevelopmental and Adult Mental Disorders. Int J Mol Sci 2024; 25:6521. [PMID: 38928227 PMCID: PMC11203689 DOI: 10.3390/ijms25126521] [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: 05/17/2024] [Revised: 06/09/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Glutamate is the main excitatory neurotransmitter in the brain wherein it controls cognitive functional domains and mood. Indeed, brain areas involved in memory formation and consolidation as well as in fear and emotional processing, such as the hippocampus, prefrontal cortex, and amygdala, are predominantly glutamatergic. To ensure the physiological activity of the brain, glutamatergic transmission is finely tuned at synaptic sites. Disruption of the mechanisms responsible for glutamate homeostasis may result in the accumulation of excessive glutamate levels, which in turn leads to increased calcium levels, mitochondrial abnormalities, oxidative stress, and eventually cell atrophy and death. This condition is known as glutamate-induced excitotoxicity and is considered as a pathogenic mechanism in several diseases of the central nervous system, including neurodevelopmental, substance abuse, and psychiatric disorders. On the other hand, these disorders share neuroplasticity impairments in glutamatergic brain areas, which are accompanied by structural remodeling of glutamatergic neurons. In the current narrative review, we will summarize the role of glutamate-induced excitotoxicity in both the pathophysiology and therapeutic interventions of neurodevelopmental and adult mental diseases with a focus on autism spectrum disorders, substance abuse, and psychiatric disorders. Indeed, glutamatergic drugs are under preclinical and clinical development for the treatment of different mental diseases that share glutamatergic neuroplasticity dysfunctions. Although clinical evidence is still limited and more studies are required, the regulation of glutamate homeostasis is attracting attention as a potential crucial target for the control of brain diseases.
Collapse
Affiliation(s)
- Noemi Nicosia
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (N.N.); (M.G.); (P.M.)
- PhD Program in Neuroscience, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Mattia Giovenzana
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (N.N.); (M.G.); (P.M.)
- PhD Program in Neuroscience, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Paulina Misztak
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (N.N.); (M.G.); (P.M.)
| | - Jessica Mingardi
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (N.N.); (M.G.); (P.M.)
| | - Laura Musazzi
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (N.N.); (M.G.); (P.M.)
- Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| |
Collapse
|
20
|
Mahmoud AH, Elhefnawei DM, EL-Desouky MA, Kadry MO. Reciprocal crosslink among MeCP2/BDNF /CREB signaling pinpointed in autism spectrum disorder. Toxicol Rep 2024; 12:91-99. [PMID: 38229920 PMCID: PMC10789594 DOI: 10.1016/j.toxrep.2023.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 12/01/2023] [Accepted: 12/22/2023] [Indexed: 01/18/2024] Open
Abstract
Autism spectrum disorder, or individual disability (ID), is a condition characterized by complications in social interaction, restricted repetitive behavior, and difficulties in social communication. Neuquinon (NQ) possess a powerful therapeutic potential in various neurodegenerative disease. Nevertheless, contributing to NQ's low water solubility and bioavailability, its medicinal use has been constrained. Liposomes were supposed to be prospective drug-delivering agents for NQ, crossing the blood-brain barrier (BBB), and reaching the target organs. The current investigation aims to track the signaling pathways that govern NQ and liposomal neuquinon (LNQ) action in autistic models generated by ethyl formic acid. The neurotransmitters gamma amino-butyric acid (GABA), acetylcholine (ACh), and acetylcholinesterase (AChE) in addition to, the gene expressions of brain-derived neurotrophic factor (BDNF), cAMP response element-binding protein (CREB), and methyl-CpG-binding protein 2 (MeCP2) and the DNA damage COMET analysis at different time intervals of the study, were assessed. EFA in a dose of 500 mg/kg BW was used to induce autism in rats, and then NQ and LNQ were administered in 10 mg/kg and 2 mg/kg BW, respectively. The results revealed that NQ and LNQ significantly down-regulated BDNF, GABA, and AChE; on the other hand, they up-regulated MeCP2, CREB gene expressions, and ACh action. NQ and LNQ displayed improvement in DNA damage in almost all brain regions after EFA alterations; even better results were noticed post-LNQ therapy. Therefore, it may be concluded that neuquinon and liposomal-loaded neuquinon have a therapeutic index versus EFA-induced autism in a rat model.
Collapse
Affiliation(s)
- Ahlam H. Mahmoud
- Department of Therapeutic Chemistry, National Research Centre, Dokki, Giza 12622, Egypt
| | - Doaa M. Elhefnawei
- Department of Therapeutic Chemistry, National Research Centre, Dokki, Giza 12622, Egypt
| | | | - Mai O. Kadry
- Department of Therapeutic Chemistry, National Research Centre, Dokki, Giza 12622, Egypt
| |
Collapse
|
21
|
Tran H, Le L, Singh BN, Kramer J, Steward R. Tet controls axon guidance in early brain development through glutamatergic signaling. iScience 2024; 27:109634. [PMID: 38655199 PMCID: PMC11035372 DOI: 10.1016/j.isci.2024.109634] [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: 08/14/2023] [Revised: 12/18/2023] [Accepted: 03/26/2024] [Indexed: 04/26/2024] Open
Abstract
Mutations in ten-eleven translocation (TET) proteins are associated with human neurodevelopmental disorders. We find a function of Tet in regulating Drosophila early brain development. The Tet DNA-binding domain (TetAXXC) is required for axon guidance in the mushroom body (MB). Glutamine synthetase 2 (Gs2), a key enzyme in glutamatergic signaling, is significantly down-regulated in the TetAXXC brains. Loss of Gs2 recapitulates the TetAXXC phenotype. Surprisingly, Tet and Gs2 act in the insulin-producing cells (IPCs) to control MB axon guidance, and overexpression of Gs2 in IPCs rescues the defects of TetAXXC. Feeding TetAXXC with metabotropic glutamate receptor antagonist MPEP rescues the phenotype while glutamate enhances it. Mutants in Tet and Drosophila Fmr1, the homolog of human FMR1, have similar defects, and overexpression of Gs2 in IPCs also rescues the Fmr1 phenotype. We provide the first evidence that Tet controls the guidance of developing brain axons by modulating glutamatergic signaling.
Collapse
Affiliation(s)
- Hiep Tran
- Waksman Institute, Rutgers University, Piscataway, NJ 08854, USA
| | - Le Le
- Waksman Institute, Rutgers University, Piscataway, NJ 08854, USA
| | - Badri Nath Singh
- Waksman Institute, Rutgers University, Piscataway, NJ 08854, USA
| | - Joseph Kramer
- Department of Pathology and Laboratory Medicine, Rutgers Biomedical and Health Sciences, Rutgers University, New Brunswick, NJ 08901, USA
| | - Ruth Steward
- Waksman Institute, Rutgers University, Piscataway, NJ 08854, USA
- Department of Molecular Biology and Biochemistry, Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ 08901, USA
| |
Collapse
|
22
|
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
|
23
|
Vakilzadeh G, Maseko BC, Bartely TD, McLennan YA, Martínez-Cerdeño V. Increased number of excitatory synapsis and decreased number of inhibitory synapsis in the prefrontal cortex in autism. Cereb Cortex 2024; 34:121-128. [PMID: 38696601 PMCID: PMC11065106 DOI: 10.1093/cercor/bhad268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/15/2023] [Accepted: 07/16/2023] [Indexed: 05/04/2024] Open
Abstract
Previous studies in autism spectrum disorder demonstrated an increased number of excitatory pyramidal cells and a decreased number of inhibitory parvalbumin+ chandelier interneurons in the prefrontal cortex of postmortem brains. How these changes in cellular composition affect the overall abundance of excitatory and inhibitory synapses in the cortex is not known. Herein, we quantified the number of excitatory and inhibitory synapses in the prefrontal cortex of 10 postmortem autism spectrum disorder brains and 10 control cases. To identify excitatory synapses, we used VGlut1 as a marker of the presynaptic component and postsynaptic density protein-95 as marker of the postsynaptic component. To identify inhibitory synapses, we used the vesicular gamma-aminobutyric acid transporter as a marker of the presynaptic component and gephyrin as a marker of the postsynaptic component. We used Puncta Analyzer to quantify the number of co-localized pre- and postsynaptic synaptic components in each area of interest. We found an increase in the number of excitatory synapses in upper cortical layers and a decrease in inhibitory synapses in all cortical layers in autism spectrum disorder brains compared with control cases. The alteration in the number of excitatory and inhibitory synapses could lead to neuronal dysfunction and disturbed network connectivity in the prefrontal cortex in autism spectrum disorder.
Collapse
Affiliation(s)
- Gelareh Vakilzadeh
- Department of Pathology and Laboratory Medicine, University of California, Davis School of Medicine, Sacramento, CA, United States
- Institute for Pediatric Regenerative Medicine and Shriners Hospitals for Children, Sacramento, CA, United States
| | - Busisiwe C Maseko
- Faculty of health Sciences, School of Anatomical Sciences, University of the Witwatersrand, Johannesburg, The Republic of South Africa
| | - Trevor D Bartely
- Department of Pathology and Laboratory Medicine, University of California, Davis School of Medicine, Sacramento, CA, United States
- Institute for Pediatric Regenerative Medicine and Shriners Hospitals for Children, Sacramento, CA, United States
| | - Yingratana A McLennan
- Department of Pathology and Laboratory Medicine, University of California, Davis School of Medicine, Sacramento, CA, United States
- Institute for Pediatric Regenerative Medicine and Shriners Hospitals for Children, Sacramento, CA, United States
| | - Verónica Martínez-Cerdeño
- Department of Pathology and Laboratory Medicine, University of California, Davis School of Medicine, Sacramento, CA, United States
- Institute for Pediatric Regenerative Medicine and Shriners Hospitals for Children, Sacramento, CA, United States
- MIND Institute, UC Davis School of Medicine, Sacramento, CA, United States
| |
Collapse
|
24
|
González-Madrid E, Rangel-Ramírez MA, Opazo MC, Méndez L, Bohmwald K, Bueno SM, González PA, Kalergis AM, Riedel CA. Gestational hypothyroxinemia induces ASD-like phenotypes in behavior, proinflammatory markers, and glutamatergic protein expression in mouse offspring of both sexes. Front Endocrinol (Lausanne) 2024; 15:1381180. [PMID: 38752179 PMCID: PMC11094302 DOI: 10.3389/fendo.2024.1381180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 04/15/2024] [Indexed: 05/18/2024] Open
Abstract
Background The prevalence of autism spectrum disorder (ASD) has significantly risen in the past three decades, prompting researchers to explore the potential contributions of environmental factors during pregnancy to ASD development. One such factor of interest is gestational hypothyroxinemia (HTX), a frequent condition in pregnancy associated with cognitive impairments in the offspring. While retrospective human studies have linked gestational HTX to autistic traits, the cellular and molecular mechanisms underlying the development of ASD-like phenotypes remain poorly understood. This study used a mouse model of gestational HTX to evaluate ASD-like phenotypes in the offspring. Methods To induce gestational HTX, pregnant mice were treated with 2-mercapto-1-methylimidazole (MMI), a thyroid hormones synthesis inhibitor, in the tap-drinking water from embryonic days (E) 10 to E14. A separate group received MMI along with a daily subcutaneous injection of T4, while the control group received regular tap water during the entire pregnancy. Female and male offspring underwent assessments for repetitive, anxious, and social behaviors from postnatal day (P) 55 to P64. On P65, mice were euthanized for the evaluation of ASD-related inflammatory markers in blood, spleen, and specific brain regions. Additionally, the expression of glutamatergic proteins (NLGN3 and HOMER1) was analyzed in the prefrontal cortex and hippocampus. Results The HTX-offspring exhibited anxious-like behavior, a subordinate state, and impaired social interactions. Subsequently, both female and male HTX-offspring displayed elevated proinflammatory cytokines in blood, including IL-1β, IL-6, IL-17A, and TNF-α, while only males showed reduced levels of IL-10. The spleen of HTX-offspring of both sexes showed increased Th17/Treg ratio and M1-like macrophages. In the prefrontal cortex and hippocampus of male HTX-offspring, elevated levels of IL-17A and reduced IL-10 were observed, accompanied by increased expression of hippocampal NLGN3 and HOMER1. All these observations were compared to those observed in the Control-offspring. Notably, the supplementation with T4 during the MMI treatment prevents the development of the observed phenotypes. Correlation analysis revealed an association between maternal T4 levels and specific ASD-like outcomes. Discussion This study validates human observations, demonstrating for the first time that gestational HTX induces ASD-like phenotypes in the offspring, highlighting the need of monitoring thyroid function during pregnancy.
Collapse
Affiliation(s)
- Enrique González-Madrid
- Laboratorio de Endocrino-inmunología, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ma. Andreina Rangel-Ramírez
- Laboratorio de Endocrino-inmunología, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - María C. Opazo
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Facultad de Medicina Veterinaria y Agronomía, Instituto de Ciencias Naturales, Universidad de las Américas, Santiago, Chile
| | - Luis Méndez
- Laboratorio de Endocrino-inmunología, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Karen Bohmwald
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - Susan M. Bueno
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A. González
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia A. Riedel
- Laboratorio de Endocrino-inmunología, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
25
|
Vilela J, Rasga C, Santos JX, Martiniano H, Marques AR, Oliveira G, Vicente AM. Bridging Genetic Insights with Neuroimaging in Autism Spectrum Disorder-A Systematic Review. Int J Mol Sci 2024; 25:4938. [PMID: 38732157 PMCID: PMC11084239 DOI: 10.3390/ijms25094938] [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: 02/29/2024] [Revised: 04/22/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Autism Spectrum Disorder (ASD) is an early onset neurodevelopmental disorder characterized by impaired social interaction and communication, and repetitive patterns of behavior. Family studies show that ASD is highly heritable, and hundreds of genes have previously been implicated in the disorder; however, the etiology is still not fully clear. Brain imaging and electroencephalography (EEG) are key techniques that study alterations in brain structure and function. Combined with genetic analysis, these techniques have the potential to help in the clarification of the neurobiological mechanisms contributing to ASD and help in defining novel therapeutic targets. To further understand what is known today regarding the impact of genetic variants in the brain alterations observed in individuals with ASD, a systematic review was carried out using Pubmed and EBSCO databases and following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. This review shows that specific genetic variants and altered patterns of gene expression in individuals with ASD may have an effect on brain circuits associated with face processing and social cognition, and contribute to excitation-inhibition imbalances and to anomalies in brain volumes.
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.); (C.R.); (J.X.S.); (H.M.); (A.R.M.)
- BioISI-Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Campo Grande, C8, 1749-016 Lisboa, Portugal
| | - 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.); (C.R.); (J.X.S.); (H.M.); (A.R.M.)
- 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.); (C.R.); (J.X.S.); (H.M.); (A.R.M.)
- 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.); (C.R.); (J.X.S.); (H.M.); (A.R.M.)
- 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.); (C.R.); (J.X.S.); (H.M.); (A.R.M.)
- 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.); (C.R.); (J.X.S.); (H.M.); (A.R.M.)
- BioISI-Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Campo Grande, C8, 1749-016 Lisboa, Portugal
| |
Collapse
|
26
|
Fu Z, Yang X, Jiang Y, Mao X, Liu H, Yang Y, Chen J, Chen Z, Li H, Zhang XS, Mao X, Li N, Wang D, Jiang J. Microbiota profiling reveals alteration of gut microbial neurotransmitters in a mouse model of autism-associated 16p11.2 microduplication. Front Microbiol 2024; 15:1331130. [PMID: 38596370 PMCID: PMC11002229 DOI: 10.3389/fmicb.2024.1331130] [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: 11/29/2023] [Accepted: 02/27/2024] [Indexed: 04/11/2024] Open
Abstract
The gut-brain axis is evident in modulating neuropsychiatric diseases including autism spectrum disorder (ASD). Chromosomal 16p11.2 microduplication 16p11.2dp/+ is among the most prevalent genetic copy number variations (CNV) linked with ASD. However, the implications of gut microbiota status underlying the development of ASD-like impairments induced by 16p11.2dp/+ remains unclear. To address this, we initially investigated a mouse model of 16p11.2dp/+, which exhibits social novelty deficit and repetitive behavior characteristic of ASD. Subsequently, we conducted a comparative analysis of the gut microbial community and metabolomic profiles between 16p11.2dp/+ and their wild-type counterparts using 16S rRNA sequencing and liquid chromatography-mass spectrometry (LC/MS). Our microbiota analysis revealed structural dysbiosis in 16p11.2dp/+ mice, characterized by reduced biodiversity and alterations in species abundance, as indicated by α/β-diversity analysis. Specifically, we observed reduced relative abundances of Faecalibaculum and Romboutsia, accompanied by an increase in Turicibacter and Prevotellaceae UCG_001 in 16p11.2dp/+ group. Metabolomic analysis identified 19 significantly altered metabolites and unveiled enriched amino acid metabolism pathways. Notably, a disruption in the predominantly histamine-centered neurotransmitter network was observed in 16p11.2dp/+ mice. Collectively, our findings delineate potential alterations and correlations among the gut microbiota and microbial neurotransmitters in 16p11.2dp/+ mice, providing new insights into the pathogenesis of and treatment for 16p11.2 CNV-associated ASD.
Collapse
Affiliation(s)
- Zhang Fu
- Tomas Lindhal Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Xiuyan Yang
- Tomas Lindhal Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Youheng Jiang
- Tomas Lindhal Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Xinliang Mao
- Guangdong Perfect Life Health Science and Technology Research Institute Co., Ltd., Zhongshan, Guangdong, China
| | - Hualin Liu
- Guangdong Perfect Life Health Science and Technology Research Institute Co., Ltd., Zhongshan, Guangdong, China
| | - Yanming Yang
- Tomas Lindhal Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Jia Chen
- Tomas Lindhal Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Zhumei Chen
- Tomas Lindhal Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
- Department of Anesthesiology, The Seventh Affiliated Hospital of Sun Yat-Sen University (SYSU), Shenzhen, Guangdong, China
| | - Huiliang Li
- Division of Medicine, Wolfson Institute for Biomedical Research, Faculty of Medical Sciences, University College London, London, United Kingdom
- China-UK Institute for Frontier Science, Shenzhen, Guangdong, China
| | - Xue-Song Zhang
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, United States
| | - Xinjun Mao
- Department of Anesthesiology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Ningning Li
- Tomas Lindhal Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
- China-UK Institute for Frontier Science, Shenzhen, Guangdong, China
| | - Dilong Wang
- Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jian Jiang
- Tomas Lindhal Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| |
Collapse
|
27
|
Oberman LM, Francis SM, Beynel L, Hynd M, Jaime M, Robins PL, Deng ZD, Stout J, van der Veen JW, Lisanby SH. Design and methodology for a proof of mechanism study of individualized neuronavigated continuous Theta burst stimulation for auditory processing in adolescents with autism spectrum disorder. Front Psychiatry 2024; 15:1304528. [PMID: 38389984 PMCID: PMC10881663 DOI: 10.3389/fpsyt.2024.1304528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 01/24/2024] [Indexed: 02/24/2024] Open
Abstract
It has been suggested that aberrant excitation/inhibition (E/I) balance and dysfunctional structure and function of relevant brain networks may underlie the symptoms of autism spectrum disorder (ASD). However, the nomological network linking these constructs to quantifiable measures and mechanistically relating these constructs to behavioral symptoms of ASD is lacking. Herein we describe a within-subject, controlled, proof-of-mechanism study investigating the pathophysiology of auditory/language processing in adolescents with ASD. We utilize neurophysiological and neuroimaging techniques including magnetic resonance spectroscopy (MRS), diffusion-weighted imaging (DWI), functional magnetic resonance imaging (fMRI), and magnetoencephalography (MEG) metrics of language network structure and function. Additionally, we apply a single, individually targeted session of continuous theta burst stimulation (cTBS) as an experimental probe of the impact of perturbation of the system on these neurophysiological and neuroimaging outcomes. MRS, fMRI, and MEG measures are evaluated at baseline and immediately prior to and following cTBS over the posterior superior temporal cortex (pSTC), a region involved in auditory and language processing deficits in ASD. Also, behavioral measures of ASD and language processing and DWI measures of auditory/language network structures are obtained at baseline to characterize the relationship between the neuroimaging and neurophysiological measures and baseline symptom presentation. We hypothesize that local gamma-aminobutyric acid (GABA) and glutamate concentrations (measured with MRS), and structural and functional activity and network connectivity (measured with DWI and fMRI), will significantly predict MEG indices of auditory/language processing and behavioral deficits in ASD. Furthermore, a single session of cTBS over left pSTC is hypothesized to lead to significant, acute changes in local glutamate and GABA concentration, functional activity and network connectivity, and MEG indices of auditory/language processing. We have completed the pilot phase of the study (n=20 Healthy Volunteer adults) and have begun enrollment for the main phase with adolescents with ASD (n=86; age 14-17). If successful, this study will establish a nomological network linking local E/I balance measures to functional and structural connectivity within relevant brain networks, ultimately connecting them to ASD symptoms. Furthermore, this study will inform future therapeutic trials using cTBS to treat the symptoms of ASD.
Collapse
Affiliation(s)
- Lindsay M Oberman
- Noninvasive Neuromodulation Unit, Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Sunday M Francis
- Noninvasive Neuromodulation Unit, Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Lysianne Beynel
- Noninvasive Neuromodulation Unit, Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Megan Hynd
- Clinical Affective Neuroscience Laboratory, Department of Psychology & Neuroscience, University of North Carolina, Chapel Hill, NC, United States
| | - Miguel Jaime
- Noninvasive Neuromodulation Unit, Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Pei L Robins
- Noninvasive Neuromodulation Unit, Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Zhi-De Deng
- Noninvasive Neuromodulation Unit, Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Jeff Stout
- Magnetoencephalography Core, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Jan Willem van der Veen
- Magnetic Resonance Spectroscopy Core, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Sarah H Lisanby
- Noninvasive Neuromodulation Unit, Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
28
|
Mhanna A, Martini N, Hmaydoosh G, Hamwi G, Jarjanazi M, Zaifah G, Kazzazo R, Haji Mohamad A, Alshehabi Z. The correlation between gut microbiota and both neurotransmitters and mental disorders: A narrative review. Medicine (Baltimore) 2024; 103:e37114. [PMID: 38306525 PMCID: PMC10843545 DOI: 10.1097/md.0000000000037114] [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: 11/07/2023] [Accepted: 01/09/2024] [Indexed: 02/04/2024] Open
Abstract
The gastrointestinal tract is embedded with microorganisms of numerous genera, referred to as gut microbiota. Gut microbiota has multiple effects on many body organs, including the brain. There is a bidirectional connection between the gut and brain called the gut-brain-axis, and these connections are formed through immunological, neuronal, and neuroendocrine pathways. In addition, gut microbiota modulates the synthesis and functioning of neurotransmitters. Therefore, the disruption of the gut microbiota in the composition or function, which is known as dysbiosis, is associated with the pathogenesis of many mental disorders, such as schizophrenia, depression, and other psychiatric disorders. This review aims to summarize the modulation role of the gut microbiota in 4 prominent neurotransmitters (tryptophan and serotonergic system, dopamine, gamma-aminobutyric acid, and glutamate), as well as its association with 4 psychiatric disorders (schizophrenia, depression, anxiety disorders, and autism spectrum disorder). More future research is required to develop efficient gut-microbiota-based therapies for these illnesses.
Collapse
Affiliation(s)
- Amjad Mhanna
- Faculty of Medicine, Tishreen University, Latakia, Syrian Arab Republic
- Stemosis for Scientific Research, Damascus, Syrian Arab Republic
| | - Nafiza Martini
- Stemosis for Scientific Research, Damascus, Syrian Arab Republic
- Damascus University, Faculty of Medicine, Damascus, Syrian Arab Republic
| | - Ghefar Hmaydoosh
- Faculty of Medicine, Tishreen University, Latakia, Syrian Arab Republic
- Stemosis for Scientific Research, Damascus, Syrian Arab Republic
| | - George Hamwi
- Faculty of Medicine, Tishreen University, Latakia, Syrian Arab Republic
- Stemosis for Scientific Research, Damascus, Syrian Arab Republic
| | - Mulham Jarjanazi
- Pediatric Surgery Resident, Pediatric Surgery Department, Aleppo University Hospital, Aleppo, Syrian Arab Republic
| | - Ghaith Zaifah
- Faculty of Medicine, Tishreen University, Latakia, Syrian Arab Republic
- Stemosis for Scientific Research, Damascus, Syrian Arab Republic
| | - Reem Kazzazo
- Faculty of Medicine, Tishreen University, Latakia, Syrian Arab Republic
- Stemosis for Scientific Research, Damascus, Syrian Arab Republic
| | - Aya Haji Mohamad
- Stemosis for Scientific Research, Damascus, Syrian Arab Republic
- Faculty of Medicine, Aleppo University, Aleppo University Hospital, Aleppo, Syrian Arab Republic
| | - Zuheir Alshehabi
- Department of Pathology, Tishreen University Hospital, Latakia, Syrian Arab Republic
| |
Collapse
|
29
|
Tripathi MK, Ojha SK, Kartawy M, Khaliulin I, Hamoudi W, Amal H. Mutations associated with autism lead to similar synaptic and behavioral alterations in both sexes of male and female mouse brain. Sci Rep 2024; 14:10. [PMID: 38177238 PMCID: PMC10766975 DOI: 10.1038/s41598-023-50248-4] [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: 09/10/2023] [Accepted: 12/17/2023] [Indexed: 01/06/2024] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder based on synaptic abnormalities. The estimated prevalence rate of male individuals diagnosed with ASD prevails over females is in a proportion of 4:1. Consequently, males remain the main focus in ASD studies in clinical and experimental settings. Meanwhile, some studies point to an underestimation of this disorder in females. In this work, we studied the sex differences of the synaptic and behavioral phenotypes of ASD mouse models. Juvenile male and female Shank3Δ4-22 and Cntnap2-/- mutant mice and their WT littermates were used in the experiments. The animals were subjected to a Three-Chamber Sociability Test, then euthanized, and the whole cortex was used for the evaluation of the synaptic phenotype. Protein levels of glutamatergic (NR1) and GABAergic (GAD1 and VGAT) neuronal markers were measured. Protein level of synaptophysin (Syp) was also measured. Dendritic spine density in somatosensory neurons was analyzed by Golgi staining methods. Spine Density and GAD1, NR1, VGAT, and Syp levels were significantly reduced in Shank3Δ4-22 and Cntnap2-/- mice compared to the control group irrespective of sex, indicating impaired synaptic development in the mutant mice. These results were consistent with the lack of differences in the three-chamber sociability test between male and female mice. In conclusion, female ASD mice of both mutations undergo similar synaptic aberrations as their male counterparts and need to be studied along with the male animals. Finally, this work urges the psychiatry scientific community to use both sexes in their investigations.
Collapse
Affiliation(s)
- Manish Kumar Tripathi
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Shashank Kumar Ojha
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Maryam Kartawy
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Igor Khaliulin
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Wajeha Hamoudi
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Haitham Amal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
30
|
Li Y, Hu W, Lin B, Ma T, Zhang Z, Hu W, Zhou R, Kwok LY, Sun Z, Zhu C, Zhang H. Omic characterizing and targeting gut dysbiosis in children with autism spectrum disorder: symptom alleviation through combined probiotic and medium-carbohydrate diet intervention - a pilot study. Gut Microbes 2024; 16:2434675. [PMID: 39632378 PMCID: PMC11622613 DOI: 10.1080/19490976.2024.2434675] [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: 08/27/2024] [Revised: 10/22/2024] [Accepted: 11/11/2024] [Indexed: 12/07/2024] Open
Abstract
Autism spectrum disorder (ASD) currently lacks effective diagnostic and therapeutic approaches. Disruptions in the gut ecosystem have been observed in individuals with ASD, suggesting that targeting gut microbiota through probiotic and dietary supplementation may serve as a potential treatment strategy. This two-phase study aimed to characterize the fecal metagenome of children with ASD and investigate the beneficial effects of a combined probiotic and medium-carbohydrate intervention in ASD. Fecal metagenomes of children with ASD were compared to those of typically developing children, revealing intestinal dysbiosis in ASD, characterized by reduced levels of Prevotella sp. Dialister invisus, and Bacteroides sp. along with increased predicted abundances of inosine, glutamate, xanthine, and methylxanthine. The gut bacteriome and phageome exhibited high cooperativity. In a 3-month pilot study, Bifidobacterium animalis subsp. lactis Probio-M8 (Probio-M8) was administered alongside a medium-carbohydrate diet to Chinese children with ASD. The primary endpoint was the Childhood Autism Rating Scale (CARS), while the secondary endpoint was the Gastrointestinal Symptom Rating Scale (GSRS). A total of 72 autistic children were initially recruited for the intervention study, but only 53 completed the intervention. Probio-M8, in combination with dietary intervention, significantly improved CARS and GSRS scores, increased fecal levels of Bifidobacterium animalis, Akkermansia muciniphila, Fusicatenibacter saccharivorans, and Sutterella sp. while also reducing Blautia obeum (Benjamini-Hochberg corrected p ≤ 0.05 for all cases). The intervention also modulated fecal metabolites associated with the metabolism of amino acids (lysine), neurotransmitters (glutamate, γ-aminobutyric acid), polyunsaturated fatty acids (arachidonate, myristic acid), and vitamin B3. In conclusion, Probio-M8 combined with medium-carbohydrate diet effectively improved ASD symptoms, with associated changes in the gut microbiome and metabolome, supporting its potential as an adjunctive therapy for ASD.
Collapse
Affiliation(s)
- Yalin Li
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Weiwei Hu
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Bing Lin
- Department of Clinical Nutrition Shenzhen Hospital, Southern Medical University, Guangdong, China
- Shenzhen Key Laboratory of Gastrointestinal Microbiota and Disease, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Teng Ma
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Zhentian Zhang
- Department of Clinical Nutrition Shenzhen Hospital, Southern Medical University, Guangdong, China
- Shenzhen Key Laboratory of Gastrointestinal Microbiota and Disease, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Weiqian Hu
- Department of Clinical Nutrition Shenzhen Hospital, Southern Medical University, Guangdong, China
- Shenzhen Key Laboratory of Gastrointestinal Microbiota and Disease, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Rui Zhou
- Department of Clinical Nutrition Shenzhen Hospital, Southern Medical University, Guangdong, China
- Shenzhen Key Laboratory of Gastrointestinal Microbiota and Disease, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Lai-Yu Kwok
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Zhihong Sun
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Cuifeng Zhu
- Department of Clinical Nutrition Shenzhen Hospital, Southern Medical University, Guangdong, China
- Shenzhen Key Laboratory of Gastrointestinal Microbiota and Disease, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Heping Zhang
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| |
Collapse
|
31
|
Al-Beltagi M. Pre-autism: What a paediatrician should know about early diagnosis of autism. World J Clin Pediatr 2023; 12:273-294. [PMID: 38178935 PMCID: PMC10762597 DOI: 10.5409/wjcp.v12.i5.273] [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: 07/10/2023] [Revised: 09/07/2023] [Accepted: 09/25/2023] [Indexed: 12/08/2023] Open
Abstract
Autism, also known as an autism spectrum disorder, is a complex neurodevelopmental disorder usually diagnosed in the first three years of a child's life. A range of symptoms characterizes it and can be diagnosed at any age, including adolescence and adulthood. However, early diagnosis is crucial for effective management, prognosis, and care. Unfortunately, there are no established fetal, prenatal, or newborn screening programs for autism, making early detection difficult. This review aims to shed light on the early detection of autism prenatally, natally, and early in life, during a stage we call as "pre-autism" when typical symptoms are not yet apparent. Some fetal, neonatal, and infant biomarkers may predict an increased risk of autism in the coming baby. By developing a biomarker array, we can create an objective diagnostic tool to diagnose and rank the severity of autism for each patient. These biomarkers could be genetic, immunological, hormonal, metabolic, amino acids, acute phase reactants, neonatal brainstem function biophysical activity, behavioral profile, body measurements, or radiological markers. However, every biomarker has its accuracy and limitations. Several factors can make early detection of autism a real challenge. To improve early detection, we need to overcome various challenges, such as raising community awareness of early signs of autism, improving access to diagnostic tools, reducing the stigma attached to the diagnosis of autism, and addressing various culturally sensitive concepts related to the disorder.
Collapse
Affiliation(s)
- Mohammed Al-Beltagi
- Department of Pediatric, Faculty of Medicine, Tanta University, Tanta 31511, Algahrbia, Egypt
- Department of Pediatric, University Medical Center, King Abdulla Medical City, Arabian Gulf University, Dr. Sulaiman Al Habib Medical Group, Manama 26671, Manama, Bahrain
| |
Collapse
|
32
|
Nisar S, Haris M. Neuroimaging genetics approaches to identify new biomarkers for the early diagnosis of autism spectrum disorder. Mol Psychiatry 2023; 28:4995-5008. [PMID: 37069342 PMCID: PMC11041805 DOI: 10.1038/s41380-023-02060-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 03/23/2023] [Accepted: 03/28/2023] [Indexed: 04/19/2023]
Abstract
Autism-spectrum disorders (ASDs) are developmental disabilities that manifest in early childhood and are characterized by qualitative abnormalities in social behaviors, communication skills, and restrictive or repetitive behaviors. To explore the neurobiological mechanisms in ASD, extensive research has been done to identify potential diagnostic biomarkers through a neuroimaging genetics approach. Neuroimaging genetics helps to identify ASD-risk genes that contribute to structural and functional variations in brain circuitry and validate biological changes by elucidating the mechanisms and pathways that confer genetic risk. Integrating artificial intelligence models with neuroimaging data lays the groundwork for accurate diagnosis and facilitates the identification of early diagnostic biomarkers for ASD. This review discusses the significance of neuroimaging genetics approaches to gaining a better understanding of the perturbed neurochemical system and molecular pathways in ASD and how these approaches can detect structural, functional, and metabolic changes and lead to the discovery of novel biomarkers for the early diagnosis of ASD.
Collapse
Affiliation(s)
- Sabah Nisar
- Laboratory of Molecular and Metabolic Imaging, Sidra Medicine, Doha, Qatar
- Department of Diagnostic Imaging, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Mohammad Haris
- Laboratory of Molecular and Metabolic Imaging, Sidra Medicine, Doha, Qatar.
- Center for Advanced Metabolic Imaging in Precision Medicine, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Laboratory Animal Research Center, Qatar University, Doha, Qatar.
| |
Collapse
|
33
|
Alhowikan AM, Elamin NE, Aldayel SS, AlSiddiqi SA, Alrowais FS, Hassan WM, El-Ansary A, Alghamdi FA, AL-Ayadhi LY. Children with Autism Spectrum Disorder Exhibit Elevated Physical Activity and Reduced Sedentary Behavior. Brain Sci 2023; 13:1575. [PMID: 38002535 PMCID: PMC10670306 DOI: 10.3390/brainsci13111575] [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: 11/03/2023] [Accepted: 11/05/2023] [Indexed: 11/26/2023] Open
Abstract
According to previous research, individuals with autism spectrum disorder (ASD) have lower levels of physical activity than their typically developed (TD) counterparts. There have been conflicting reports about physical activity (PA) levels in people with ASD. Given the conflicting evidence, further investigation is required. We believe that evaluating PA in individuals with ASD is critical in order to offer PA intervention plans aiming at increasing their health-related physical fitness on a daily, systematic, and individualized basis. In the current study, an ActiGraph monitor (GT3X+) was used to accurately measure PA and sedentary activity in 21 children with autism aged 6.43 ± 2.29 years and 30 TD children aged 7.2 ± 3.14 years. Our data indicated that while the light and moderate activity counts were not significantly different between the two groups, the vigorous activity was significantly higher in ASD compared to TD. This finding was attributed to ASD characteristic stereotypy and self-stimulating behaviors. The significantly higher vigorous PA is discussed in relation to altered neurochemistry, oxidative stress, and neuroinflammation as etiological mechanisms in ASD. This research provides a better understanding of the status of PA participation in individuals with ASD.
Collapse
Affiliation(s)
- Abdulrahman M. Alhowikan
- Department of Physiology, College of Medicine, King Saud University, P.O. Box 2925, Riyadh 11461, Saudi Arabia;
| | - Nadra E. Elamin
- Autism Research and Treatment Center, Department of Physiology, Faculty of Medicine, King Saud University, Riyadh 11461, Saudi Arabia;
| | - Sarah S. Aldayel
- Department of Medicine, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia; (S.S.A.); (F.S.A.)
| | - Sara A. AlSiddiqi
- King Abdullah Bin Abdulaziz University Hospital (KAAUH), Riyadh 11461, Saudi Arabia;
| | - Fai S. Alrowais
- Department of Medicine, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia; (S.S.A.); (F.S.A.)
| | - Wail M. Hassan
- Department of Biomedical Sciences, University of Missouri-Kansas City School of Medicine, Kansas City, MO 64108, USA;
| | - Afaf El-Ansary
- Autism Research and Treatment Center, Department of Physiology, Faculty of Medicine, King Saud University, Riyadh 11461, Saudi Arabia;
- Autism Center, Lotus Holistic Medical Center, Abu Dhabi P.O. Box 110281, United Arab Emirates
| | - Farah Ali Alghamdi
- College of Medicine, Dar Al-Olum University (DAU), Riyadh 13314, Saudi Arabia;
| | - Laila Y. AL-Ayadhi
- Department of Physiology, College of Medicine, King Saud University, P.O. Box 2925, Riyadh 11461, Saudi Arabia;
- Autism Research and Treatment Center, Department of Physiology, Faculty of Medicine, King Saud University, Riyadh 11461, Saudi Arabia;
| |
Collapse
|
34
|
Schmitz I, da Silva A, Bobermin LD, Gonçalves CA, Steiner J, Quincozes-Santos A. The Janus face of antipsychotics in glial cells: Focus on glioprotection. Exp Biol Med (Maywood) 2023; 248:2120-2130. [PMID: 38230521 PMCID: PMC10800129 DOI: 10.1177/15353702231222027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2024] Open
Abstract
Antipsychotics are commonly prescribed to treat several neuropsychiatric disorders, including schizophrenia, mania in bipolar disorder, autism spectrum disorder, delirium, and organic or secondary psychosis, for example, in dementias such as Alzheimer's disease. There is evidence that typical antipsychotics such as haloperidol are more effective in reducing positive symptoms than negative symptoms and/or cognitive deficits. In contrast, atypical antipsychotic agents have gained popularity over typical antipsychotics, due to fewer extrapyramidal side effects and their theoretical efficacy in controlling both positive and negative symptoms. Although these therapies focus on neuron-based therapeutic schemes, glial cells have been recognized as important regulators of the pathophysiology of neuropsychiatric disorders, as well as targets to improve the efficacy of these drugs. Glial cells (astrocytes, oligodendrocytes, and microglia) are critical for the central nervous system in both physiological and pathological conditions. Astrocytes are the most abundant glial cells and play important roles in brain homeostasis, regulating neurotransmitter systems and gliotransmission, since they express a wide variety of functional receptors for different neurotransmitters. In addition, converging lines of evidence indicate that psychiatric disorders are commonly associated with the triad neuroinflammation, oxidative stress, and excitotoxicity, and that glial cells may contribute to the gliotoxicity process. Conversely, glioprotective molecules attenuate glial damage by generating specific responses that can protect glial cells themselves and/or neurons, resulting in improved central nervous system (CNS) functioning. In this regard, resveratrol is well-recognized as a glioprotective molecule, including in clinical studies of schizophrenia and autism. This review will provide a summary of the dual role of antipsychotics on neurochemical parameters associated with glial functions and will highlight the potential activity of glioprotective molecules to improve the action of antipsychotics.
Collapse
Affiliation(s)
- Izaviany Schmitz
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003, Brazil
| | - Amanda da Silva
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003, Brazil
| | - Larissa Daniele Bobermin
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003, Brazil
| | - Carlos-Alberto Gonçalves
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003, Brazil
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003, Brazil
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003, Brazil
| | - Johann Steiner
- Department of Psychiatry, University of Magdeburg, Magdeburg 39120, Germany
| | - André Quincozes-Santos
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003, Brazil
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003, Brazil
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003, Brazil
| |
Collapse
|
35
|
Wang F, Yang X, Ren Z, Chen C, Liu C. Alternative splicing in mouse brains affected by psychological stress is enriched in the signaling, neural transmission and blood-brain barrier pathways. Mol Psychiatry 2023; 28:4707-4718. [PMID: 37217679 DOI: 10.1038/s41380-023-02103-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 04/26/2023] [Accepted: 05/03/2023] [Indexed: 05/24/2023]
Abstract
Psychological stress increases the risk of major psychiatric disorders. Psychological stress on mice was reported to induce differential gene expression (DEG) in mice brain regions. Alternative splicing is a fundamental aspect of gene expression and has been associated with psychiatric disorders but has not been investigated in the stressed brain yet. This study investigated changes in gene expression and splicing under psychological stress, the related pathways, and possible relationship with psychiatric disorders. RNA-seq raw data of 164 mouse brain samples from 3 independent datasets with stressors including chronic social defeat stress (CSDS), early life stress (ELS), and two-hit stress of combined CSDS and ELS were collected. There were more changes in splicing than in gene expression in the ventral hippocampus and medial prefrontal cortex, but stress-induced changes of individual genes by differential splicing and differential expression could not be replicated. In contrast, pathway analyses produced robust findings: stress-induced differentially spliced genes (DSGs) were reproducibly enriched in neural transmission and blood-brain barrier systems, and DEGs were reproducibly enriched in stress response-related functions. The hub genes of DSG-related PPI networks were enriched in synaptic functions. The corresponding human homologs of stress-induced DSGs were robustly enriched in AD-related DSGs as well as BD and SCZ in GWAS. These results suggested that stress-induced DSGs from different datasets belong to the same biological system throughout the stress response process, resulting in consistent stress response effects.
Collapse
Affiliation(s)
- Feiran Wang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, and Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiuju Yang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, and Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zongyao Ren
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, and Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chao Chen
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, and Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center on Mental Disorders, The Second Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Chunyu Liu
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, and Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China.
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, 13210, USA.
| |
Collapse
|
36
|
El-Ansary A, Al-Ayadhi L. Effects of Walnut and Pumpkin on Selective Neurophenotypes of Autism Spectrum Disorders: A Case Study. Nutrients 2023; 15:4564. [PMID: 37960217 PMCID: PMC10647375 DOI: 10.3390/nu15214564] [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: 09/19/2023] [Revised: 10/23/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
Special diets or nutritional supplements are regularly given to treat children with autism spectrum disorder (ASD). The increased consumption of particular foods has been demonstrated in numerous trials to lessen autism-related symptoms and comorbidities. A case study on a boy with moderate autism who significantly improved after three years of following a healthy diet consisting of pumpkin and walnuts was examined in this review in connection to a few different neurophenotypes of ASD. We are able to suggest that a diet high in pumpkin and walnuts was useful in improving the clinical presentation of the ASD case evaluated by reducing oxidative stress, neuroinflammation, glutamate excitotoxicity, mitochondrial dysfunction, and altered gut microbiota, all of which are etiological variables. Using illustrated figures, a full description of the ways by which a diet high in pumpkin and nuts could assist the included case is offered.
Collapse
Affiliation(s)
- Afaf El-Ansary
- Autism Center, Lotus Holistic Alternative Medical Center, Abu Dhabi P.O. Box 110281, United Arab Emirates
- Autism Research and Treatment Center, P.O. Box 2925, Riyadh 11461, Saudi Arabia;
| | - Laila Al-Ayadhi
- Autism Research and Treatment Center, P.O. Box 2925, Riyadh 11461, Saudi Arabia;
- Department of Physiology, Faculty of Medicine, King Saud University, P.O. Box 2925, Riyadh 11461, Saudi Arabia
| |
Collapse
|
37
|
Wei J, Chen J, Fang X, Liu T, Yuan Y, Zhang J. Protocol for the safety and efficacy of fecal microbiota transplantation liquid in children with autism spectrum disorder: a randomized controlled study. Front Microbiol 2023; 14:1236904. [PMID: 37675433 PMCID: PMC10477363 DOI: 10.3389/fmicb.2023.1236904] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/08/2023] [Indexed: 09/08/2023] Open
Abstract
Background Autism Spectrum Disorder (ASD) is a neurodevelopmental disorder characterized by deficits in social interaction, repetitive behavior and language impairment, and its worldwide prevalence has been found to be increasing annually in recent years. Till now, ASD is uncurable as its pathogenesis remains unknown. However, studies on both animals and humans have demonstrated that fecal microbiota transplantation (FMT) may ameliorate the symptoms of ASD, as well as gastrointestinal symptoms. Nonetheless, there is still no agreement regarding the optimal dosage or duration of FMT treatment for individuals with ASD. Methods This clinical study is a double-blind, randomized, interventional trial conducted at a single center. The aim is to investigate the safety and efficacy of a pediatric formulation of FMT for ASD. A total of 42 children between the ages of 3-9 with ASD will be randomly assigned in a 2:1 ratio to either an FMT treatment group (n = 28) or a placebo group (n = 14), forming cohort 1. Additionally, 30 healthy children of similar age and gender will be recruited as the control group (cohort 2). Cohort 1 will be assessed using a variety of scales, including the Autism Behavior Checklist, Childhood Autism Rating Scale, Social Responsiveness Scale, Gastrointestinal Symptom Rating Scale, Children's Sleep Habits Questionnaire, and Psychoeducational Profile (Third Edition). These assessments will evaluate the effectiveness of FMT in reducing core symptoms and comorbidities (such as gastrointestinal symptoms and sleep disturbances) in children with ASD. The study will use metagenomic and metabolomic sequencing to assess changes in the composition and structure of the intestinal flora and its metabolites in blood, urine, and feces following treatment. Furthermore, the study will evaluate the acceptability of the FMT formulation by participants' legal guardians and investigate differences in the intestinal flora and metabolism in the FMT group before and after treatment compared to 30 healthy children. Clinical trial registration https://www.chictr.org.cn/, identifier ChiCTR2200058459.
Collapse
Affiliation(s)
- Jinying Wei
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Jiayi Chen
- Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaohui Fang
- Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianyu Liu
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Yanhan Yuan
- Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinping Zhang
- Pediatrics, Shanghai Sixth People’s Hospital, Shanghai, China
| |
Collapse
|
38
|
Oya M, Matsuoka K, Kubota M, Fujino J, Tei S, Takahata K, Tagai K, Yamamoto Y, Shimada H, Seki C, Itahashi T, Aoki YY, Ohta H, Hashimoto RI, Sugihara G, Obata T, Zhang MR, Suhara T, Nakamura M, Kato N, Takado Y, Takahashi H, Higuchi M. Increased glutamate and glutamine levels and their relationship to astrocytes and dopaminergic transmissions in the brains of adults with autism. Sci Rep 2023; 13:11655. [PMID: 37468523 DOI: 10.1038/s41598-023-38306-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 07/06/2023] [Indexed: 07/21/2023] Open
Abstract
Increased excitatory neuronal tones have been implicated in autism, but its mechanism remains elusive. The amplified glutamate signals may arise from enhanced glutamatergic circuits, which can be affected by astrocyte activation and suppressive signaling of dopamine neurotransmission. We tested this hypothesis using magnetic resonance spectroscopy and positron emission tomography scan with 11C-SCH23390 for dopamine D1 receptors in the anterior cingulate cortex (ACC). We enrolled 18 male adults with high-functioning autism and 20 typically developed (TD) male subjects. The autism group showed elevated glutamate, glutamine, and myo-inositol (mI) levels compared with the TD group (p = 0.045, p = 0.044, p = 0.030, respectively) and a positive correlation between glutamine and mI levels in the ACC (r = 0.54, p = 0.020). In autism and TD groups, ACC D1 receptor radioligand binding was negatively correlated with ACC glutamine levels (r = - 0.55, p = 0.022; r = - 0.58, p = 0.008, respectively). The enhanced glutamate-glutamine metabolism might be due to astroglial activation and the consequent reinforcement of glutamine synthesis in autistic brains. Glutamine synthesis could underly the physiological inhibitory control of dopaminergic D1 receptor signals. Our findings suggest a high neuron excitation-inhibition ratio with astrocytic activation in the etiology of autism.
Collapse
Affiliation(s)
- Masaki Oya
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba, 263-8555, Japan
- Department of Psychiatry and Behavioral Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Kiwamu Matsuoka
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba, 263-8555, Japan.
- Department of Psychiatry, Nara Medical University, Kashihara-shi, Nara, Japan.
| | - Manabu Kubota
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba, 263-8555, Japan
- Department of Psychiatry, Graduate School of Medicine, Kyoto University, Kyoto-shi, Kyoto, Japan
- Medical Institute of Developmental Disabilities Research, Showa University, Setagaya-ku, Tokyo, Japan
| | - Junya Fujino
- Department of Psychiatry and Behavioral Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
- Medical Institute of Developmental Disabilities Research, Showa University, Setagaya-ku, Tokyo, Japan
| | - Shisei Tei
- Department of Psychiatry, Graduate School of Medicine, Kyoto University, Kyoto-shi, Kyoto, Japan
- Medical Institute of Developmental Disabilities Research, Showa University, Setagaya-ku, Tokyo, Japan
- Institute of Applied Brain Sciences, Waseda University, Tokorozawa-shi, Saitama, Japan
- School of Human and Social Sciences, Tokyo International University, Kawagoe-shi, Saitama, Japan
| | - Keisuke Takahata
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba, 263-8555, Japan
- Department of Neuropsychiatry, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Kenji Tagai
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba, 263-8555, Japan
| | - Yasuharu Yamamoto
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba, 263-8555, Japan
- Department of Neuropsychiatry, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Hitoshi Shimada
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba, 263-8555, Japan
- Center for Integrated Human Brain Science, Brain Research Institute, Niigata University, Niigata-shi, Niigata, Japan
| | - Chie Seki
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba, 263-8555, Japan
| | - Takashi Itahashi
- Medical Institute of Developmental Disabilities Research, Showa University, Setagaya-ku, Tokyo, Japan
| | - Yuta Y Aoki
- Medical Institute of Developmental Disabilities Research, Showa University, Setagaya-ku, Tokyo, Japan
| | - Haruhisa Ohta
- Medical Institute of Developmental Disabilities Research, Showa University, Setagaya-ku, Tokyo, Japan
- Department of Psychiatry, School of Medicine, Showa University, Setagaya-ku, Tokyo, Japan
| | - Ryu-Ichiro Hashimoto
- Medical Institute of Developmental Disabilities Research, Showa University, Setagaya-ku, Tokyo, Japan
- Department of Language Sciences, Graduate School of Humanities, Tokyo Metropolitan University, Hachioji-shi, Tokyo, Japan
| | - Genichi Sugihara
- Department of Psychiatry and Behavioral Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Takayuki Obata
- Department of Molecular Imaging and Theranostics, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba-shi, Chiba, Japan
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba-shi, Chiba, Japan
| | - Tetsuya Suhara
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba, 263-8555, Japan
| | - Motoaki Nakamura
- Medical Institute of Developmental Disabilities Research, Showa University, Setagaya-ku, Tokyo, Japan
- Kanagawa Psychiatric Center, Yokohama-shi, Kanagawa, Japan
| | - Nobumasa Kato
- Medical Institute of Developmental Disabilities Research, Showa University, Setagaya-ku, Tokyo, Japan
| | - Yuhei Takado
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba, 263-8555, Japan
| | - Hidehiko Takahashi
- Department of Psychiatry and Behavioral Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
- Center for Brain Integration Research, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Makoto Higuchi
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba, 263-8555, Japan
| |
Collapse
|
39
|
Yang Y, Zhou S, Xing Y, Yang G, You M. Impact of pesticides exposure during neurodevelopmental period on autism spectrum disorders - A focus on gut microbiota. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 260:115079. [PMID: 37262968 DOI: 10.1016/j.ecoenv.2023.115079] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/03/2023]
Abstract
Accumulating evidence indicates exposure to pesticides during the crucial neurodevelopmental period increases susceptibility to many diseases, including the neurodevelopmental disorder known as autism spectrum disorder (ASD). In the last few years, it has been hypothesized that gut microbiota dysbiosis is strongly implicated in the aetiopathogenesis of ASD. Recently, new studies have suggested that the gut microbiota may be involved in the neurological and behavioural defects caused by pesticides, including ASD symptoms. This review highlights the available evidence from recent animal and human studies on the relationship between pesticides that have the potential to disturb intestinal microbiota homeostasis, and ASD symptoms. The mechanisms through which gut microbiota dysbiosis may trigger ASD-like behaviours induced by pesticides exposure during the neurodevelopmental period via the altered production of bacterial metabolites (short chain fatty acids, lipids, retinol, and amino acid) are also described. According to recent research, gut microbiota dysbiosis may be a major contributor to the symptoms of ASD associated with pesticides exposure. However, to determine the detailed mechanism of action of gut microbiota on pesticide-induced ASD behaviours, actual population exposure scenarios from epidemiological studies should be used as the basis for the appropriate exposure pattern and dosage to be used in animal studies.
Collapse
Affiliation(s)
- Yongyong Yang
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Shun Zhou
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Ying Xing
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550025, China; Guizhou Provincial Center for Disease Control and Prevention, Guiyang, Guizhou 550004, China; School of Public Health, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Guanghong Yang
- Guizhou Provincial Center for Disease Control and Prevention, Guiyang, Guizhou 550004, China; School of Public Health, Guizhou Medical University, Guiyang, Guizhou 550025, China.
| | - Mingdan You
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550025, China.
| |
Collapse
|
40
|
Bagheri J, Fallahnezhad S, Alipour N, Babaloo H, Tahmasebi F, Kheradmand H, Sazegar G, Haghir H. Maternal diabetes decreases the expression of GABA Aα1, GABA B1, and mGlu2 receptors in the visual cortex of male rat neonates. Neurosci Lett 2023; 809:137309. [PMID: 37230455 DOI: 10.1016/j.neulet.2023.137309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/12/2023] [Accepted: 05/17/2023] [Indexed: 05/27/2023]
Abstract
AIMS This study examines the impact of maternal diabetes on the expression of GABAB1, GABAAα1, and mGlu2 receptors in the primary visual cortex layers of male rat newborns. MAIN METHODS In diabetic group (Dia), diabetes was induced in adult female rats using an intraperitoneal dose of Streptozotocin (STZ) 65 (mg/kg). Diabetes was managed by daily subcutaneous injection of NPH insulin in insulin-treated diabetic group (Ins). Control group (Con) received normal saline intraperitoneally rather than STZ. Male offspring born to each group of female rats were euthanized via CO2 inhalation at P0, P7, and P14 days after delivery and the expression of GABAB1, GABAAα1, and mGlu2 receptors in their primary visual cortex was determined using immunohistochemistry (IHC). KEY FINDINGS The expression of GABAB1, GABAAα1, and mGlu2 receptors increased gradually with age in the male offspring born to Con group while the highest expression was detected in layer IV of the primary visual cortex. In Dia group newborns, the expression of these receptors was significantly reduced in all layers of the primary visual cortex at every three days. Insulin treatment in diabetic mothers restored the expression of these receptors to normal levels in their newborns. SIGNIFICANCE The study indicates that diabetes reduces the expression of GABAB1, GABAAα1, and mGlu2 receptors in the primary visual cortex of male offspring born to diabetic rats at P0, P7, and P14. However, insulin treatment can counteract these effects.
Collapse
Affiliation(s)
- Javad Bagheri
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Somaye Fallahnezhad
- Nervous System Stem Cell Research Center, Semnan University of Medical Sciences, Semnan, Iran; Department of Anatomical Sciences, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Nasim Alipour
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Hamideh Babaloo
- Regenerative Medicine, Organ Procurement and Transplantation Multidisciplinary Center, Razi Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Fatemeh Tahmasebi
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Hamed Kheradmand
- Department of Neurosurgery, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ghasem Sazegar
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Hossein Haghir
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Genetic Research Center (MGRC), School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
41
|
Tran H, Le L, Singh BN, Kramer J, Steward R. Tet Controls Axon Guidance in Early Brain Development through Glutamatergic Signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.02.539069. [PMID: 37398066 PMCID: PMC10312521 DOI: 10.1101/2023.05.02.539069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Mutations in human TET proteins have been found in individuals with neurodevelopmental disorders. Here we report a new function of Tet in regulating Drosophila early brain development. We found that mutation in the Tet DNA-binding domain ( Tet AXXC ) resulted in axon guidance defects in the mushroom body (MB). Tet is required in early brain development during the outgrowth of MB β axons. Transcriptomic study shows that glutamine synthetase 2 (Gs2), a key enzyme in glutamatergic signaling, is significantly downregulated in the Tet AXXC mutant brains. CRISPR/Cas9 mutagenesis or RNAi knockdown of Gs2 recapitulates the Tet AXXC mutant phenotype. Surprisingly, Tet and Gs2 act in the insulin-producing cells (IPCs) to control MB axon guidance, and overexpression of Gs2 in these cells rescues the axon guidance defects of Tet AXXC . Treating Tet AXXC with the metabotropic glutamate receptor antagonist MPEP can rescue while treating with glutamate enhances the phenotype confirming Tet function in regulating glutamatergic signaling. Tet AXXC and the Drosophila homolog of Fragile X Messenger Ribonucleoprotein protein mutant ( Fmr1 3 ) have similar axon guidance defects and reduction in Gs2 mRNA levels. Interestingly, overexpression of Gs2 in the IPCs also rescues the Fmr1 3 phenotype, suggesting functional overlapping of the two genes. Our studies provide the first evidence that Tet can control the guidance of axons in the developing brain by modulating glutamatergic signaling and the function is mediated by its DNA-binding domain.
Collapse
|
42
|
Al-Mazidi SH, El-Ansary A, Abualnaja A, AlZarroug A, Alharbi T, Al-Ayadhi LY. Exploring the Potential Role of ADAM 17 and ADAM 22 in the Etiology of Autism Spectrum Disorders. Brain Sci 2023; 13:972. [PMID: 37371450 DOI: 10.3390/brainsci13060972] [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: 05/30/2023] [Revised: 06/16/2023] [Accepted: 06/18/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) encompasses a group of disorders characterized by difficulties with social interaction and repetitive behavior. The condition is supposed to originate from early shifts in brain development, while the underlying processes are unknown. Moreover, a considerable number of patients with ASD experience digestive difficulties. Metalloproteases (ADAMs) are a class of enzymes capable of cleaving membrane-bound proteins. Members of this family, ADAM17 and ADAM22, have the ability to cleave proteins like the pro-inflammatory cytokine TNF-ά and glutamate synaptic molecules, which are both engaged in neuro-inflammation and glutamate excitotoxicity as crucial etiological mechanisms in ASD. ADAM17 and ADAM22 may also have a role in ASD microbiota-gut-brain axis connections by regulating immunological and inflammatory responses in the intestinal tract. SUBJECTS AND METHODS Using ELISA kits, the plasma levels of ADAM17 and ADAM22 were compared in 40 children with ASD and 40 typically developing children. All of the autistic participants' childhood autism rating scores (CARS), social responsiveness scales (SRS), and short sensory profiles (SSP) were evaluated as indicators of ASD severity. RESULTS Our results showed that plasma levels of ADAM17 were significantly lower in ASD children than in control children, while ADAM22 demonstrated non-significantly lower levels. Our data also indicate that while ADAM17 correlates significantly with age, ADAM22 correlates significantly with CARS as a marker of ASD severity. CONCLUSIONS Our interpreted data showed that alteration in ADAM17 and ADAM22 might be associated with glutamate excitotoxicity, neuroinflammation, and altered gut microbiota as etiological mechanisms of ASD and could be an indicator of the severity of the disorder.
Collapse
Affiliation(s)
- Sarah H Al-Mazidi
- Department of Physiology, Faculty of Medicine, Imam Mohammed Ibn Saud Islamic University, Riyadh 11432, Saudi Arabia
| | - Afaf El-Ansary
- Autism Center, Lotus Holistic Alternative Medical Center, Abu Dhabi 110281, United Arab Emirates
- Autism Research and Treatment Centre, King Saud University, Riyadh 11461, Saudi Arabia
| | - Amani Abualnaja
- Department of Physiology, Faculty of Medicine, Imam Mohammed Ibn Saud Islamic University, Riyadh 11432, Saudi Arabia
| | - Abdullah AlZarroug
- Department of Physiology, Faculty of Medicine, Imam Mohammed Ibn Saud Islamic University, Riyadh 11432, Saudi Arabia
| | - Turki Alharbi
- Department of Physiology, Faculty of Medicine, Imam Mohammed Ibn Saud Islamic University, Riyadh 11432, Saudi Arabia
| | - Laila Y Al-Ayadhi
- Autism Research and Treatment Centre, King Saud University, Riyadh 11461, Saudi Arabia
- Department of Physiology, Faculty of Medicine, King Saud University, Riyadh 11461, Saudi Arabia
| |
Collapse
|
43
|
Al-Mazidi S, Al-Ayadhi L, Alqahtany F, Abualnaja A, Alzarroug A, Alharbi T, Farhat K, AlMnaizel A, El-Ansary A. The possible role of sodium leakage channel localization factor-1 in the pathophysiology and severity of autism spectrum disorders. Sci Rep 2023; 13:9747. [PMID: 37328585 PMCID: PMC10275888 DOI: 10.1038/s41598-023-36953-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 06/13/2023] [Indexed: 06/18/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by social, stereotypical, and repetitive behaviors. Neural dysregulation was proposed as an etiological factor in ASD. The sodium leakage channel (NCA), regulated by NLF-1 (NCA localization factor-1), has a major role in maintaining the physiological excitatory function of neurons. We aimed to examine the level of NLF-1 in ASD children and correlate it with the severity of the disease. We examined the plasma levels of NLF-1 in 80 ASD and neurotypical children using ELISA. The diagnosis and severity of ASD were based on the Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition (DSM-IV), Childhood Autism Rating Score, Social Responsiveness Scale, and Short Sensory Profile. Then, we compared the levels of NLF-1 with the severity of the disease and behavioral and sensory symptoms. Our results showed a significant decrease in the plasma levels of NLF-1 in ASD children compared to neurotypical children (p < 0.001). Additionally, NLF-1 was significantly correlated with the severity of the behavioral symptoms of ASD (p < 0.05). The low levels of NLF-1 in ASD children potentially affect the severity of their behavioral symptoms by reducing neuron excitability through NCA. These novel findings open a new venue for pharmacological and possible genetic research involving NCA in ASD children.
Collapse
Affiliation(s)
- Sarah Al-Mazidi
- Physiology Department, College of Medicine, Imam Mohammad Ibn Saud Islamic University, P.O.Box: 5701, Riyadh, 11432, Saudi Arabia.
| | - Laila Al-Ayadhi
- Physiology, King Saud University College of Medicine, Riyadh, Saudi Arabia
- Autism Research and Treatment Center, King Saud University College of Medicine, Riyadh, Saudi Arabia
| | - Fatmah Alqahtany
- Hematopathology Unit, Department of Pathology, College of Medicine, King Saud University, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Amani Abualnaja
- College of Medicine, Imam Muhammad bin Saud Islamic University, Riyadh, Saudi Arabia
| | - Abdullah Alzarroug
- College of Medicine, Imam Muhammad bin Saud Islamic University, Riyadh, Saudi Arabia
| | - Turki Alharbi
- College of Medicine, Imam Muhammad bin Saud Islamic University, Riyadh, Saudi Arabia
| | - Karim Farhat
- Cancer Research Chair, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Ahmad AlMnaizel
- Research office, John Hopkins Aramco Healthcare, Dahran, Saudi Arabia
| | - Afaf El-Ansary
- Autism Research and Treatment Center, King Saud University College of Medicine, Riyadh, Saudi Arabia
| |
Collapse
|
44
|
Kamalmaz N, Ben Bacha A, Alonazi M, Albasher G, Khayyat AIA, El-Ansary A. Unveiling sex-based differences in developing propionic acid-induced features in mice as a rodent model of ASD. PeerJ 2023; 11:e15488. [PMID: 37334116 PMCID: PMC10274690 DOI: 10.7717/peerj.15488] [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: 01/25/2023] [Accepted: 05/10/2023] [Indexed: 06/20/2023] Open
Abstract
Background Males are more likely to develop autism as a neurodevelopmental disorder than females are, although the mechanisms underlying male vulnerability are not fully understood. Therefore, studying the role of autism etiologies considering sex differences in the propionic acid (PPA) rodent model of autism would build greater understanding of how females are protected from autism spectrum disorder, which may be used as a treatment strategy for males with autism. Objectives This study aimed to investigate the sex differences in oxidative stress, glutamate excitotoxicity, neuroinflammation, and gut microbiota impairment as etiological mechanisms for many neurological diseases, with specific reference to autism. Method Forty albino mice were divided into four groups of 10 animals each with two control and two treated groups of both sexes received only phosphate-buffered saline or a neurotoxic dose of PPA (250 mg/kg body weight) for 3 days, respectively. Biochemical markers of energy metabolism, oxidative stress, neuroinflammation, and excitotoxicity were measured in mouse brain homogenates, whereas the presence of pathogenic bacteria was assessed in mouse stool samples. Furthermore, the repetitive behavior, cognitive ability, and physical-neural coordination of the animals were examined. Results Collectively, selected variables related to oxidative stress, glutamate excitotoxicity, neuroinflammation, and gut bacteria were impaired concomitantly with altered behavior in PPA-induced rodent model, with males being more susceptible than females. Conclusion This study explains the role of sex in the higher vulnerability of males to develop autistic biochemical and behavioral features compared with females. Female sex hormones and the higher detoxification capacity and higher glycolytic flux in females serve as neuroprotective contributors in a rodent model of autism.
Collapse
Affiliation(s)
- Nasreen Kamalmaz
- Biochemistry Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Abir Ben Bacha
- Biochemistry Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Mona Alonazi
- Biochemistry Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Gadah Albasher
- Zoology Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Arwa Ishaq A. Khayyat
- Biochemistry Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Afaf El-Ansary
- Central Research Laboratory, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
45
|
Alsubaiei SRM, Alfawaz HA, Bhat RS, El-Ansary A. Nutritional Intervention as a Complementary Neuroprotective Approach against Propionic Acid-Induced Neurotoxicity and Associated Biochemical Autistic Features in Rat Pups. Metabolites 2023; 13:738. [PMID: 37367896 DOI: 10.3390/metabo13060738] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/26/2023] [Accepted: 06/07/2023] [Indexed: 06/28/2023] Open
Abstract
Since there is no known cure for autism spectrum disorder (ASD), its incidence rate is on the rise. Common comorbidities like gastrointestinal problems are observed as common signs of ASD and play a major role in controlling social and behavioral symptoms. Although there is a lot of interest in dietary treatments, no harmony exists with regard to the ideal nutritional therapy. To better direct prevention and intervention measures for ASD, the identification of risk and protective factors is required. Through the use of a rat model, our study aims to assess the possible danger of exposure to neurotoxic doses of propionic acid (PPA) and the nutritional protective effects of prebiotics and probiotics. Here, we conducted a biochemical assessment of the effects of dietary supplement therapy in the PPA model of autism. We used 36 male Sprague Dawley albino rat pups divided into six groups. Standard food and drink were given to the control group. The PPA-induced ASD model was the second group; it was fed a conventional diet for 27 days before receiving 250 mg/kg of PPA orally for three days. The four other groups were given 3 mL/kg of yoghurt daily, 400 mg/Kg of artichokes daily, 50 mg/kg of luteolin daily and Lacticaseibacillus rhamnosus GG at 0.2 mL daily for 27 days before being given PPA (250 mg/kg BW) for three days along with their normal diet. All groups had their brain homogenates tested for biochemical markers, which included gamma-aminobutyric acid (GABA), glutathione peroxidase 1 (GPX1), glutathione (GSH), interleukin 6 (IL-6), interleukin 10 (IL-10) and tumor necrosis factor-alpha (TNF). When compared with the control group, the PPA-induced model presented increased oxidative stress and neuroinflammation but groups treated with all four dietary therapies presented improvements in biochemical characteristics for oxidative stress and neuroinflammation. As all of the therapies show sufficient anti-inflammatory and antioxidant effects, they can be used as a useful dietary component to help prevent ASD.
Collapse
Affiliation(s)
- Sana Razhan M Alsubaiei
- Department of Food Science and Nutrition, College of Food & Agriculture Sciences, King Saud University, Riyadh 11495, Saudi Arabia
| | - Hanan A Alfawaz
- Department of Food Science and Nutrition, College of Food & Agriculture Sciences, King Saud University, Riyadh 11495, Saudi Arabia
| | - Ramesa Shafi Bhat
- Biochemistry Department, Science College, King Saud University, Riyadh 11495, Saudi Arabia
| | - Afaf El-Ansary
- Central Research Laboratory, Female Campus, King Saud University, Riyadh 11495, Saudi Arabia
| |
Collapse
|
46
|
Ilchibaeva T, Tsybko A, Lipnitskaya M, Eremin D, Milutinovich K, Naumenko V, Popova N. Brain-Derived Neurotrophic Factor (BDNF) in Mechanisms of Autistic-like Behavior in BTBR Mice: Crosstalk with the Dopaminergic Brain System. Biomedicines 2023; 11:biomedicines11051482. [PMID: 37239153 DOI: 10.3390/biomedicines11051482] [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: 04/25/2023] [Revised: 05/17/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
Disturbances in neuroplasticity undoubtedly play an important role in the development of autism spectrum disorders (ASDs). Brain neurotransmitters and brain-derived neurotrophic factor (BDNF) are known as crucial players in cerebral and behavioral plasticity. Such an important neurotransmitter as dopamine (DA) is involved in the behavioral inflexibility of ASD. Additionally, much evidence from human and animal studies implicates BDNF in ASD pathogenesis. Nonetheless, crosstalk between BDNF and the DA system has not been studied in the context of an autistic-like phenotype. For this reason, the aim of our study was to compare the effects of either the acute intracerebroventricular administration of a recombinant BDNF protein or hippocampal adeno-associated-virus-mediated BDNF overexpression on autistic-like behavior and expression of key DA-related and BDNF-related genes in BTBR mice (a widely recognized model of autism). The BDNF administration failed to affect autistic-like behavior but downregulated Comt mRNA in the frontal cortex and hippocampus; however, COMT protein downregulation in the hippocampus and upregulation in the striatum were insignificant. BDNF administration also reduced the receptor TrkB level in the frontal cortex and midbrain and the BDNF/proBDNF ratio in the striatum. In contrast, hippocampal BDNF overexpression significantly diminished stereotypical behavior and anxiety; these alterations were accompanied only by higher hippocampal DA receptor D1 mRNA levels. The results indicate an important role of BDNF in mechanisms underlying anxiety and repetitive behavior in ASDs and implicates BDNF-DA crosstalk in the autistic-like phenotype of BTBR mice.
Collapse
Affiliation(s)
- Tatiana Ilchibaeva
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Prospekt Akad. Lavrentyeva 10, 630090 Novosibirsk, Russia
| | - Anton Tsybko
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Prospekt Akad. Lavrentyeva 10, 630090 Novosibirsk, Russia
| | - Marina Lipnitskaya
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Prospekt Akad. Lavrentyeva 10, 630090 Novosibirsk, Russia
| | - Dmitry Eremin
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Prospekt Akad. Lavrentyeva 10, 630090 Novosibirsk, Russia
| | - Kseniya Milutinovich
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Prospekt Akad. Lavrentyeva 10, 630090 Novosibirsk, Russia
| | - Vladimir Naumenko
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Prospekt Akad. Lavrentyeva 10, 630090 Novosibirsk, Russia
| | - Nina Popova
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Prospekt Akad. Lavrentyeva 10, 630090 Novosibirsk, Russia
| |
Collapse
|
47
|
Al-Ayadhi L, Bhat RS, Alghamdi FA, Alhadlaq AS, El-Ansary A. Influence of Auditory Integrative Training on Casein Kinase 2 and Its Impact on Behavioral and Social Interaction in Children with Autism Spectrum Disorder. Curr Issues Mol Biol 2023; 45:4317-4330. [PMID: 37232743 DOI: 10.3390/cimb45050274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/07/2023] [Accepted: 05/08/2023] [Indexed: 05/27/2023] Open
Abstract
Considerable disturbances in post-translational protein phosphorylation have recently been discovered in multiple neurological disorders. Casein kinase-2 (CK2) is a tetrameric Ser/Thr protein kinase that phosphorylates a large number of substrates and contributes in several cellular physiological and pathological processes. CK2 is highly expressed in the mammalian brain and catalyzes the phosphorylation of a large number of substrates that are crucial in neuronal or glial homeostasis and inflammatory signaling processes across synapses. In this study, we investigated the impact of auditory integration therapy (AIT) for the treatment of sensory processing abnormalities in autism on plasma CK2 levels. A total of 25 ASD children, aged between 5 and 12 years, were enrolled and participated in the present research study. AIT was performed for two weeks, for a period of 30 min, twice a day, with a 3 h interval between sessions. Before and after AIT, the Childhood Autism Rating Scale (CARS), Social Responsiveness Scale (SRS), and Short Sensory Profile (SSP) scores were calculated, and plasma CK2 levels were assayed using an ELISA test. The CARS and SRS indices of autism severity improved as a result of AIT, which could be related to the decreased level of plasma CK2. However, the mean value of the SSP scores was not significantly increased after AIT. The relationship between CK2 downregulation and glutamate excitotoxicity, neuro-inflammation, and leaky gut, as etiological mechanisms in ASD, was proposed and discussed. Further research, conducted on a larger scale and with a longer study duration, are required to assess whether the cognitive improvement in ASD children after AIT is related to the downregulation of CK2.
Collapse
Affiliation(s)
- Laila Al-Ayadhi
- Department of Physiology, Faculty of Medicine, King Saud University, Riyadh 11495, Saudi Arabia
- Autism Research and Treatment Center, Riyadh 12713, Saudi Arabia
| | - Ramesa Shafi Bhat
- Biochemistry Department, College of Science, King Saud University, Riyadh 11495, Saudi Arabia
| | - Farah Ali Alghamdi
- College of Medicine, Dar Al Uloom University, Riyadh 13314, Saudi Arabia
| | | | - Afaf El-Ansary
- Autism Research and Treatment Center, Riyadh 12713, Saudi Arabia
- Autism Center, Lotus Holistic Medical Center, Abu Dhabi 110281, United Arab Emirates
| |
Collapse
|
48
|
Chamtouri M, Merghni A, Salazar N, Redruello B, Gaddour N, Mastouri M, Arboleya S, de los Reyes-Gavilán CG. An Overview on Fecal Profiles of Amino Acids and Related Amino-Derived Compounds in Children with Autism Spectrum Disorder in Tunisia. Molecules 2023; 28:molecules28073269. [PMID: 37050030 PMCID: PMC10096484 DOI: 10.3390/molecules28073269] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 04/08/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental pathology characterized by the impairment of social interaction, difficulties in communication, and repetitive behaviors. Alterations in the metabolism of amino acids have been reported. We performed a chromatographic analysis of fecal amino acids, ammonium, biogenic amines, and gamma aminobutyric acid (GABA) in Tunisian autistic children from 4 to 10 years, and results were compared with their siblings (SIB) and children from the general population (GP). ASD presented significantly higher levels of fecal amino acids than SIB and GP; differences being more pronounced in younger (4–7 years) than in older (8–10 years) individuals whereas no changes were found for the remaining compounds. Lower levels of histidine were the only difference related with severe symptoms of autism (CARS scale). A linear discriminant analysis (LDA) based on fecal amino acid profiles clearly separated ASD, SIB, and GP at 4 to 7 years but not at more advanced age (8–10 years), evidencing more pronounced alterations in younger children. The relationship of fecal amino acids with autism needs deeper research integrating blood analytical parameters, brain metabolism, and intestinal microbiota. Fecal amino acids could be targeted for designing personalized diets to prevent or minimize cognitive impairments associated with ASD.
Collapse
Affiliation(s)
- Mariem Chamtouri
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain
- Laboratory of Transmissible Diseases and Biologically Active Substances LR99ES27, Faculty of Pharmacy, University of Monastir, Monastir 5000, Tunisia
| | - Abderrahmen Merghni
- Laboratory of Antimicrobial Resistance LR99ES09, Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis 1068, Tunisia
| | - Nuria Salazar
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain
- Diet, Microbiota, and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Begoña Redruello
- Scientific and Technical Services, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain
| | - Naoufel Gaddour
- Unit of Child Psychiatry, Monastir University Hospital, Monastir 5000, Tunisia
| | - Maha Mastouri
- Laboratory of Transmissible Diseases and Biologically Active Substances LR99ES27, Faculty of Pharmacy, University of Monastir, Monastir 5000, Tunisia
| | - Silvia Arboleya
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain
- Diet, Microbiota, and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Clara G. de los Reyes-Gavilán
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain
- Diet, Microbiota, and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| |
Collapse
|
49
|
Chakraborty P, Dey A, Gopalakrishnan AV, Swati K, Ojha S, Prakash A, Kumar D, Ambasta RK, Jha NK, Jha SK, Dewanjee S. Glutamatergic neurotransmission: A potential pharmacotherapeutic target for the treatment of cognitive disorders. Ageing Res Rev 2023; 85:101838. [PMID: 36610558 DOI: 10.1016/j.arr.2022.101838] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 12/27/2022] [Accepted: 12/29/2022] [Indexed: 01/06/2023]
Abstract
In the mammalian brain, glutamate is regarded to be the primary excitatory neurotransmitter due to its widespread distribution and wide range of metabolic functions. Glutamate plays key roles in regulating neurogenesis, synaptogenesis, neurite outgrowth, and neuron survival in the brain. Ionotropic and metabotropic glutamate receptors, neurotransmitters, neurotensin, neurosteroids, and others co-ordinately formulate a complex glutamatergic network in the brain that maintains optimal excitatory neurotransmission. Cognitive activities are potentially synchronized by the glutamatergic activities in the brain via restoring synaptic plasticity. Dysfunctional glutamate receptors and other glutamatergic components are responsible for the aberrant glutamatergic activity in the brain that cause cognitive impairments, loss of synaptic plasticity, and neuronal damage. Thus, controlling the brain's glutamatergic transmission and modifying glutamate receptor function could be a potential therapeutic strategy for cognitive disorders. Certain drugs that regulate glutamate receptor activities have shown therapeutic promise in improving cognitive functions in preclinical and clinical studies. However, several issues regarding precise functional information of glutamatergic activity are yet to be comprehensively understood. The present article discusses the scope of developing glutamatergic systems as prospective pharmacotherapeutic targets to treat cognitive disorders. Special attention has been given to recent developments, challenges, and future prospects.
Collapse
Affiliation(s)
- Pratik Chakraborty
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata 700073, West Bengal, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India
| | - Kumari Swati
- Department of Biotechnology, School of Life Science, Mahatma Gandhi Central University, Motihari, Bihar, India
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Anand Prakash
- Department of Biotechnology, School of Life Science, Mahatma Gandhi Central University, Motihari, Bihar, India
| | - Dhruv Kumar
- School of Health Sciences & Technology, UPES University, Dehradun, Uttarakhand 248007, India
| | - Rashmi K Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi 110042, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, UP, India; School of Bioengineering & Biosciences, Lovely Professional University, Phagwara, Punjab 144411, India.
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, UP, India; Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali 140413, India; Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun 248007, India.
| | - Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India.
| |
Collapse
|
50
|
Alijanpour S, Miryounesi M, Ghafouri-Fard S. The role of excitatory amino acid transporter 2 (EAAT2) in epilepsy and other neurological disorders. Metab Brain Dis 2023; 38:1-16. [PMID: 36173507 DOI: 10.1007/s11011-022-01091-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/15/2022] [Indexed: 02/03/2023]
Abstract
Glutamate is the major excitatory neurotransmitter in the central nervous system (CNS). Excitatory amino acid transporters (EAATs) have important roles in the uptake of glutamate and termination of glutamatergic transmission. Up to now, five EAAT isoforms (EAAT1-5) have been identified in mammals. The main focus of this review is EAAT2. This protein has an important role in the pathoetiology of epilepsy. De novo dominant mutations, as well as inherited recessive mutation in this gene, have been associated with epilepsy. Moreover, dysregulation of this protein is implicated in a range of neurological diseases, namely amyotrophic lateral sclerosis, alzheimer's disease, parkinson's disease, schizophrenia, epilepsy, and autism. In this review, we summarize the role of EAAT2 in epilepsy and other neurological disorders, then provide an overview of the therapeutic modulation of this protein.
Collapse
Affiliation(s)
- Sahar Alijanpour
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Miryounesi
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|