1
|
Zhang Y, Chen H, Cao J, Gao L, Jing Y. Maternal separation alters peripheral immune responses associated with IFN-γ and OT in mice. Peptides 2024; 182:171318. [PMID: 39486747 DOI: 10.1016/j.peptides.2024.171318] [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: 04/30/2024] [Revised: 10/03/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024]
Abstract
The co-evolution of social behavior and the immune system plays a critical role in individuals' adaptation to their environment. However, also need for further research on the key molecules that co-regulate social behavior and immunity. This study focused on neonatal mice that were separated from their mothers for 4 hours per day between the 6th and 16th day after birth. The results showed that these mice had lower plasma levels of IFN-γ and oxytocin, but higher levels of plasma glucocorticoids (GC), then impacting their social abilities. Additionally, maternal separation led to decreased levels of BDNF, IGF2, and CREB mRNAs in the hippocampus, while levels in the prefrontal cortex (PFC) remained unaffected. Maternal separation also resulted in increased levels of oxytocin and CRH mRNA in the hypothalamus, as well as an increase in CD45+ lymphocyte subsets in the meninges and choroid plexus (CP), with CD8+ lymphocytes in meninges and CD4+ lymphocytes in CP showing an increase. In IFN-γ-/- mice, a decrease in social preference was observed alongside lower plasma oxytocin levels. Moreover, IFN-γ-/- mice exhibited reduced numbers of oxytocin neurons in the paraventricular nucleus of the paraventricular nucleus of hypothalamus (PVN), decreased BDNF levels in the PFC and hippocampus, and alterations in CD45+ lymphocytes in CP and meninges, with an increase in CD8+ lymphocytes in meninges and CD4+ lymphocytes in CP. These findings highlight the immunological impact of social stress on IFN-γ regulation, suggesting that the immunomodulatory molecule IFN-γ may influence social behavior by affecting synaptic efficiency in brain regions such as the hippocampus and PFC, which are linked to oxytocin in the PVN.
Collapse
Affiliation(s)
- Yishu Zhang
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, PR China
| | - HaiChao Chen
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, PR China
| | - JiaXin Cao
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, PR China
| | - LiPing Gao
- Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, PR China
| | - YuHong Jing
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, PR China; Key Laboratory of Preclinical Study for New Drugs of Gansu province, Lanzhou University, Lanzhou, Gansu, PR China.
| |
Collapse
|
2
|
Moreno RJ, Amara R, Ashwood P. Toward a better understanding of T cell dysregulation in autism: An integrative review. Brain Behav Immun 2024:S0889-1591(24)00649-4. [PMID: 39378971 DOI: 10.1016/j.bbi.2024.10.009] [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: 05/23/2024] [Revised: 08/28/2024] [Accepted: 10/05/2024] [Indexed: 10/10/2024] Open
Abstract
Autism spectrum disorder (ASD) is a highly heterogeneous disorder characterized by impairments in social, communicative, and restrictive behaviors. Over the past 20 years, research has highlighted the role of the immune system in regulating neurodevelopment and behavior. In ASD, immune abnormalities are frequently observed, such as elevations in pro-inflammatory cytokines, alterations in immune cell frequencies, and dysregulated mechanisms of immune suppression. The adaptive immune system - the branch of the immune system conferring cellular immunity - may be involved in the etiology of ASD. Specifically, dysregulated T cell activity, characterized by altered cellular function and increased cytokine release, presence of inflammatory phenotypes and altered cellular signaling, has been consistently observed in several studies across multiple laboratories and geographic regions. Similarly, mechanisms regulating their activation are also disrupted. T cells at homeostasis coordinate the healthy development of the central nervous system (CNS) during early prenatal and postnatal development, and aid in CNS maintenance into adulthood. Thus, T cell dysregulation may play a role in neurodevelopment and the behavioral and cognitive manifestations observed in ASD. Outside of the CNS, aberrant T cell activity may also be responsible for the increased frequency of immune based conditions in the ASD population, such as allergies, gut inflammation and autoimmunity. In this review, we will discuss the current understanding of T cell biology in ASD and speculate on mechanisms behind their dysregulation. This review also evaluates how aberrant T cell biology affects gastrointestinal issues and behavior in the context of ASD.
Collapse
Affiliation(s)
- R J Moreno
- Department of Medical Microbiology and Immunology, UC Davis, CA, USA; The M.I.N.D. Institute, University of California at Davis, CA, USA
| | - R Amara
- Department of Medical Microbiology and Immunology, UC Davis, CA, USA; The M.I.N.D. Institute, University of California at Davis, CA, USA
| | - P Ashwood
- Department of Medical Microbiology and Immunology, UC Davis, CA, USA; The M.I.N.D. Institute, University of California at Davis, CA, USA.
| |
Collapse
|
3
|
Buxbaum Grice AS, Sloofman L, Levy T, Walker H, Ganesh G, Rodriguez de Los Santos M, Amini P, Buxbaum JD, Kolevzon A, Kostic A, Breen MS. Transient peripheral blood transcriptomic response to ketamine treatment in children with ADNP syndrome. Transl Psychiatry 2024; 14:307. [PMID: 39054328 PMCID: PMC11272924 DOI: 10.1038/s41398-024-03005-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 06/26/2024] [Accepted: 07/02/2024] [Indexed: 07/27/2024] Open
Abstract
Activity-dependent neuroprotective protein (ADNP) syndrome is a rare neurodevelopmental disorder resulting in intellectual disability, developmental delay and autism spectrum disorder (ASD) and is due to mutations in the ADNP gene. Ketamine treatment has emerged as a promising therapeutic option for ADNP syndrome, showing safety and apparent behavioral improvements in a first open label study. However, the molecular perturbations induced by ketamine remain poorly understood. Here, we investigated the longitudinal effect of ketamine on the blood transcriptome of 10 individuals with ADNP syndrome. Transcriptomic profiling was performed before and at multiple time points after a single low-dose intravenous ketamine infusion (0.5 mg/kg). We show that ketamine triggers immediate and profound gene expression alterations, with specific enrichment of monocyte-related expression patterns. These acute alterations encompass diverse signaling pathways and co-expression networks, implicating upregulation of immune and inflammatory-related processes and down-regulation of RNA processing mechanisms and metabolism. Notably, these changes exhibit a transient nature, returning to baseline levels 24 hours to 1 week after treatment. These findings enhance our understanding of ketamine's molecular effects and lay the groundwork for further research elucidating its specific cellular and molecular targets. Moreover, they contribute to the development of therapeutic strategies for ADNP syndrome and potentially, ASD more broadly.
Collapse
Affiliation(s)
- Ariela S Buxbaum Grice
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Laura Sloofman
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tess Levy
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hannah Walker
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gauri Ganesh
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miguel Rodriguez de Los Santos
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Pardis Amini
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joseph D Buxbaum
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexander Kolevzon
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ana Kostic
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael S Breen
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
4
|
Shao W, Su Y, Liu J, Liu Y, Zhao J, Fan X. Understanding the link between different types of maternal diabetes and the onset of autism spectrum disorders. DIABETES & METABOLISM 2024; 50:101543. [PMID: 38761920 DOI: 10.1016/j.diabet.2024.101543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 05/03/2024] [Accepted: 05/04/2024] [Indexed: 05/20/2024]
Abstract
Autism spectrum disorders (ASD) encompass a collection of neurodevelopmental disorders that exhibit impaired social interactions and repetitive stereotypic behaviors. Although the exact cause of these disorders remains unknown, it is widely accepted that both genetic and environmental factors contribute to their onset and progression. Recent studies have highlighted the potential negative impact of maternal diabetes on embryonic neurodevelopment, suggesting that intrauterine hyperglycemia could pose an additional risk to early brain development and contribute to the development of ASD. This paper presents a comprehensive analysis of the current research on the relationship between various forms of maternal diabetes, such as type 1 diabetes mellitus, type 2 diabetes mellitus, and gestational diabetes mellitus, and the likelihood of ASD in offspring. The study elucidates the potential mechanisms through which maternal hyperglycemia affects fetal development, involving metabolic hormones, immune dysregulation, heightened oxidative stress, and epigenetic alterations. The findings of this review offer valuable insights for potential preventive measures and evidence-based interventions targeting ASD.
Collapse
Affiliation(s)
- Wenyu Shao
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yichun Su
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jiayin Liu
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yulong Liu
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jinghui Zhao
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China.
| | - Xiaotang Fan
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
5
|
He H, Zhang X, He H, Xiao C, Xu G, Li L, Liu YE, Yang C, Zhou T, You Z, Zhang J. Priming of hippocampal microglia by IFN-γ/STAT1 pathway impairs social memory in mice. Int Immunopharmacol 2024; 134:112191. [PMID: 38759369 DOI: 10.1016/j.intimp.2024.112191] [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: 11/07/2023] [Revised: 03/29/2024] [Accepted: 04/29/2024] [Indexed: 05/19/2024]
Abstract
Social behavior is inextricably linked to the immune system. Although IFN-γ is known to be involved in social behavior, yet whether and how it encodes social memory remains unclear. In the current study, we injected with IFN-γ into the lateral ventricle of male C57BL/6J mice, and three-chamber social test was used to examine the effects of IFN-γ on their social preference and social memory. The morphology of microglia in the hippocampus, prelimbic cortex and amygdala was examined using immunohistochemistry, and the phenotype of microglia were examined using immunohistochemistry and enzyme-linked immunosorbent assays. The IFN-γ-injected mice were treated with lipopolysaccharide, and effects of IFN-γ on behavior and microglial responses were evaluated. STAT1 pathway and microglia-neuron interactions were examined in vivo or in vitro using western blotting and immunohistochemistry. Finally, we use STAT1 inhibitor or minocycline to evaluated the role of STAT1 in mediating the microglial priming and effects of primed microglia in IFN-γ-induced social dysfunction. We demonstrated that 500 ng of IFN-γ injection results in significant decrease in social index and social novelty recognition index, and induces microglial priming in hippocampus, characterized by enlarged cell bodies, shortened branches, increased expression of CD68, CD86, CD74, CD11b, CD11c, CD47, IL-33, IL-1β, IL-6 and iNOS, and decreased expression of MCR1, Arg-1, IGF-1 and BDNF. This microglia subpopulation is more sensitive to LPS challenge, which characterized by more significant morphological changes and inflammatory responses, as well as induced increased sickness behaviors in mice. IFN-γ upregulated pSTAT1 and STAT1 and promoted the nuclear translocation of STAT1 in the hippocampal microglia and in the primary microglia. Giving minocycline or STAT1 inhibitor fludarabin blocked the priming of hippocampal microglia induced by IFN-γ, ameliorated the dysfunction in hippocampal microglia-neuron interactions and synapse pruning by microglia, thereby improving social memory deficits in IFN-γ injected mice. IFN-γ initiates STAT1 pathway to induce priming of hippocampal microglia, thereby disrupts hippocampal microglia-neuron interactions and neural circuit link to social memory. Blocking STAT1 pathway or inhibiting microglial priming may be strategies to reduce the effects of IFN-γ on social behavior.
Collapse
Affiliation(s)
- Haili He
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Xiaomei Zhang
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Hui He
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Chenghong Xiao
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Gaojie Xu
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Liangyuan Li
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Yu-E Liu
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Chengyan Yang
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Tao Zhou
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Zili You
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China.
| | - Jinqiang Zhang
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| |
Collapse
|
6
|
Long J, Dang H, Su W, Moneruzzaman M, Zhang H. Interactions between circulating inflammatory factors and autism spectrum disorder: a bidirectional Mendelian randomization study in European population. Front Immunol 2024; 15:1370276. [PMID: 38742104 PMCID: PMC11089225 DOI: 10.3389/fimmu.2024.1370276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 04/16/2024] [Indexed: 05/16/2024] Open
Abstract
Background Extensive observational studies have reported an association between inflammatory factors and autism spectrum disorder (ASD), but their causal relationships remain unclear. This study aims to offer deeper insight into causal relationships between circulating inflammatory factors and ASD. Methods Two-sample bidirectional Mendelian randomization (MR) analysis method was used in this study. The genetic variation of 91 circulating inflammatory factors was obtained from the genome-wide association study (GWAS) database of European ancestry. The germline GWAS summary data for ASD were also obtained (18,381 ASD cases and 27,969 controls). Single nucleotide polymorphisms robustly associated with the 91 inflammatory factors were used as instrumental variables. The random-effects inverse-variance weighted method was used as the primary analysis, and the Bonferroni correction for multiple comparisons was applied. Sensitivity tests were carried out to assess the validity of the causal relationship. Results The forward MR analysis results suggest that levels of sulfotransferase 1A1, natural killer cell receptor 2B4, T-cell surface glycoprotein CD5, Fms-related tyrosine kinase 3 ligand, and tumor necrosis factor-related apoptosis-inducing ligand are positively associated with the occurrence of ASD, while levels of interleukin-7, interleukin-2 receptor subunit beta, and interleukin-2 are inversely associated with the occurrence of ASD. In addition, matrix metalloproteinase-10, caspase 8, tumor necrosis factor-related activation-induced cytokine, and C-C motif chemokine 19 were considered downstream consequences of ASD. Conclusion This MR study identified additional inflammatory factors in patients with ASD relative to previous studies, and raised a possibility of ASD-caused immune abnormalities. These identified inflammatory factors may be potential biomarkers of immunologic dysfunction in ASD.
Collapse
Affiliation(s)
- Junzi Long
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Neurorehabilitation, China Rehabilitation Research Center, Beijing, China
- Division of Brain Sciences, Changping Laboratory, Beijing, China
| | - Hui Dang
- Department of Neurorehabilitation, China Rehabilitation Research Center, Beijing, China
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Wenlong Su
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Neurorehabilitation, China Rehabilitation Research Center, Beijing, China
| | - Md. Moneruzzaman
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Neurorehabilitation, China Rehabilitation Research Center, Beijing, China
| | - Hao Zhang
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Neurorehabilitation, China Rehabilitation Research Center, Beijing, China
- Division of Brain Sciences, Changping Laboratory, Beijing, China
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
7
|
Hassan A, Mohsen R, Rezk A, Bangay G, Rijo P, Soliman MFM, G. A. Hablas M, Swidan KAK, Mohammed TS, Zoair MA, Mohamed AAK, Abdalrhman TI, Abdel-aleem Desoky AM, Mohamed DD, Mohamed DD, Abd El Maksoud AI, Mohamed AF. Enhancement of Vitamin C's Protective Effect against Thimerosal-Induced Neurotoxicity in the Cerebral Cortex of Wistar Albino Rats: An In Vivo and Computational Study. ACS OMEGA 2024; 9:8973-8984. [PMID: 38434836 PMCID: PMC10905602 DOI: 10.1021/acsomega.3c07239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/29/2023] [Accepted: 01/09/2024] [Indexed: 03/05/2024]
Abstract
Vitamin C was examined to ameliorate the neurotoxicity of thimerosal (THIM) in an animal model (Wistar albino rats). In our work, oxidative and antioxidative biomarkers such as SOD, LPO, and GSH were investigated at various doses of THIM with or without concurrent vitamin C administration. Furthermore, the adverse effects of THIM on hepatic tissue and cerebral cortex morphology were examined in the absence or presence of associated vitamin C administration. Also, we studied the effect of vitamin C on the metallothionein isoforms (MT-1, MT-2, and MT-3) in silico and in vivo using the RT-PCR assay. The results showed that the antioxidant biomarker was reduced as the THIM dose was raised and vice versa. THIM-associated vitamin C reduced the adverse effects of the THIM dose. The computation studies demonstrated that vitamin C has a lower ΔG of -4.9 kcal/mol compared to -4.1 kcal/mol for THIM to bind to the MT-2 protein, which demonstrated that vitamin C has a greater ability to bind with MT-2 than THIM. This is due to multiple hydrogen bonds that exist between vitamin C and MT-2 residues Lys31, Gln23, Cys24, and Cys29, and the sodium ion represents key stabilizing interactions. Hydrogen bonds involve electrostatic interactions between hydrogen atom donors (e.g., hydroxyl groups) and acceptors (e.g., carbonyl oxygens). The distances between heavy atoms are typically 2.5-3.5 Å. H-bonds provide directed, high-affinity interactions to anchor the ligand to the binding site. The five H-bonds formed by vitamin C allow it to form a stable complex with MT, while THIM can form two H-bonds with Gln23 and Cys24. This provides less stabilization in the binding pocket, contributing to the lower affinity compared to vitamin C. The histopathological morphologies in hepatic tissue displayed an expansion in the portal tract and the hepatocytes surrounding the portal tract, including apoptosis, binucleation, and karyomegaly. The histopathological morphologies in the brain tissue revealed a significant decrease in the number of Purkinje cells due to THIM toxicity. Interestingly, THIM toxicity was associated with hemorrhage and astrogliosis. Both intracellular and vasogenic edema appeared as the concentrations of THIM rose. Finally, vitamin C ameliorated the adverse effect on the cerebral cortex in Wistar albino rats.
Collapse
Affiliation(s)
- Amr Hassan
- Department
of Bioinformatics, Genetic Engineering and Biotechnology Research
Institute (GEBRI), University of Sadat City, Sadat 32897, Egypt
| | - Reham Mohsen
- College
of Biotechnology, October University for Modern Science and Arts (MSA), University Giza, Giza 11456, Egypt
| | - Ahmed Rezk
- College
of Biotechnology, October University for Modern Science and Arts (MSA), University Giza, Giza 11456, Egypt
| | - Gabrielle Bangay
- CBIOS—Research
Center for Biosciences & Health Technologies, Universidade Lusófona de Humanidades e Tecnologias, Lisboa 1749-024, Portugal
- Facultad
de Farmacia, Departamento de Ciencias Biomédicas (Área
de Farmacología; Nuevos agentes antitumorales, Acción
tóxica sobre células leucémicas), Universidad de Alcalá de Henares, Ctra. Madrid-Barcelona km. 33, Alcalá de Henares 600 28805, Madrid, Espana
| | - Patrícia Rijo
- CBIOS—Research
Center for Biosciences & Health Technologies, Universidade Lusófona de Humanidades e Tecnologias, Lisboa 1749-024, Portugal
- Instituto
de Investigação do Medicamento (iMed.ULisboa), Faculdade
de Farmácia, Universidade de Lisboa, Lisbon 1749-024, Portugal
| | - Mona F. M. Soliman
- Department
of Histology and Cell Biology, Faculty Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Mohamed G. A. Hablas
- Department
of Histology and Cell Biology, Faculty of Medicine, Suez University, Suez 43221, Egypt
| | | | - Tahseen S. Mohammed
- Department
of Public Health and Community Medicine, Faculty of Medicine, Al-Azhar University, Cairo 11884, Egypt
| | - Mohammad A. Zoair
- Department
of Physiology, Faculty of Medicine, Al-Azhar
University, Cairo 11884, Egypt
| | - Abir A. Khalil Mohamed
- Department
of Zoology, Faculty of Science, Girls Branch, Al-Azhar University, Cairo 11884, Egypt
| | - Tamer I. Abdalrhman
- Department of Histology, Faculty of Medicine, Al-Azhar University, Assiut 71524, Egypt
| | | | - Dalia D. Mohamed
- Department of Industrial Biotechnology,
Genetic Engineering and Biotechnology
Research Institute (GEBRI), University of
Sadat City, Sadat 32897, Egypt
| | - Doaa D. Mohamed
- Department of Industrial Biotechnology,
Genetic Engineering and Biotechnology
Research Institute (GEBRI), University of
Sadat City, Sadat 32897, Egypt
| | - Ahmed I. Abd El Maksoud
- Department of Industrial Biotechnology,
Genetic Engineering and Biotechnology
Research Institute (GEBRI), University of
Sadat City, Sadat 32897, Egypt
| | - Aly F. Mohamed
- Holding
Company for Vaccine and Sera Production (VACSERA), Giza 22311, Egypt
| |
Collapse
|
8
|
Cui J, Zhai Z, Wang S, Song X, Qiu T, Yu L, Zhai Q, Zhang H. The role and impact of abnormal vitamin levels in autism spectrum disorders. Food Funct 2024; 15:1099-1115. [PMID: 38221882 DOI: 10.1039/d3fo03735e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
The prevalence of autism spectrum disorder (ASD), a neurodevelopmental disorder with a predominance of social behavioral disorders, has increased dramatically in various countries in recent decades. The interplay between genetic and environmental factors is believed to underlie ASD pathogenesis. Recent analyses have shown that abnormal vitamin levels in early life are associated with an increased risk of autism. As essential substances for growth and development, vitamins have been shown to have significant benefits for the nervous and immune systems. However, it is unknown whether certain vitamin types influence the emergence or manifestation of ASD symptoms. Several studies have focused on vitamin levels in children with autism, and neurotypical children have provided different insights into the types of vitamins and their intake. Here, we review the mechanisms and significance of several vitamins (A, B, C, D, E, and K) that are closely associated with the development of ASD in order to prevent, mitigate, and treat ASD. Efforts have been made to discover and develop new indicators for nutritional assessment of children with ASD to play a greater role in the early detection of ASD and therapeutic remission after diagnosis.
Collapse
Affiliation(s)
- Jingjing Cui
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214002, China.
- Department of child health care, Wuxi Maternity and Child Health Care Hospital, Affiliated Women's Hositipal of Jiangnan University, Wuxi, Jiangsu, 214002, China.
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, 214122, China.
| | - Zidan Zhai
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214002, China.
- Department of child health care, Wuxi Maternity and Child Health Care Hospital, Affiliated Women's Hositipal of Jiangnan University, Wuxi, Jiangsu, 214002, China.
| | - Shumin Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China.
| | - Xiaoyue Song
- Department of Toxicology, School of Public Health, Anhui Medical University/Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei 230032, Anhui, China.
| | - Ting Qiu
- Department of child health care, Wuxi Maternity and Child Health Care Hospital, Affiliated Women's Hositipal of Jiangnan University, Wuxi, Jiangsu, 214002, China.
| | - Leilei Yu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, 214122, China.
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, 214122, China.
| | - Heng Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214002, China.
- Department of child health care, Wuxi Maternity and Child Health Care Hospital, Affiliated Women's Hositipal of Jiangnan University, Wuxi, Jiangsu, 214002, China.
- Department of Toxicology, School of Public Health, Anhui Medical University/Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei 230032, Anhui, China.
| |
Collapse
|
9
|
Tamayo JM, Osman HC, Schwartzer JJ, Ashwood P. The influence of asthma on neuroinflammation and neurodevelopment: From epidemiology to basic models. Brain Behav Immun 2024; 116:218-228. [PMID: 38070621 DOI: 10.1016/j.bbi.2023.12.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 11/08/2023] [Accepted: 12/04/2023] [Indexed: 12/18/2023] Open
Abstract
Asthma is a highly heterogeneous inflammatory disease that can have a significant effect on both the respiratory system and central nervous system. Population based studies and animal models have found asthma to be comorbid with a number of neurological conditions, including depression, anxiety, and neurodevelopmental disorders. In addition, maternal asthma during pregnancy has been associated with neurodevelopmental disorders in the offspring, such as autism spectrum disorders and attention deficit hyperactivity disorder. In this article, we review the most current epidemiological studies of asthma that identify links to neurological conditions, both as it relates to individuals that suffer from asthma and the impacts asthma during pregnancy may have on offspring neurodevelopment. We also discuss the relevant animal models investigating these links, address the gaps in knowledge, and explore the potential future directions in this field.
Collapse
Affiliation(s)
- Juan M Tamayo
- Department of Medical Microbiology and Immunology, and the M.I.N.D. Institute, University of California at Davis, CA 95817, USA
| | - Hadley C Osman
- Department of Medical Microbiology and Immunology, and the M.I.N.D. Institute, University of California at Davis, CA 95817, USA
| | - Jared J Schwartzer
- Program in Neuroscience and Behavior, Department of Psychology and Education, Mount Holyoke College, 50 College Street, South Hadley, MA 01075, USA
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, and the M.I.N.D. Institute, University of California at Davis, CA 95817, USA.
| |
Collapse
|
10
|
Grice ASB, Sloofman L, Levy T, Walker H, Ganesh G, de Los Santos MR, Armini P, Buxbaum JD, Kolevzon A, Kostic A, Breen MS. Transient peripheral blood transcriptomic response to ketamine treatment in children with ADNP syndrome. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.29.24301949. [PMID: 38352457 PMCID: PMC10863029 DOI: 10.1101/2024.01.29.24301949] [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: 02/25/2024]
Abstract
Activity-dependent neuroprotective protein (ADNP) syndrome is a rare neurodevelopmental disorder resulting in intellectual disability, developmental delay and autism spectrum disorder (ASD) and is due to mutations in the ADNP gene. Ketamine treatment has emerged as a promising therapeutic option for ADNP syndrome, showing safety and apparent behavioral improvements in a first open label study. However, the molecular perturbations induced by ketamine remain poorly understood. Here, we investigated the longitudinal effect of ketamine on the blood transcriptome of 10 individuals with ADNP syndrome. Transcriptomic profiling was performed before and at multiple time points after a single low-dose intravenous ketamine infusion (0.5mg/kg). We show that ketamine triggers immediate and profound gene expression alterations, with specific enrichment of monocyte-related expression patterns. These acute alterations encompass diverse signaling pathways and co-expression networks, implicating up-regulation of immune and inflammatory-related processes and down-regulation of RNA processing mechanisms and metabolism. Notably, these changes exhibit a transient nature, returning to baseline levels 24 hours to 1 week after treatment. These findings enhance our understanding of ketamine's molecular effects and lay the groundwork for further research elucidating its specific cellular and molecular targets. Moreover, they contribute to the development of therapeutic strategies for ADNP syndrome and potentially, ASD more broadly.
Collapse
Affiliation(s)
- Ariela S Buxbaum Grice
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Laura Sloofman
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tess Levy
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hannah Walker
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gauri Ganesh
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miguel Rodriguez de Los Santos
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Pardis Armini
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joseph D Buxbaum
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexander Kolevzon
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ana Kostic
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael S Breen
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
11
|
Bäckström F, Ahl M, Wickham J, Ekdahl CT. Reduced epilepsy development in synapsin 2 knockout mice with autistic behavior following early systemic treatment with interleukin-6 receptor antibody. Epilepsy Res 2023; 191:107114. [PMID: 36870094 DOI: 10.1016/j.eplepsyres.2023.107114] [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: 12/15/2022] [Revised: 02/09/2023] [Accepted: 02/23/2023] [Indexed: 02/27/2023]
Abstract
BACKGROUND Individuals with autism spectrum disorder (ASD) have an increased risk of developing epilepsy. Both ASD and epilepsy have been associated with increased levels of immune factors in the blood, including the proinflammatory cytokine interleukin 6 (IL-6). Mice lacking the synapsin 2 gene (Syn2 KO) exhibit ASD-like behavior and develop epileptic seizures. Their brains display neuroinflammatory changes including elevated IL-6 levels. We aimed to investigate the effect of systemic IL-6 receptor antibody (IL-6R ab) treatment on seizure development and frequency in Syn2 KO mice. MATERIAL AND METHODS Weekly systemic (i.p.) injections of IL-6R ab or saline were given to Syn2 KO mice starting either early in life at 1 month of age, before seizure debut or at 3 months of age, directly after seizure debut and continued for 4 or 2 months, respectively. Seizures were provoked by handling the mice three times per week. The neuroinflammatory response and synaptic protein levels in the brain were determined by ELISA, immunohistochemistry and western blots. In an additional group of Syn2 KO mice, with IL-6R ab treatment early in life, ASD-related behavioral tests including social interaction and repetitive self-grooming, as well as cognitive memory and depressive-/anxiety-like tests, and actigraphy measurements of circadian sleep-awake rhythm were analyzed. RESULTS The IL-6R ab treatment reduced seizure development and frequency in Syn2 KO mice when initiated before, but not after, seizure debut. However, early treatment did not reverse the neuroinflammatory response or the imbalance in synaptic protein levels in the brain previously reported in Syn2 KO mice. The treatment did not affect social interaction, performance in memory, depressive-/anxiety-like tests or the sleep-awake rhythm of Syn2 KO mice. CONCLUSION These findings suggest the involvement of IL-6 receptor signaling during epilepsy development in Syn2 KO mice, without significant alterations of the immune reaction in the brain, and independently of cognitive performance, mood and circadian sleep-awake rhythm.
Collapse
Affiliation(s)
- Filip Bäckström
- Inflammation and Stem Cell Therapy Group, Division of Clinical Neurophysiology, Department of Clinical Sciences, Lund University, Lund, Sweden; Lund Epilepsy Center, Department of Clinical Sciences, Lund University, Lund, Sweden; Translational Neurogenetics Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Matilda Ahl
- Inflammation and Stem Cell Therapy Group, Division of Clinical Neurophysiology, Department of Clinical Sciences, Lund University, Lund, Sweden; Lund Epilepsy Center, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Jenny Wickham
- Inflammation and Stem Cell Therapy Group, Division of Clinical Neurophysiology, Department of Clinical Sciences, Lund University, Lund, Sweden; Lund Epilepsy Center, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Christine T Ekdahl
- Inflammation and Stem Cell Therapy Group, Division of Clinical Neurophysiology, Department of Clinical Sciences, Lund University, Lund, Sweden; Lund Epilepsy Center, Department of Clinical Sciences, Lund University, Lund, Sweden.
| |
Collapse
|
12
|
Indika NLR, Frye RE, Rossignol DA, Owens SC, Senarathne UD, Grabrucker AM, Perera R, Engelen MPKJ, Deutz NEP. The Rationale for Vitamin, Mineral, and Cofactor Treatment in the Precision Medical Care of Autism Spectrum Disorder. J Pers Med 2023; 13:252. [PMID: 36836486 PMCID: PMC9964499 DOI: 10.3390/jpm13020252] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/27/2023] [Accepted: 01/28/2023] [Indexed: 01/31/2023] Open
Abstract
Children with autism spectrum disorder may exhibit nutritional deficiencies due to reduced intake, genetic variants, autoantibodies interfering with vitamin transport, and the accumulation of toxic compounds that consume vitamins. Importantly, vitamins and metal ions are essential for several metabolic pathways and for neurotransmitter functioning. The therapeutic benefits of supplementing vitamins, minerals (Zinc, Magnesium, Molybdenum, and Selenium), and other cofactors (coenzyme Q10, alpha-lipoic acid, and tetrahydrobiopterin) are mediated through their cofactor as well as non-cofactor functions. Interestingly, some vitamins can be safely administered at levels far above the dose typically used to correct the deficiency and exert effects beyond their functional role as enzyme cofactors. Moreover, the interrelationships between these nutrients can be leveraged to obtain synergistic effects using combinations. The present review discusses the current evidence for using vitamins, minerals, and cofactors in autism spectrum disorder, the rationale behind their use, and the prospects for future use.
Collapse
Affiliation(s)
- Neluwa-Liyanage R. Indika
- Department of Biochemistry, Faculty of Medical Sciences, University of Sri Jayewardenepura, Nugegoda 10250, Sri Lanka
| | - Richard E. Frye
- Autism Discovery and Research Foundation, Phoenix, AZ 85050, USA
- Rossignol Medical Center, Phoenix, AZ 85050, USA
| | - Daniel A. Rossignol
- Rossignol Medical Center, Phoenix, AZ 85050, USA
- Rossignol Medical Center, Aliso Viejo, CA 92656, USA
| | - Susan C. Owens
- Autism Oxalate Project at the Autism Research Institute, San Diego, CA 92116, USA
| | - Udara D. Senarathne
- Department of Biochemistry, Faculty of Medical Sciences, University of Sri Jayewardenepura, Nugegoda 10250, Sri Lanka
| | - Andreas M. Grabrucker
- Department of Biological Sciences, University of Limerick, V94 T9PX Limerick, Ireland
- Bernal Institute, University of Limerick, V94 T9PX Limerick, Ireland
- Health Research Institute (HRI), University of Limerick, V94 T9PX Limerick, Ireland
| | - Rasika Perera
- Department of Biochemistry, Faculty of Medical Sciences, University of Sri Jayewardenepura, Nugegoda 10250, Sri Lanka
| | - Marielle P. K. J. Engelen
- Center for Translational Research in Aging & Longevity, Texas A&M University, College Station, TX 77843, USA
| | - Nicolaas E. P. Deutz
- Center for Translational Research in Aging & Longevity, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
13
|
Breach MR, Lenz KM. Sex Differences in Neurodevelopmental Disorders: A Key Role for the Immune System. Curr Top Behav Neurosci 2023; 62:165-206. [PMID: 35435643 PMCID: PMC10286778 DOI: 10.1007/7854_2022_308] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Sex differences are prominent defining features of neurodevelopmental disorders. Understanding the sex biases in these disorders can shed light on mechanisms leading to relative risk and resilience for the disorders, as well as more broadly advance our understanding of how sex differences may relate to brain development. The prevalence of neurodevelopmental disorders is increasing, and the two most common neurodevelopmental disorders, Autism Spectrum Disorder (ASD) and Attention-Deficit/Hyperactivity Disorder (ADHD) exhibit male-biases in prevalence rates and sex differences in symptomology. While the causes of neurodevelopmental disorders and their sex differences remain to be fully understood, increasing evidence suggests that the immune system plays a critical role in shaping development. In this chapter we discuss sex differences in prevalence and symptomology of ASD and ADHD, review sexual differentiation and immune regulation of neurodevelopment, and discuss findings from human and rodent studies of immune dysregulation and perinatal immune perturbation as they relate to potential mechanisms underlying neurodevelopmental disorders. This chapter will give an overview of how understanding sex differences in neuroimmune function in the context of neurodevelopmental disorders could lend insight into their etiologies and better treatment strategies.
Collapse
Affiliation(s)
- Michaela R Breach
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Kathryn M Lenz
- Department of Psychology, The Ohio State University, Columbus, OH, USA.
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA.
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
14
|
Association of complement component 4 with neuroimmune abnormalities in the subventricular zone in schizophrenia and autism spectrum disorders. Neurobiol Dis 2022; 173:105840. [PMID: 35995342 PMCID: PMC9582995 DOI: 10.1016/j.nbd.2022.105840] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 08/12/2022] [Accepted: 08/14/2022] [Indexed: 02/06/2023] Open
Abstract
An early inflammatory insult is the most recognized risk factor associated with neurodevelopmental psychiatric disorders, even more so than genetic variants. Notably, complement component 4 (C4), a molecule involved in inflammatory responses, has been strongly associated with schizophrenia (SZ) and its role in other neurodevelopmental disorders, such as autism (ASD), is an area of active investigation. However, while C4 in SZ has been implicated in the context of synaptic pruning, little is known about its neuroinflammatory role. The subventricular zone (SVZ) is a region heavily involved in neurodevelopment and neuroimmune interactions through the lifespan; thus, it is a region wherein C4 may play a vital role in disease pathology. Using in situ hybridization with radioactive riboprobes and RNAscope, we identified robust astrocytic expression of C4 in the SVZ and in the septum pellucidum. C4 was also expressed in ependyma, neurons, and Ki67+ progenitor cells. Examination of mRNA levels showed elevated C4 in both ASD and SZ, with higher expression in SZ compared to controls. Targeted transcriptomic analysis of inflammatory pathways revealed a strong association of complement system genes with SZ, and to a lesser extent, ASD, as well as generalized immune dysregulation without a strong association with known infectious pathways. Analysis of differentially expressed genes (DEGs) showed that ASD DEGs were enriched in adaptive immune system functions such as Th cell differentiation, while SZ DEGs were enriched in innate immune system functions, including NF-κB and toll like receptor signaling. Moreover, the number of Ki67+ cells was significantly higher in ASD compared to SZ and controls. Taken together, these results support a role for C4 into inflammatory-neuroimmune dysregulation observed in SZ and ASD pathology.
Collapse
|
15
|
Gevezova M, Sarafian V, Anderson G, Maes M. Inflammation and Mitochondrial Dysfunction in Autism Spectrum Disorder. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 19:320-333. [PMID: 32600237 DOI: 10.2174/1871527319666200628015039] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 03/30/2020] [Accepted: 05/01/2020] [Indexed: 12/11/2022]
Abstract
Autism Spectrum Disorders (ASD) is a severe childhood psychiatric condition with an array of cognitive, language and social impairments that can significantly impact family life. ASD is classically characterized by reduced communication skills and social interactions, with limitations imposed by repetitive patterns of behavior, interests, and activities. The pathophysiology of ASD is thought to arise from complex interactions between environmental and genetic factors within the context of individual development. A growing body of research has raised the possibility of identifying the aetiological causes of the disorder. This review highlights the roles of immune-inflammatory pathways, nitro-oxidative stress and mitochondrial dysfunctions in ASD pathogenesis and symptom severity. The role of NK-cells, T helper, T regulatory and B-cells, coupled with increased inflammatory cytokines, lowered levels of immune-regulatory cytokines, and increased autoantibodies and microglial activation is elucidated. It is proposed that alterations in mitochondrial activity and nitrooxidative stress are intimately associated with activated immune-inflammatory pathways. Future research should determine as to whether the mitochondria, immune-inflammatory activity and nitrooxidative stress changes in ASD affect the development of amygdala-frontal cortex interactions. A number of treatment implications may arise, including prevention-orientated prenatal interventions, treatment of pregnant women with vitamin D, and sodium butyrate. Treatments of ASD children and adults with probiotics, sodium butyrate and butyrate-inducing diets, antipurinergic therapy with suramin, melatonin, oxytocin and taurine are also discussed.
Collapse
Affiliation(s)
- Maria Gevezova
- Department of Medical Biology, Faculty of Medicine, Medical University-Plovdiv, Plovdiv, Bulgaria,Research Institute at Medical University-Plovdiv, Plovdiv, Bulgaria
| | - Victoria Sarafian
- Department of Medical Biology, Faculty of Medicine, Medical University-Plovdiv, Plovdiv, Bulgaria,Research Institute at Medical University-Plovdiv, Plovdiv, Bulgaria
| | | | - Michael Maes
- Department of Medical Biology, Faculty of Medicine, Medical University-Plovdiv, Plovdiv, Bulgaria,Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand,IMPACT Strategic Research Center, Deakin University, Geelong, Australia
| |
Collapse
|
16
|
Ahadullah, Yau SY, Lu HX, Lee TMC, Guo H, Chan CCH. PM 2.5 as a potential risk factor for autism spectrum disorder: Its possible link to neuroinflammation, oxidative stress and changes in gene expression. Neurosci Biobehav Rev 2021; 128:534-548. [PMID: 34216652 DOI: 10.1016/j.neubiorev.2021.06.043] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/10/2021] [Accepted: 06/29/2021] [Indexed: 10/21/2022]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by behavioral deficits including impairments in social communication, social interaction, and repetitive behaviors. Because the etiology of ASD is still largely unknown, there is no cure for ASD thus far. Although it has been established that genetic components play a vital role in ASD development, the influence of epigenetic regulation induced by environmental factors could also contribute to ASD susceptibility. Accumulated evidence has suggested that exposure to atmospheric particulate matter (PM) in polluted air could affect neurodevelopment, thus possibly leading to ASD. Particles with a size of 2.5 μm (PM2.5) or less have been shown to have negative effects on human health, and could be linked to ASD symptoms in children. This review summarizes evidence from clinical and animal studies to demonstrate the possible linkage between PM2.5 exposure and the incidence of ASD in children. An attempt was made to explore the possible mechanisms of this linkage, including changes of gene expression, oxidative stress and neuroinflammation induced by PM2.5 exposure.
Collapse
Affiliation(s)
- Ahadullah
- Department of Rehabilitation Sciences, Hong Kong Polytechnic University, Hong Kong, China
| | - Suk-Yu Yau
- Department of Rehabilitation Sciences, Hong Kong Polytechnic University, Hong Kong, China; Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou 510515, China.
| | - Hao-Xian Lu
- Department of Civil and Environmental Engineering, Hong Kong Polytechnic University, Hong Kong, China
| | - Tatia M C Lee
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China; Laboratory of Neuropsychology and Human Neuroscience, The University of Hong Kong, Hong Kong, China
| | - Hai Guo
- Department of Civil and Environmental Engineering, Hong Kong Polytechnic University, Hong Kong, China.
| | - Chetwyn C H Chan
- Department of Psychology, The Education University of Hong Kong, Tai Po, Hong Kong, China
| |
Collapse
|
17
|
Jhun M, Panwar A, Cordner R, Irvin DK, Veiga L, Yeager N, Pechnick RN, Schubloom H, Black KL, Wheeler CJ. CD103 Deficiency Promotes Autism (ASD) and Attention-Deficit Hyperactivity Disorder (ADHD) Behavioral Spectra and Reduces Age-Related Cognitive Decline. Front Neurol 2021; 11:557269. [PMID: 33424735 PMCID: PMC7786306 DOI: 10.3389/fneur.2020.557269] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 12/03/2020] [Indexed: 11/17/2022] Open
Abstract
The incidence of autism spectrum disorders (ASD) and attention deficit hyperactivity disorder (ADHD), which frequently co-occur, are both rising. The causes of ASD and ADHD remain elusive, even as both appear to involve perturbation of the gut-brain-immune axis. CD103 is an integrin and E-cadherin receptor most prominently expressed on CD8 T cells that reside in gut, brain, and other tissues. CD103 deficiency is well-known to impair gut immunity and resident T cell function, but it's impact on neurodevelopmental disorders has not been examined. We show here that CD8 T cells influence neural progenitor cell function, and that CD103 modulates this impact both directly and potentially by controlling CD8 levels in brain. CD103 knockout (CD103KO) mice exhibited a variety of behavioral abnormalities, including superior cognitive performance coupled with repetitive behavior, aversion to novelty and social impairment in females, with hyperactivity with delayed learning in males. Brain protein markers in female and male CD103KOs coincided with known aspects of ASD and ADHD in humans, respectively. Surprisingly, CD103 deficiency also decreased age-related cognitive decline in both sexes, albeit by distinct means. Together, our findings reveal a novel role for CD103 in brain developmental function, and identify it as a unique factor linking ASD and ADHD etiology. Our data also introduce a new animal model of combined ASD and ADHD with associated cognitive benefits, and reveal potential therapeutic targets for these disorders and age-related cognitive decline.
Collapse
Affiliation(s)
- Michelle Jhun
- Department of Neurosurgery, Cedars-Sinai Medical Center, Maxine Dunitz Neurosurgical Institute, Los Angeles, CA, United States
| | - Akanksha Panwar
- Department of Neurosurgery, Cedars-Sinai Medical Center, Maxine Dunitz Neurosurgical Institute, Los Angeles, CA, United States
| | - Ryan Cordner
- Department of Neurosurgery, Cedars-Sinai Medical Center, Maxine Dunitz Neurosurgical Institute, Los Angeles, CA, United States.,Department Biomedical & Translational Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Dwain K Irvin
- Department of Neurosurgery, Cedars-Sinai Medical Center, Maxine Dunitz Neurosurgical Institute, Los Angeles, CA, United States.,StemVax Therapeutics, Chesterland, OH, United States
| | - Lucia Veiga
- Department of Neurosurgery, Cedars-Sinai Medical Center, Maxine Dunitz Neurosurgical Institute, Los Angeles, CA, United States
| | - Nicole Yeager
- Department Biomedical & Translational Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Robert N Pechnick
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States
| | - Hanna Schubloom
- Department of Neurosurgery, Cedars-Sinai Medical Center, Maxine Dunitz Neurosurgical Institute, Los Angeles, CA, United States
| | - Keith L Black
- Department of Neurosurgery, Cedars-Sinai Medical Center, Maxine Dunitz Neurosurgical Institute, Los Angeles, CA, United States
| | - Christopher J Wheeler
- Department of Neurosurgery, Cedars-Sinai Medical Center, Maxine Dunitz Neurosurgical Institute, Los Angeles, CA, United States.,Society for Brain Mapping & Therapeutics, Brain Mapping Foundation, Santa Monica, CA, United States.,T-Neuro Pharma, Inc., Albuquerque, NM, United States
| |
Collapse
|
18
|
Perinatal Exposure to Diesel Exhaust-Origin Secondary Organic Aerosol Induces Autism-Like Behavior in Rats. Int J Mol Sci 2021; 22:ijms22020538. [PMID: 33430368 PMCID: PMC7828068 DOI: 10.3390/ijms22020538] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/04/2021] [Accepted: 01/04/2021] [Indexed: 02/06/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by impaired social communication, poor social interactions, and repetitive behaviors. We aimed to examine autism-like behaviors and related gene expressions in rats exposed to diesel exhaust (DE)-origin secondary organic aerosol (DE-SOA) perinatally. Sprague–Dawley pregnant rats were exposed to clean air (control), DE, and DE-SOA in the exposure chamber from gestational day 14 to postnatal day 21. Behavioral phenotypes of ASD were investigated in 10~13-week-old offspring using a three-chambered social behavior test, social dominance tube test, and marble burying test. Prefrontal cortex was collected to examine molecular analyses including neurological and immunological markers and glutamate concentration, using RT-PCR and ELISA methods. DE-SOA-exposed male and female rats showed poor sociability and social novelty preference, socially dominant behavior, and increased repetitive behavior. Serotonin receptor (5-HT(5B)) and brain-derived neurotrophic factor (BDNF) mRNAs were downregulated whereas interleukin 1 β (IL-β) and heme oxygenase 1 (HO-1) mRNAs were upregulated in the prefrontal cortex of male and female rats exposed to DE-SOA. Glutamate concentration was also increased significantly in DE-SOA-exposed male and female rats. Our results indicate that perinatal exposure to DE-SOA may induce autism-like behavior by modulating molecules such as neurological and immunological markers in rats.
Collapse
|
19
|
Uddin MN, Yao Y, Manley K, Lawrence DA. Development, phenotypes of immune cells in BTBR T +Itpr3 tf/J mice. Cell Immunol 2020; 358:104223. [PMID: 33137646 DOI: 10.1016/j.cellimm.2020.104223] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 09/18/2020] [Accepted: 09/20/2020] [Indexed: 01/01/2023]
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental condition that is characterized by a lack of social interaction, decreased verbal and non-verbal communication skills, and stereotyped repetitive behavior. There is strong evidence that a dysregulated immune response may influence neurodevelopment and thus may have a role in the development of ASD. This study focuses on the characterization of immune cell phenotypes in the BTBR T+Itpr3tf/J (BTBR) mouse strain, a widely used animal model for autism research. Our study demonstrated that BTBR mice have a different immune profile compared to C57BL/6J (B6) mice, which do not display ASD-like characteristics. Thymic cells of BTBR mice have more single positive (SP) CD4+ and CD8+ T cells and fewer double positive (DP) T cells than B6 mice. The development of T cells is increased in BTBR mice with regard to the double negative (DN4) population being much higher in BTBR mice. The spleens and blood of BTBR mice also have more T helper type 1 (Th1), T helper type 2 (Th2) and T regulatory (Treg) cells compared to B6 mice. Aire expression in the thymus and spleen of BTBR mice compared to B6 mice was equivalent and lower, respectively. The mature natural killer (NK) innate immune cell population in blood and spleen is lower in BTBR than B6 mice; NK cell development is blocked prior to the double positive (DN) CD11b+CD27+ stage in BTBR mice. Since BTBR mice have more CD4+ T cells and elevated numbers of Th1 (T-bet+) and Th2 (GATA3+) cells, their low defense against pathogen may be explained by the lower number of NK cells and the significantly lower Th1 to Th2 ratio. The elevated number of plasma cells and autoantibodies of BTBR mice may be due to less presence and function of splenic AIRE.
Collapse
Affiliation(s)
- Mohammad Nizam Uddin
- Wadsworth Center, New York State Department of Health, Albany, NY, United States
| | - Yunyi Yao
- Wadsworth Center, New York State Department of Health, Albany, NY, United States
| | - Kevin Manley
- Wadsworth Center, New York State Department of Health, Albany, NY, United States
| | - David A Lawrence
- Wadsworth Center, New York State Department of Health, Albany, NY, United States; University at Albany School of Public Health, Rensselaer, NY, United States.
| |
Collapse
|
20
|
Analyzing the Potential Biological Determinants of Autism Spectrum Disorder: From Neuroinflammation to the Kynurenine Pathway. Brain Sci 2020; 10:brainsci10090631. [PMID: 32932826 PMCID: PMC7563403 DOI: 10.3390/brainsci10090631] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/31/2020] [Accepted: 09/10/2020] [Indexed: 12/22/2022] Open
Abstract
Autism Spectrum Disorder (ASD) etiopathogenesis is still unclear and no effective preventive and treatment measures have been identified. Research has focused on the potential role of neuroinflammation and the Kynurenine pathway; here we review the nature of these interactions. Pre-natal or neonatal infections would induce microglial activation, with secondary consequences on behavior, cognition and neurotransmitter networks. Peripherally, higher levels of pro-inflammatory cytokines and anti-brain antibodies have been identified. Increased frequency of autoimmune diseases, allergies, and recurring infections have been demonstrated both in autistic patients and in their relatives. Genetic studies have also identified some important polymorphisms in chromosome loci related to the human leukocyte antigen (HLA) system. The persistence of immune-inflammatory deregulation would lead to mitochondrial dysfunction and oxidative stress, creating a self-sustaining cytotoxic loop. Chronic inflammation activates the Kynurenine pathway with an increase in neurotoxic metabolites and excitotoxicity, causing long-term changes in the glutamatergic system, trophic support and synaptic function. Furthermore, overactivation of the Kynurenine branch induces depletion of melatonin and serotonin, worsening ASD symptoms. Thus, in genetically predisposed subjects, aberrant neurodevelopment may derive from a complex interplay between inflammatory processes, mitochondrial dysfunction, oxidative stress and Kynurenine pathway overexpression. To validate this hypothesis a new translational research approach is necessary.
Collapse
|
21
|
Matta SM, Moore Z, Walker FR, Hill-Yardin EL, Crack PJ. An altered glial phenotype in the NL3 R451C mouse model of autism. Sci Rep 2020; 10:14492. [PMID: 32879325 PMCID: PMC7468159 DOI: 10.1038/s41598-020-71171-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 06/26/2020] [Indexed: 12/26/2022] Open
Abstract
Autism Spectrum Disorder (ASD; autism) is a neurodevelopmental disorder characterised by deficits in social communication, and restricted and/or repetitive behaviours. While the precise pathophysiologies are unclear, increasing evidence supports a role for dysregulated neuroinflammation in the brain with potential effects on synapse function. Here, we studied characteristics of microglia and astrocytes in the Neuroligin-3 (NL3R451C) mouse model of autism since these cell types are involved in regulating both immune and synapse function. We observed increased microglial density in the dentate gyrus (DG) of NL3R451C mice without morphological differences. In contrast, WT and NL3R451C mice had similar astrocyte density but astrocyte branch length, the number of branch points, as well as cell radius and area were reduced in the DG of NL3R451C mice. Because retraction of astrocytic processes has been linked to altered synaptic transmission and dendrite formation, we assessed for regional changes in pre- and postsynaptic protein expression in the cortex, striatum and cerebellum in NL3R451C mice. NL3R451C mice showed increased striatal postsynaptic density 95 (PSD-95) protein levels and decreased cortical expression of synaptosomal-associated protein 25 (SNAP-25). These changes could contribute to dysregulated neurotransmission and cognition deficits previously reported in these mice.
Collapse
Affiliation(s)
- Samantha M Matta
- Department of Pharmacology and Therapeutics, The University of Melbourne, Grattan St, Parkville, VIC, Australia.,School of Health & Biomedical Sciences, RMIT University, 225-245 Clements Drive, Bundoora, VIC, Australia
| | - Zachery Moore
- Department of Pharmacology and Therapeutics, The University of Melbourne, Grattan St, Parkville, VIC, Australia
| | - Frederick Rohan Walker
- School of Biomedical Sciences and Pharmacy, University of Newcastle, University Drive, Callaghan, NSW, Australia.,Hunter Medical Research Institute, Locked Bag 1000, New Lambton, NSW, Australia
| | - Elisa L Hill-Yardin
- School of Health & Biomedical Sciences, RMIT University, 225-245 Clements Drive, Bundoora, VIC, Australia.,Department of Physiology, The University of Melbourne, Grattan St, Parkville, VIC, Australia
| | - Peter J Crack
- Department of Pharmacology and Therapeutics, The University of Melbourne, Grattan St, Parkville, VIC, Australia.
| |
Collapse
|
22
|
Lee KM, Hawi ZH, Parkington HC, Parish CL, Kumar PV, Polo JM, Bellgrove MA, Tong J. The application of human pluripotent stem cells to model the neuronal and glial components of neurodevelopmental disorders. Mol Psychiatry 2020; 25:368-378. [PMID: 31455859 DOI: 10.1038/s41380-019-0495-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 05/19/2019] [Accepted: 06/24/2019] [Indexed: 12/24/2022]
Abstract
Cellular models of neurodevelopmental disorders provide a valuable experimental system to uncover disease mechanisms and novel therapeutic strategies. The ability of induced pluripotent stem cells (iPSCs) to generate diverse brain cell types offers great potential to model several neurodevelopmental disorders. Further patient-derived iPSCs have the unique genetic and molecular signature of the affected individuals, which allows researchers to address limitations of transgenic behavioural models, as well as generate hypothesis-driven models to study disorder-relevant phenotypes at a cellular level. In this article, we review the extant literature that has used iPSC-based modelling to understand the neuronal and glial contributions to neurodevelopmental disorders including autism spectrum disorder (ASD), Rett syndrome, bipolar disorder (BP), and schizophrenia. For instance, several molecular candidates have been shown to influence cellular phenotypes in three-dimensional iPSC-based models of ASD patients. Delays in differentiation of astrocytes and morphological changes of neurons are associated with Rett syndrome. In the case of bipolar disorders and schizophrenia, patient-derived models helped to identify cellular phenotypes associated with neuronal deficits (e.g., excitability) and mutation-specific abnormalities in oligodendrocytes (e.g., CSPG4). Further we provide a critical review of the current limitations of this field and provide methodological suggestions to enhance future modelling efforts of neurodevelopmental disorders. Future developments in experimental design and methodology of disease modelling represent an exciting new avenue relevant to neurodevelopmental disorders.
Collapse
Affiliation(s)
- K M Lee
- Turner Institute for Brain and Mental Health and the School of Psychological Sciences, Monash University, Melbourne, Australia
| | - Z H Hawi
- Turner Institute for Brain and Mental Health and the School of Psychological Sciences, Monash University, Melbourne, Australia
| | - H C Parkington
- Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - C L Parish
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - P V Kumar
- Turner Institute for Brain and Mental Health and the School of Psychological Sciences, Monash University, Melbourne, Australia
| | - J M Polo
- Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - M A Bellgrove
- Turner Institute for Brain and Mental Health and the School of Psychological Sciences, Monash University, Melbourne, Australia
| | - J Tong
- Turner Institute for Brain and Mental Health and the School of Psychological Sciences, Monash University, Melbourne, Australia.
| |
Collapse
|
23
|
Rose DR, Yang H, Careaga M, Angkustsiri K, Van de Water J, Ashwood P. T cell populations in children with autism spectrum disorder and co-morbid gastrointestinal symptoms. Brain Behav Immun Health 2020; 2:100042. [PMID: 34589832 PMCID: PMC8474588 DOI: 10.1016/j.bbih.2020.100042] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 01/17/2020] [Accepted: 01/24/2020] [Indexed: 01/09/2023] Open
Abstract
Children with ASD are more likely to experience gastrointestinal (GI) symptoms than typically-developed children. Numerous studies have reported immune abnormalities and inflammatory profiles in the majority of individuals with ASD. Immune dysfunction is often hypothesized as a driving factor in many GI diseases and it has been suggested that it is more apparent in children with ASD that exhibit GI symptoms. In this study we sought to characterize peripheral T cell subsets in children with and without GI symptoms, compared to healthy typically-developing children. Peripheral blood mononuclear cells were isolated from participants, who were categorized into three groups: children with ASD who experience GI symptoms (n = 14), children with ASD who do not experience GI symptoms (n = 10) and typically-developing children who do not experience GI symptoms (n = 15). In order to be included in the GI group, GI symptoms such as diarrhea, constipation, and/or pain while defecating, had to be present in the child regularly for the past 6 months; likewise, in order to be placed in the no GI groups, bowel movements could not include the above symptoms present throughout development. Cells were assessed for surface markers and intracellular cytokines to identify T cell populations. Children with ASD and GI symptoms displayed elevated TH17 populations (0.757% ± 0.313% compared to 0.297% ± 0.197), while children with ASD who did not experience GI symptoms showed increased frequency of TH2 populations (2.02% ± 1.08% compared to 1.01% ± 0.58%). Both ASD groups showed evidence of reduced gut homing regulatory T cell populations compared to typically developing children (ASDGI:1.93% ± 0.75% and ASDNoGI:1.85% ± 0.89 compared to 2.93% ± 1.16%). Children with ASD may have deficits in immune regulation that lead to differential inflammatory T cell subsets that could be linked to associated co-morbidities.
Collapse
Affiliation(s)
- Destanie R. Rose
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
- MIND Institute, University of California Davis, Davis, CA, USA
| | - Houa Yang
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
- MIND Institute, University of California Davis, Davis, CA, USA
| | - Milo Careaga
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
- MIND Institute, University of California Davis, Davis, CA, USA
| | - Kathy Angkustsiri
- MIND Institute, University of California Davis, Davis, CA, USA
- Department of Pediatrics, University of California Davis, CA, USA
- Children’s Center for Environmental Health, University of California Davis, CA, USA
| | - Judy Van de Water
- MIND Institute, University of California Davis, Davis, CA, USA
- Children’s Center for Environmental Health, University of California Davis, CA, USA
- Division of Rheumatology, Allergy and Clinical Immunology, University of California Davis, CA, USA
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
- MIND Institute, University of California Davis, Davis, CA, USA
- Public Health Sciences, University of California Davis, CA, USA
| |
Collapse
|
24
|
Alzghoul L. Role of Vitamin D in Autism Spectrum Disorder. Curr Pharm Des 2020; 25:4357-4367. [DOI: 10.2174/1381612825666191122092215] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 11/15/2019] [Indexed: 12/19/2022]
Abstract
:
Autism spectrum disorder (ASD) is a pervasive developmental disorder with heterogeneous etiology.
Vitamin D can function as a fat-soluble vitamin as well as a hormone, and can exert its effect through both genomic
and non-genomic mechanisms. In the last decades, several studies have examined the relationship between
vitamin D levels and ASD. These studies demonstrated that low vitamin D status in early development has been
hypothesized as an environmental risk factor for ASD. Both in vivo and in vitro studies have demonstrated that
vitamin D deficiency in early life can alter brain development, dysregulates neurotransmitter balance in the brain,
decreases body and brain antioxidant ability, and alters the immune system in ways that resemble pathological
features commonly seen in ASD. In this review, we focused on the association between vitamin D and ASD. In
addition, the above-mentioned mechanisms of action that link vitamin D deficiency with ASD were also discussed.
Finally, clinical trials of vitamin D supplementation treatment of ASD have also been discussed.
Collapse
Affiliation(s)
- Loai Alzghoul
- Department of Physiology and Biochemistry, School of Medicine, The University of Jordan, Amman, Jordan
| |
Collapse
|
25
|
Deb S, Phukan BC, Dutta A, Paul R, Bhattacharya P, Manivasagam T, Thenmozhi AJ, Babu CS, Essa MM, Borah A. Natural Products and Their Therapeutic Effect on Autism Spectrum Disorder. ADVANCES IN NEUROBIOLOGY 2020; 24:601-614. [PMID: 32006376 DOI: 10.1007/978-3-030-30402-7_22] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Autism is a complex neurodevelopmental disorder that is evident in early childhood and can persist throughout the entire life. The disease is basically characterized by hurdles in social interaction where the individuals demonstrate repetitive and stereotyped interests or patterns of behavior. A wide number of neuroanatomical studies with autistic patients revealed alterations in brain development which lead to diverse cellular and anatomical processes including atypical neurogenesis, neuronal migration, maturation, differentiation, and degeneration. Special education programs, speech and language therapy, have been employed for the amelioration of behavioral deficits in autism. Although commonly prescribed antidepressants, antipsychotics, anticonvulsants, and stimulants have revealed satisfactory responses in autistic individuals, adverse side effects and increased risk of several other complications including obesity, dyslipidemia, diabetes mellitus, thyroid disorders, etc. have compelled the researchers to turn their attention toward herbal remedies. Alternative approaches with natural compounds are on continuous clinical trial to confirm their efficacy and to understand their potential in autism treatment. This chapter aims to cover the major plant-based natural products which hold promising outcomes in the field of reliable therapeutic interventions for autism.
Collapse
Affiliation(s)
- Satarupa Deb
- Cellular and Molecular Neurobiology Laboratory, Department of Life Science and Bioinformatics, Assam University, Silchar, Assam, India
| | - Banashree Chetia Phukan
- Cellular and Molecular Neurobiology Laboratory, Department of Life Science and Bioinformatics, Assam University, Silchar, Assam, India
| | - Ankumoni Dutta
- Cellular and Molecular Neurobiology Laboratory, Department of Life Science and Bioinformatics, Assam University, Silchar, Assam, India
| | - Rajib Paul
- Department of Zoology, Pandit Deendayal Upadhyaya Adarsha Mahavidyalaya (PDUAM), Karimganj, Assam, India
| | - Pallab Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India
| | - Thamilarasan Manivasagam
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Chidambaram, Tamil Nadu, India
| | - Arokiasamy Justin Thenmozhi
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Chidambaram, Tamil Nadu, India
| | - Chidambaram Saravana Babu
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSSAHER), Mysuru, Karnataka, India
| | - Musthafa Mohamed Essa
- Department of Food Science and Nutrition, CAMS, Sultan Qaboos University, Muscat, Oman.,Ageing and Dementia Research Group, Sultan Qaboos University, Muscat, Oman.,Food and Brain Research Foundation, Chennai, Tamil Nadu, India
| | - Anupom Borah
- Cellular and Molecular Neurobiology Laboratory, Department of Life Science and Bioinformatics, Assam University, Silchar, Assam, India.
| |
Collapse
|
26
|
Afroz KF, Alviña K. Maternal elevated salt consumption and the development of autism spectrum disorder in the offspring. J Neuroinflammation 2019; 16:265. [PMID: 31837704 PMCID: PMC6911292 DOI: 10.1186/s12974-019-1666-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 11/27/2019] [Indexed: 01/15/2023] Open
Abstract
Autism spectrum disorder (ASD) is a prevalent neurodevelopmental condition with no known etiology or cure. Several possible contributing factors, both genetic and environmental, are being actively investigated. Amongst these, maternal immune dysregulation has been identified as potentially involved in promoting ASD in the offspring. Indeed, ASD-like behaviors have been observed in studies using the maternal immune activation mouse model. Furthermore, recent studies have shed light on maternal dietary habits and their impact on the gut microbiome as factors possibly facilitating ASD. However, most of these studies have been limited to the effects of high fat and/or high sugar. More recent data, however, have shown that elevated salt consumption has a significant effect on the immune system and gut microbiome, often resulting in gut dysbiosis and induction of pro-inflammatory pathways. Specifically, high salt alters the gut microbiome and induces the differentiation of T helper-17 cells that produce pro-inflammatory cytokines such as interleukin-17 and interleukin-23. Moreover, elevated salt can also reduce the differentiation of regulatory T cells that help maintaining a balanced immune system. While in the innate immune system, high salt can cause over activation of M1 pro-inflammatory macrophages and downregulation of M2 regulatory macrophages. These changes to the immune system are alarming because excessive consumption of salt is a documented worldwide problem. Thus, in this review, we discuss recent findings on high salt intake, gut microbiome, and immune system dysregulation while proposing a hypothesis to link maternal overconsumption of salt and children’s ASD.
Collapse
Affiliation(s)
- Kazi Farhana Afroz
- Department of Biological Sciences, Texas Tech University, 2901 Main St. Room #05, Biology Building, Lubbock, TX, 79409, USA
| | - Karina Alviña
- Department of Biological Sciences, Texas Tech University, 2901 Main St. Room #05, Biology Building, Lubbock, TX, 79409, USA. .,Department of Neuroscience, University of Florida, 1149 Newell Drive, Room L1-100, Gainesville, FL, 32611, USA.
| |
Collapse
|
27
|
Saffari A, Arno M, Nasser E, Ronald A, Wong CCY, Schalkwyk LC, Mill J, Dudbridge F, Meaburn EL. RNA sequencing of identical twins discordant for autism reveals blood-based signatures implicating immune and transcriptional dysregulation. Mol Autism 2019; 10:38. [PMID: 31719968 PMCID: PMC6839145 DOI: 10.1186/s13229-019-0285-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 09/01/2019] [Indexed: 11/13/2022] Open
Abstract
Background A gap exists in our mechanistic understanding of how genetic and environmental risk factors converge at the molecular level to result in the emergence of autism symptoms. We compared blood-based gene expression signatures in identical twins concordant and discordant for autism spectrum condition (ASC) to differentiate genetic and environmentally driven transcription differences, and establish convergent evidence for biological mechanisms involved in ASC. Methods Genome-wide gene expression data were generated using RNA-seq on whole blood samples taken from 16 pairs of monozygotic (MZ) twins and seven twin pair members (39 individuals in total), who had been assessed for ASC and autism traits at age 12. Differential expression (DE) analyses were performed between (a) affected and unaffected subjects (N = 36) and (b) within discordant ASC MZ twin pairs (total N = 11) to identify environmental-driven DE. Gene set enrichment and pathway testing was performed on DE gene lists. Finally, an integrative analysis using DNA methylation data aimed to identify genes with consistent evidence for altered regulation in cis. Results In the discordant twin analysis, three genes showed evidence for DE at FDR < 10%: IGHG4, EVI2A and SNORD15B. In the case-control analysis, four DE genes were identified at FDR < 10% including IGHG4, PRR13P5, DEPDC1B, and ZNF501. We find enrichment for DE of genes curated in the SFARI human gene database. Pathways showing evidence of enrichment included those related to immune cell signalling and immune response, transcriptional control and cell cycle/proliferation. Integrative methylomic and transcriptomic analysis identified a number of genes showing suggestive evidence for cis dysregulation. Limitations Identical twins stably discordant for ASC are rare, and as such the sample size was limited and constrained to the use of peripheral blood tissue for transcriptomic and methylomic profiling. Given these primary limitations, we focused on transcript-level analysis. Conclusions Using a cohort of ASC discordant and concordant MZ twins, we add to the growing body of transcriptomic-based evidence for an immune-based component in the molecular aetiology of ASC. Whilst the sample size was limited, the study demonstrates the utility of the discordant MZ twin design combined with multi-omics integration for maximising the potential to identify disease-associated molecular signals.
Collapse
Affiliation(s)
- Ayden Saffari
- 1Department of Non-communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
- 2Centre for Brain and Cognitive Development, Department of Psychological Sciences, Birkbeck, University of London, London, UK
| | - Matt Arno
- 3Edinburgh Genomics, University of Edinburgh, Edinburgh, Scotland UK
- 4King's Genomics Centre, King's College London, London, UK
| | - Eric Nasser
- 4King's Genomics Centre, King's College London, London, UK
| | - Angelica Ronald
- 2Centre for Brain and Cognitive Development, Department of Psychological Sciences, Birkbeck, University of London, London, UK
| | - Chloe C Y Wong
- 5Social Genetic and Developmental Psychology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | | | - Jonathan Mill
- 7University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Frank Dudbridge
- 1Department of Non-communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
- 8Department of Health Sciences, University of Leicester, Leicester, UK
| | - Emma L Meaburn
- 2Centre for Brain and Cognitive Development, Department of Psychological Sciences, Birkbeck, University of London, London, UK
| |
Collapse
|
28
|
Hamed NO, Osman MA, Elkhawad AO, Bjørklund G, Qasem H, Zayed N, El-Ansary A. Determination of neuroinflammatory biomarkers in autistic and neurotypical Saudi children. Metab Brain Dis 2019; 34:1049-1060. [PMID: 31147808 DOI: 10.1007/s11011-019-00420-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 04/17/2019] [Indexed: 12/16/2022]
Abstract
To identify neuroinflammatory biomarkers in patients with various severity of autism spectrum disorder (ASD) increases the insight about the pathogenesis and pathophysiology of this neurodevelopmental disorder. The aim of the present study was to analyze the levels in plasma of TGFβ2, Heat shock protein 70 (HSP70), and hematopoietic prostaglandin D2 synthase (H-PGDS) in Saudi ASD children and healthy age-matched neurotypical controls. Also, it was in the present study examined the correlation among these neuroinflammatory biomarkers and the sensory deficit exhibited by the ASD children. Blood samples from 38 Saudi children with ASD and 32 age-matched neurotypical controls were withdrawn after an overnight fast. For the blood taking 3 mL EDTA containing blood collection tubes was used. The samples were centrifuged for 20 min (4 °C; 3000×g) directly after the blood sampling. The harvested plasma was used for in vitro quantification of TGF-β2, HSP70, and H-PGDS by using the sandwich enzyme immunoassay. Receiver operating characteristic (ROC) analysis and predictiveness curves showed that each of TGF-β2, HSP70 or H-PGDS alone could not be used as a predictive neuroinflammatory biomarker for ASD. However, when TGF-β2 and HSP70 were combined in one ROC curve, the AUC was increased to an appreciable value that makes them together robust predictors of variation between the ASD and neurotypical control groups. Overall, it was in the present study found significant differences for TGF-β2 and HSP70 when the ASD and neurotypical control groups were compared, independently of the sensory deficit level. In conclusion, the present study highlights the usefulness of TGF-β2, HSP70, and H-PGDS as diagnostic tools to differentiate between ASD and neurotypical control children, but not among subgroups of ASD children exhibiting different severity levels of sensory dysfunction. The presented data also suggest the effectiveness of ROC as a powerful statistical tool, which precisely can measure a combined effect of neuroinflammatory biomarkers intended for diagnostic purposes.
Collapse
Affiliation(s)
- Najat O Hamed
- University of Medical Sciences and Technology, Khartoum, Sudan
- Almaaref Colleges for Sciences and Technology, Riyadh, Saudi Arabia
| | - Mohamed A Osman
- Kirkwood College, Cedar Rapids, IA, USA
- Sudan Medical and Scientific Research Institute, Khartoum, Sudan
| | - Abdalla O Elkhawad
- University of Medical Sciences and Technology, Khartoum, Sudan
- Sudan Medical and Scientific Research Institute, Khartoum, Sudan
| | - Geir Bjørklund
- Council for Nutritional and Environmental Medicine, Toften 24, 8610, Mo i Rana, Norway.
| | - Hanan Qasem
- Autism Research and Treatment Center, Riyadh, Saudi Arabia
| | - Naima Zayed
- Therapeutic Chemistry Department, National Research Centre, Dokki, Cairo, Egypt
| | - Afaf El-Ansary
- Autism Research and Treatment Center, Riyadh, Saudi Arabia
- Shiek Al-Amodi Autism Research Chair, King Saud University, Riyadh, Saudi Arabia
- Therapeutic Chemistry Department, National Research Centre, Dokki, Cairo, Egypt
- Central Laboratory, Female Center for Scientific and Medical Studies, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
29
|
Matta SM, Hill-Yardin EL, Crack PJ. The influence of neuroinflammation in Autism Spectrum Disorder. Brain Behav Immun 2019; 79:75-90. [PMID: 31029798 DOI: 10.1016/j.bbi.2019.04.037] [Citation(s) in RCA: 207] [Impact Index Per Article: 41.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 04/15/2019] [Accepted: 04/24/2019] [Indexed: 12/12/2022] Open
Abstract
Autism Spectrum Disorder (ASD) is a neurodevelopmental disorder characterised by deficits in social communication and restricted or repetitive behaviours. The clinical presentation of ASD is highly variable and diagnosis is based on the presence of impaired social communication and repetitive and/or restricted behaviours. Although the precise pathophysiologies underlying ASD are unclear, growing evidence supports a role for dysregulated neuroinflammation. The potential involvement of microglia and astrocytes reactive to inflammatory stimuli in ASD has generated much interest due to their varied roles including in mounting an immune response and regulating synaptic function. Increased numbers of reactive microglial and astrocytes in both ASD postmortem tissue and animal models have been reported. Whether dysregulation of glial subtypes exacerbates alterations in neural connectivity in the brain of autistic patients is not well explored. A role for the gut-brain axis involving microbial-immune-neuronal cross talk is also a growing area of neuroinflammation research. Greater understanding of these interactions under patho/physiological conditions and the identification of consistent immune profile abnormalities can potentially lead to more reliable diagnostic measures and treatments in ASD.
Collapse
Affiliation(s)
- Samantha M Matta
- Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Elisa L Hill-Yardin
- School of Health & Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia; Department of Physiology, The University of Melbourne, Parkville, VIC 3010, Australia.
| | - Peter J Crack
- Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
30
|
Barfi S, Narges C, Pouretemad HR, Poortahmasebi V, Norouzi M, Farahmand M, Yahyapour Y, Ghorbani S, Ghalichi L, Ofoghi H, Jazayeri SM. Measurement of serum hepatitis B surface antibody levels in Iranian autistic children and evaluation of immunological memory after booster dose injection in comparison with controls. J Med Virol 2019; 91:1272-1278. [PMID: 30761573 DOI: 10.1002/jmv.25429] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 01/29/2019] [Accepted: 02/07/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Responsiveness to hepatitis B vaccine among patients with autism spectrum disorders (ASD) has not been evaluated worldwide. We aimed to determine the anti-HBs antibody duration in autistic and healthy children few years after primary vaccination and evaluate their immunological memory against hepatitis B virus (HBV) vaccine with booster dose administration. METHODS One hundred seven and 147 HBsAg-negative children from ASD and normal population were recruited, respectively. HBV seromarkers (HBc-Ab, HBsAg, and HBs-Ab) were assessed and subsequently, molecular tests were used on all the subjects. A booster dose of vaccine was injected for those who showed low levels (<10 mIU/mL) of anti-HBs and their antibody levels was measured 4 weeks later. RESULTS The mean ages of ASD and control groups were 7.14 ± 2.42 and 8.68 ± 1.96, respectively. Seven (6.5%) of the ASD group were positive for anti-HBc and one child was positive for occult hepatitis B infection (HBsAg negative, HBV DNA positive). In ASD, 54 (50.4%) and 53 (49.6%) had adequate (>10 mIU/mL) and low anti-HBs levels, respectively. Among control group, 74 (50.4%) and 73 (49.6%) had sufficient and low antibody levels, respectively. After injection of a booster dose for all children with low antibody, 100% of ASD and 92% (59 of 64) of control pupils contained >10 mIU/mL of antibody, respectively. In both the groups, the HBs-Ab titer increased similarly in response to the booster injection (P < 0.05). CONCLUSION Despite previous investigations regarding immune impairment in individuals with autism, the immune system of these individuals was able to manage the hepatitis B vaccine challenge.
Collapse
Affiliation(s)
- Shahram Barfi
- Department of Virology, School of Public Health, International Campus, Tehran University of Medical Sciences, Tehran, Iran
| | - Chimeh Narges
- Rehabilitation Clinic for Children with Special needs (RCCS), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Reza Pouretemad
- Clinical Neurophysiology Section, Department of Psychological Medicine, Institute of Psychiatry, Shahid Beheshti University, Tehran, Iran
| | - Vahdat Poortahmasebi
- Department of Virology, Tehran University of Medical Science, Tehran, Iran.,Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Norouzi
- Department of Virology, Tehran University of Medical Science, Tehran, Iran.,Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Farahmand
- Department of Virology, Tehran University of Medical Science, Tehran, Iran
| | - Yousef Yahyapour
- Infectious Diseases and Tropical Medicine Research Center, Babol University of Medical Sciences, Babol, Iran
| | - Saeid Ghorbani
- Iran University of Medical Sciences Tehrān, Tehran, Iran
| | - Leila Ghalichi
- Mental Health Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Haleh Ofoghi
- Rehabilitation Clinic for Children with Special needs (RCCS), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Mohammad Jazayeri
- Department of Virology, Tehran University of Medical Science, Tehran, Iran.,Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
31
|
Alterations in the MicroRNA of the Blood of Autism Spectrum Disorder Patients: Effects on Epigenetic Regulation and Potential Biomarkers. Behav Sci (Basel) 2018; 8:bs8080075. [PMID: 30111726 PMCID: PMC6115946 DOI: 10.3390/bs8080075] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 07/31/2018] [Accepted: 08/11/2018] [Indexed: 12/27/2022] Open
Abstract
Aims: Autism spectrum disorder (ASD) refers to a group of heterogeneous brain-based neurodevelopmental disorders with different levels of symptom severity. Given the challenges, the clinical diagnosis of ASD is based on information gained from interviews with patients’ parents. The heterogeneous pathogenesis of this disorder appears to be driven by genetic and environmental interactions, which also plays a vital role in predisposing individuals to ASD with different commitment levels. In recent years, it has been proposed that epigenetic modifications directly contribute to the pathogenesis of several neurodevelopmental disorders, such as ASD. The microRNAs (miRNAs) comprises a species of short noncoding RNA that regulate gene expression post-transcriptionally and have an essential functional role in the brain, particularly in neuronal plasticity and neuronal development, and could be involved in ASD pathophysiology. The aim of this study is to evaluate the expression of blood miRNA in correlation with clinical findings in patients with ASD, and to find possible biomarkers for the disorder. Results: From a total of 26 miRNA studied, seven were significantly altered in ASD patients, when compared to the control group: miR34c-5p, miR92a-2-5p, miR-145-5p and miR199a-5p were up-regulated and miR27a-3p, miR19-b-1-5p and miR193a-5p were down-regulated in ASD patients. Discussion: The main targets of these miRNAs are involved in immunological developmental, immune response and protein synthesis at transcriptional and translational levels. The up-regulation of both miR-199a-5p and miR92a-2a and down-regulation of miR-193a and miR-27a was observed in AD patients, and may in turn affect the SIRT1, HDAC2, and PI3K/Akt-TSC:mTOR signaling pathways. Furthermore, MeCP2 is a target of miR-199a-5p, and is involved in Rett Syndrome (RTT), which possibly explains the autistic phenotype in male patients with this syndrome.
Collapse
|
32
|
Gładysz D, Krzywdzińska A, Hozyasz KK. Immune Abnormalities in Autism Spectrum Disorder-Could They Hold Promise for Causative Treatment? Mol Neurobiol 2018; 55:6387-6435. [PMID: 29307081 PMCID: PMC6061181 DOI: 10.1007/s12035-017-0822-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 11/05/2017] [Indexed: 12/15/2022]
Abstract
Autism spectrum disorders (ASD) are characterized by impairments in language and communication development, social behavior, and the occurrence of stereotypic patterns of behavior and interests. Despite substantial speculation about causes of ASD, its exact etiology remains unknown. Recent studies highlight a link between immune dysfunction and behavioral traits. Various immune anomalies, including humoral and cellular immunity along with abnormalities at the molecular level, have been reported. There is evidence of altered immune function both in cerebrospinal fluid and peripheral blood. Several studies hypothesize a role for neuroinflammation in ASD and are supported by brain tissue and cerebrospinal fluid analysis, as well as evidence of microglial activation. It has been shown that immune abnormalities occur in a substantial number of individuals with ASD. Identifying subgroups with immune system dysregulation and linking specific cellular immunophenotypes to different symptoms would be key to defining a group of patients with immune abnormalities as a major etiology underlying behavioral symptoms. These determinations would provide the opportunity to investigate causative treatments for a defined patient group that may specifically benefit from such an approach. This review summarizes recent insights into immune system dysfunction in individuals with ASD and discusses the potential implications for future therapies.
Collapse
Affiliation(s)
- Dominika Gładysz
- Department of Pediatrics, Institute of Mother and Child, Warsaw, Poland
| | | | - Kamil K Hozyasz
- Department of Pediatrics, Institute of Mother and Child, Warsaw, Poland.
| |
Collapse
|
33
|
Eftekharian MM, Ghafouri-Fard S, Noroozi R, Omrani MD, Arsang-jang S, Ganji M, Gharzi V, Noroozi H, Komaki A, Mazdeh M, Taheri M. Cytokine profile in autistic patients. Cytokine 2018; 108:120-126. [DOI: 10.1016/j.cyto.2018.03.034] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 12/24/2017] [Accepted: 03/22/2018] [Indexed: 12/19/2022]
|
34
|
Xu G, Snetselaar LG, Jing J, Liu B, Strathearn L, Bao W. Association of Food Allergy and Other Allergic Conditions With Autism Spectrum Disorder in Children. JAMA Netw Open 2018; 1:e180279. [PMID: 30646068 PMCID: PMC6324407 DOI: 10.1001/jamanetworkopen.2018.0279] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
IMPORTANCE The prevalence of autism spectrum disorder (ASD) in US children has increased during the past decades. Immunologic dysfunction has recently emerged as a factor associated with ASD. Although children with ASD are more likely to have gastrointestinal disorders, little is known about the association between food allergy and ASD. OBJECTIVE To examine the association of food allergy and other allergic conditions with ASD in US children. DESIGN, SETTING, AND PARTICIPANTS This population-based, cross-sectional study used data from the National Health Interview Survey collected between 1997 and 2016. The data analysis was performed in 2018. All eligible children aged 3 to 17 years were included. Food allergy, respiratory allergy, and skin allergy were defined based on an affirmative response in the questionnaire by a parent or guardian. MAIN OUTCOMES AND MEASURES Reported ASD diagnosed by a physician or other health professional. RESULTS This analysis included 199 520 children (unweighted mean [SD] age, 10.21 [4.41] years; 102 690 boys [51.47%]; 55 476 Hispanic [27.80%], 97 200 non-Hispanic white [48.72%], 30 760 non-Hispanic black [15.42%], and 16 084 non-Hispanic other race [8.06%]). Among them, 8734 (weighted prevalence, 4.31%) had food allergy, 24 555 (12.15%) had respiratory allergy, and 19 399 (9.91%) had skin allergy. A diagnosis of ASD was reported in 1868 children (0.95%). The weighted prevalence of reported food, respiratory, and skin allergies was higher in children with ASD (11.25%, 18.73%, and 16.81%, respectively) compared with children without ASD (4.25%, 12.08%, and 9.84%, respectively). In analyses adjusting for age, sex, race/ethnicity, family highest education level, family income level, geographical region, and mutual adjustment for other allergic conditions, the associations between allergic conditions and ASD remained significant. The odds ratio (OR) of ASD increased in association with food allergy (OR, 2.29; 95% CI, 1.87-2.81), respiratory allergy (OR, 1.28; 95% CI, 1.10-1.50), and skin allergy (OR, 1.50; 95% CI, 1.28-1.77) when comparing children with these conditions and those without. CONCLUSIONS AND RELEVANCE In a nationally representative sample of US children, a significant and positive association of common allergic conditions, in particular food allergy, with ASD was found. Further investigation is warranted to elucidate the causality and underlying mechanisms.
Collapse
Affiliation(s)
- Guifeng Xu
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City
- Center for Disabilities and Development, University of Iowa Stead Family Children’s Hospital, Iowa City
| | - Linda G. Snetselaar
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City
| | - Jin Jing
- Department of Maternal and Child Health, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Buyun Liu
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City
| | - Lane Strathearn
- Center for Disabilities and Development, University of Iowa Stead Family Children’s Hospital, Iowa City
- Division of Developmental and Behavioral Pediatrics, Stead Family Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City
| | - Wei Bao
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City
| |
Collapse
|
35
|
Sanctuary MR, Kain JN, Angkustsiri K, German JB. Dietary Considerations in Autism Spectrum Disorders: The Potential Role of Protein Digestion and Microbial Putrefaction in the Gut-Brain Axis. Front Nutr 2018; 5:40. [PMID: 29868601 PMCID: PMC5968124 DOI: 10.3389/fnut.2018.00040] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 04/30/2018] [Indexed: 12/13/2022] Open
Abstract
Children with autism spectrum disorders (ASD), characterized by a range of behavioral abnormalities and social deficits, display high incidence of gastrointestinal (GI) co-morbidities including chronic constipation and diarrhea. Research is now increasingly able to characterize the “fragile gut” in these children and understand the role that impairment of specific GI functions plays in the GI symptoms associated with ASD. This mechanistic understanding is extending to the interactions between diet and ASD, including food structure and protein digestive capacity in exacerbating autistic symptoms. Children with ASD and gut co-morbidities exhibit low digestive enzyme activity, impaired gut barrier integrity and the presence of antibodies specific for dietary proteins in the peripheral circulation. These findings support the hypothesis that entry of dietary peptides from the gut lumen into the vasculature are associated with an aberrant immune response. Furthermore, a subset of children with ASD exhibit high concentrations of metabolites originating from microbial activity on proteinaceous substrates. Taken together, the combination of specific protein intakes poor digestion, gut barrier integrity, microbiota composition and function all on a background of ASD represents a phenotypic pattern. A potential consequence of this pattern of conditions is that the fragile gut of some children with ASD is at risk for GI symptoms that may be amenable to improvement with specific dietary changes. There is growing evidence that shows an association between gut dysfunction and dysbiosis and ASD symptoms. It is therefore urgent to perform more experimental and clinical research on the “fragile gut” in children with ASD in order to move toward advancements in clinical practice. Identifying those factors that are of clinical value will provide an evidence-based path to individual management and targeted solutions; from real time sensing to the design of diets with personalized protein source/processing, all to improve GI function in children with ASD.
Collapse
Affiliation(s)
- Megan R Sanctuary
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Jennifer N Kain
- Department of Neurobiology, Physiology and Behavior Department, University of California, Davis, Davis, CA, United States
| | - Kathleen Angkustsiri
- School of Medicine, Department of Pediatrics, University of California, Davis, Sacramento, CA, United States.,Department of Pediatrics, UC Davis MIND Institute, Sacramento, CA, United States
| | - J Bruce German
- Department of Food Science and Technology, University of California, Davis, Davis, CA, United States.,Foods for Health Institute, University of California, Davis, Davis, CA, United States
| |
Collapse
|
36
|
Ohja K, Gozal E, Fahnestock M, Cai L, Cai J, Freedman JH, Switala A, El-Baz A, Barnes GN. Neuroimmunologic and Neurotrophic Interactions in Autism Spectrum Disorders: Relationship to Neuroinflammation. Neuromolecular Med 2018; 20:161-173. [PMID: 29691724 PMCID: PMC5942347 DOI: 10.1007/s12017-018-8488-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 03/28/2018] [Indexed: 12/30/2022]
Abstract
Autism spectrum disorders (ASD) are the most prevalent set of pediatric neurobiological disorders. The etiology of ASD has both genetic and environmental components including possible dysfunction of the immune system. The relationship of the immune system to aberrant neural circuitry output in the form of altered behaviors and communication characterized by ASD is unknown. Dysregulation of neurotrophins such as BDNF and their signaling pathways have been implicated in ASD. While abnormal cortical formation and autistic behaviors in mouse models of immune activation have been described, no one theory has been described to link activation of the immune system to specific brain signaling pathways aberrant in ASD. In this paper we explore the relationship between neurotrophin signaling, the immune system and ASD. To this effect we hypothesize that an interplay of dysregulated immune system, synaptogenic growth factors and their signaling pathways contribute to the development of ASD phenotypes.
Collapse
Affiliation(s)
- Kshama Ohja
- Department of Neurology, University of Louisville School of Medicine, Louisville, KY, USA.,Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Evelyne Gozal
- Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Margaret Fahnestock
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Lu Cai
- Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA.,Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Jun Cai
- Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Jonathan H Freedman
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Andy Switala
- Department of Bioengineering, University of Louisville, Louisville, KY, USA
| | - Ayman El-Baz
- Department of Bioengineering, University of Louisville, Louisville, KY, USA
| | - Gregory Neal Barnes
- Department of Neurology, University of Louisville School of Medicine, Louisville, KY, USA. .,Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA. .,Spafford Ackerly Chair in Child and Adolescent Psychiatry, University of Louisville Autism Center, 1405 East Burnett Avenue, Louisville, KY, 40217, USA.
| |
Collapse
|
37
|
Ashwood P. Differential T Cell Levels of Tumor Necrosis Factor Receptor-II in Children With Autism. Front Psychiatry 2018; 9:543. [PMID: 30524316 PMCID: PMC6256095 DOI: 10.3389/fpsyt.2018.00543] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 10/11/2018] [Indexed: 11/13/2022] Open
Abstract
Autism spectrum disorders (ASD) are characterized by impairments in verbal and non-verbal communication, in social interactions, and often accompanied by stereotypical interests and behaviors. A role for immune dysfunction has long been implicated in ASD pathophysiology, behavioral severity, and co-morbidities. The pro-inflammatory cytokine tumor necrosis factor alpha (TNFα) has been associated with ASD in some studies but little is known about its receptors. There are two receptors for TNFα, with TNFRI relaying many of the signals from TNFα, especially those that are rapid, whilst TNFRII relays later more long-term effects of TNFα. Proteolytic cleavage can lead to the soluble versions of these receptors which can neutralize the effects of TNFα. Here, we determined levels of TNFα and its receptors in 36 children with a confirmed diagnosis of ASD and 27 confirmed typically developing (TD) controls, 2-5 years-of-age. Children with ASD had higher levels of TNFRII on T cells compared to controls following cell stimulation. Levels of sTNFRII were decreased in cell supernatants following stimulation in ASD. Overall these data corroborate the role of inflammatory events in ASD and align with previous studies that have shown differential changes in cellular adaptive immunity in children with ASD. Future longitudinal analyzes of cellular immune function and downstream signaling from immune receptors will help further delineate the role of inflammation in ASD.
Collapse
Affiliation(s)
- Paul Ashwood
- Department of Medical Microbiology and Immunology, and The Medical Investigation of Neurodevelopmental Disorders Institute, University of California, Davis, CA, United States
| |
Collapse
|
38
|
Kalkman HO, Feuerbach D. Microglia M2A Polarization as Potential Link between Food Allergy and Autism Spectrum Disorders. Pharmaceuticals (Basel) 2017; 10:ph10040095. [PMID: 29232822 PMCID: PMC5748650 DOI: 10.3390/ph10040095] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 12/05/2017] [Accepted: 12/05/2017] [Indexed: 12/18/2022] Open
Abstract
Atopic diseases are frequently co-morbid with autism spectrum disorders (ASD). Allergic responses are associated with an activation of mast cells, innate lymphoid cells, and Th2 cells. These cells produce type-2 cytokines (IL4 and IL13), which stimulate microglia and macrophages to adopt a phenotype referred to as ‘alternative activation’ or ‘M2A’. M2A-polarized macrophages and microglia play a physiological role in tissue repair by secreting growth factors such as brain-derived neurotrophic factor (BDNF) and insulin-like growth factor-1. In ASD there is evidence for increased type-2 cytokines, microglia activation, M2A polarization, and increased levels of growth factors. In neurons, these growth factors drive a signal transduction pathway that leads to activation of the enzyme mammalian Target of Rapamycin (mTOR), and thereby to the inhibition of autophagy. Activation of mTOR is an effect that is also common to several of the genetic forms of autism. In the central nervous system, redundant synapses are removed via an autophagic process. Activation of mTOR would diminish the pruning of redundant synapses, which in the context of ASD is likely to be undesired. Based on this line of reasoning, atopic diseases like food allergy, eczema or asthma would represent risk factors for autism spectrum disorders.
Collapse
|
39
|
Ahmad SF, Nadeem A, Ansari MA, Bakheet SA, Al-Ayadhi LY, Attia SM. Upregulation of IL-9 and JAK-STAT signaling pathway in children with autism. Prog Neuropsychopharmacol Biol Psychiatry 2017; 79:472-480. [PMID: 28802860 DOI: 10.1016/j.pnpbp.2017.08.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 08/02/2017] [Accepted: 08/03/2017] [Indexed: 02/06/2023]
Abstract
Autism spectrum disorder (ASD) gradually develops predominantly neurodevelopmental disorders, which are socially diagnosed in early childhood. Though the etiopathology of ASD is not clear, immune alteration has been suggested as autism's pathophysiological mechanism. Previous studies found that several cytokines and transcription factor activation pathways were significantly increased in ASD. IL-9 has been confirmed to play a significant role in the central nervous system (CNS). The aim of the present study was to investigate the understudied role of pro- and anti-inflammatory cytokines and the JAK-STAT signaling pathway in ASD. We examined the IL-1β, IL-4, IFN-γ, and IL-9 positive immunostaining in all cells, and CD4+ T cells, in ASD and normally developing control children (TD), on peripheral blood mononuclear cells (PBMCs), using flow cytometry. We explored PBMC mRNA expression levels for IL-1β, IL-4, IFN-γ, IL-9, JAK1, and STAT5, by using real-time PCR (RT-PCR). We also explored PBMC protein expression levels for IL-1β, IL-4, IL-9, pJAK1, and pSTAT5 by using western blotting. We found that the children with ASD had increased IL-1β, IL-4, IFN-γ, and IL-9 positive immunostaining in all cells, and in CD4+ cells, relative to the TD controls. The mRNA and protein expression for IL-1β, IL-4, IFN-γ, IL-9, JAK1, pJAK1, STAT5, and pSTAT5 were also significantly elevated in ASD relative to TD controls. These results suggested that cytokines and JAK-STAT activation signaling have an essential role in immune dysfunction in ASD.
Collapse
Affiliation(s)
- Sheikh F Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mushtaq A Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Saleh A Bakheet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Laila Yousef Al-Ayadhi
- Autism Research and Treatment Center, AL-Amodi Autism Research Chair, Department of Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia; Department of Pharmacology and Toxicology, College of Pharmacy, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
40
|
Ordaz JD, Damayanti NP, Irudayaraj JMK. Toxicological effects of trichloroethylene exposure on immune disorders. Immunopharmacol Immunotoxicol 2017; 39:305-317. [PMID: 28828896 DOI: 10.1080/08923973.2017.1364262] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Trichloroethylene (TCE) is one of the most common ground water contaminants in USA. Even though recent regulation mandates restricted utilization of TCE, its use is not completely prohibited, especially in industrial and manufacturing processes. The risk of TCE on human health is an ongoing field of study and its implications on certain diseases such as cancer has been recognized and well-documented. However, the link between TCE and immune disorders is still an under-studied area. Studies on the risk of TCE on the immune system is usually focused on certain immune class disorders, but consensus on the impact of TCE on the immune system has not been established. This review presents representative work that investigates the effect of TCE on immune disorders and highlights future opportunities. We attempt to provide a broader perspective of the risks of TCE on the immune system and human health.
Collapse
Affiliation(s)
- Josue D Ordaz
- a Department of Agricultural & Biological Engineering , Bindley Bioscience Center, Purdue University , West Lafayette , IN , USA.,b Indiana University School of Medicine , Indianapolis , IN , USA
| | - Nur P Damayanti
- a Department of Agricultural & Biological Engineering , Bindley Bioscience Center, Purdue University , West Lafayette , IN , USA.,b Indiana University School of Medicine , Indianapolis , IN , USA
| | - Joseph M K Irudayaraj
- a Department of Agricultural & Biological Engineering , Bindley Bioscience Center, Purdue University , West Lafayette , IN , USA
| |
Collapse
|
41
|
Jónsdóttir U, Lang JE. How does autism spectrum disorder affect the risk and severity of childhood asthma? Ann Allergy Asthma Immunol 2017; 118:570-576. [PMID: 28477788 DOI: 10.1016/j.anai.2017.02.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 02/16/2017] [Accepted: 02/20/2017] [Indexed: 01/01/2023]
Abstract
BACKGROUND Autism spectrum disorder (ASD) and asthma are among the most common chronic disorders in childhood. Both are associated with altered immune regulation and share several risk factors. The effects of ASD on risk for later asthma and asthma severity remain unclear. OBJECTIVE To determine whether ASD in children increases the risk of incident asthma and worsens asthma severity. METHODS We performed 2 distinct analytic designs (case-control and retrospective longitudinal cohort) using a multistate electronic health records database to assess the odds of new asthma and asthma severity among children with ASD. In both designs, children with ASD were matched with children without ASD according to sex, age, race, ethnicity, location, and insurance status. Pulmonary function, controller medication prescriptions, asthma exacerbations, and asthma-related hospitalizations were collected. The effects of ASD on asthma risk and severity were assessed using multivariable linear and logistic regression. RESULTS Among children with asthma, ASD was associated with reduced exacerbations (odds ratio [OR], 0.71; 95% confidence interval [CI], 0.54-0.92), better forced expiratory volume in 1 second/forced vital capacity ratio (0.876 vs 0.841, P < .001), and lower odds of airflow obstruction (OR, 0.53; 95% CI, 0.31-0.90) but had higher odds of asthma controller prescription (OR, 2.18; 95% CI, 1.62-2.93). In a longitudinal analysis of children without asthma, ASD was found to be protective for new asthma (OR, 0.44; 95% CI, 0.26-0.74). CONCLUSION Among children with asthma, concomitant ASD is associated with better asthma-related outcomes but a higher controller treatment burden. In addition, our data did not support ASD as a risk factor for incident asthma.
Collapse
Affiliation(s)
| | - Jason E Lang
- Division of Pulmonary and Sleep Medicine, Nemours Children's Hospital, Orlando, Florida; Division of Allergy/Immunology and Pulmonary Medicine, Duke University School of Medicine, Durham, North Carolina.
| |
Collapse
|
42
|
Yenkoyan K, Grigoryan A, Fereshetyan K, Yepremyan D. Advances in understanding the pathophysiology of autism spectrum disorders. Behav Brain Res 2017; 331:92-101. [DOI: 10.1016/j.bbr.2017.04.038] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 04/16/2017] [Accepted: 04/18/2017] [Indexed: 10/19/2022]
|
43
|
Filiano AJ, Gadani SP, Kipnis J. How and why do T cells and their derived cytokines affect the injured and healthy brain? Nat Rev Neurosci 2017; 18:375-384. [PMID: 28446786 PMCID: PMC5823005 DOI: 10.1038/nrn.2017.39] [Citation(s) in RCA: 147] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The evolution of adaptive immunity provides enhanced defence against specific pathogens, as well as homeostatic immune surveillance of all tissues. Despite being 'immune privileged', the CNS uses the assistance of the immune system in physiological and pathological states. In this Opinion article, we discuss the influence of adaptive immunity on recovery after CNS injury and on cognitive and social brain function. We further extend a hypothesis that the pro-social effects of interferon-regulated genes were initially exploited by pathogens to increase host-host transmission, and that these genes were later recycled by the host to form part of an immune defence programme. In this way, the evolution of adaptive immunity may reflect a host-pathogen 'arms race'.
Collapse
Affiliation(s)
- Anthony J Filiano
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA
| | - Sachin P Gadani
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA
| | - Jonathan Kipnis
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA
| |
Collapse
|
44
|
Immune Alterations in CD8+ T Cells Are Associated with Neuronal C-C and C-X-C Chemokine Receptor Regulation Through Adenosine A2A Receptor Signaling in a BTBR T+ Itpr3tf/J Autistic Mouse Model. Mol Neurobiol 2017; 55:2603-2616. [DOI: 10.1007/s12035-017-0548-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 04/07/2017] [Indexed: 11/25/2022]
|
45
|
Voltas N, Arija V, Hernández-Martínez C, Jiménez-Feijoo R, Ferré N, Canals J. Are there early inflammatory biomarkers that affect neurodevelopment in infancy? J Neuroimmunol 2017; 305:42-50. [DOI: 10.1016/j.jneuroim.2017.01.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 12/31/2016] [Accepted: 01/11/2017] [Indexed: 12/21/2022]
|
46
|
Mizoguchi Y, Monji A. Microglial Intracellular Ca 2+ Signaling in Synaptic Development and its Alterations in Neurodevelopmental Disorders. Front Cell Neurosci 2017; 11:69. [PMID: 28367116 PMCID: PMC5355421 DOI: 10.3389/fncel.2017.00069] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 02/24/2017] [Indexed: 12/27/2022] Open
Abstract
Autism spectrum disorders (ASDs) are neurodevelopmental disorders characterized by deficits in social interaction, difficulties with language and repetitive/restricted behaviors. Microglia are resident innate immune cells which release many factors including proinflammatory cytokines, nitric oxide (NO) and brain-derived neurotrophic factor (BDNF) when they are activated in response to immunological stimuli. Recent in vivo imaging has shown that microglia sculpt and refine the synaptic circuitry by removing excess and unwanted synapses and be involved in the development of neural circuits or synaptic plasticity thereby maintaining the brain homeostasis. BDNF, one of the neurotrophins, has various important roles in cell survival, neurite outgrowth, neuronal differentiation, synaptic plasticity and the maintenance of neural circuits in the CNS. Intracellular Ca2+ signaling is important for microglial functions including ramification, de-ramification, migration, phagocytosis and release of cytokines, NO and BDNF. BDNF induces a sustained intracellular Ca2+ elevation through the upregulation of the surface expression of canonical transient receptor potential 3 (TRPC3) channels in rodent microglia. BDNF might have an anti-inflammatory effect through the inhibition of microglial activation and TRPC3 could play important roles in not only inflammatory processes but also formation of synapse through the modulation of microglial phagocytic activity in the brain. This review article summarizes recent findings on emerging dual, inflammatory and non-inflammatory, roles of microglia in the brain and reinforces the importance of intracellular Ca2+ signaling for microglial functions in both normal neurodevelopment and their potential contributing to neurodevelopmental disorders such as ASDs.
Collapse
Affiliation(s)
- Yoshito Mizoguchi
- Department of Psychiatry, Faculty of Medicine, Saga University Saga, Japan
| | - Akira Monji
- Department of Psychiatry, Faculty of Medicine, Saga University Saga, Japan
| |
Collapse
|
47
|
Onore C, Yang H, Van de Water J, Ashwood P. Dynamic Akt/mTOR Signaling in Children with Autism Spectrum Disorder. Front Pediatr 2017; 5:43. [PMID: 28361047 PMCID: PMC5350147 DOI: 10.3389/fped.2017.00043] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 02/15/2017] [Indexed: 12/15/2022] Open
Abstract
Autism spectrum disorder (ASD) is a behaviorally defined disorder affecting 1 in 68 children. Currently, there is no known cause for the majority of ASD cases nor are there physiological diagnostic tools or biomarkers to aid behavioral diagnosis. Whole-genome linkage studies, genome-wide association studies, copy number variation screening, and SNP analyses have identified several ASD candidate genes, but which vary greatly among individuals and family clusters, suggesting that a variety of genetic mutations may result in a common pathology or alter a common mechanistic pathway. The Akt/mammalian target of rapamycin (mTOR) pathway is involved in many cellular processes including synaptic plasticity and immune function that can alter neurodevelopment. In this study, we examined the activity of the Akt/mTOR pathway in cells isolated from children with ASD and typically developing controls. We observed higher activity of mTOR, extracellular receptor kinase, and p70S6 kinase and lower activity of glycogen synthase kinase 3 (GSK3)α and tuberin (TSC2) in cells from children with ASD. These data suggest a phosphorylation pattern indicative of higher activity in the Akt/mTOR pathway in children with general/idiopathic ASD and may suggest a common pathological pathway of interest for ASD.
Collapse
Affiliation(s)
- Charity Onore
- The M.I.N.D. Institute, University of California Davis, Davis, CA, USA
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
| | - Houa Yang
- The M.I.N.D. Institute, University of California Davis, Davis, CA, USA
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
| | - Judy Van de Water
- The M.I.N.D. Institute, University of California Davis, Davis, CA, USA
- Division of Rheumatology, Allergy and Clinical Immunology, University of California Davis, Davis, CA, USA
| | - Paul Ashwood
- The M.I.N.D. Institute, University of California Davis, Davis, CA, USA
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
| |
Collapse
|
48
|
Careaga M, Rogers S, Hansen RL, Amaral DG, Van de Water J, Ashwood P. Immune Endophenotypes in Children With Autism Spectrum Disorder. Biol Psychiatry 2017; 81:434-441. [PMID: 26493496 PMCID: PMC4788581 DOI: 10.1016/j.biopsych.2015.08.036] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 08/11/2015] [Accepted: 08/25/2015] [Indexed: 11/17/2022]
Abstract
BACKGROUND Autism spectrum disorder (ASD) is characterized by social communication deficits and restricted, repetitive patterns of behavior. Varied immunological findings have been reported in children with ASD. To address the question of heterogeneity in immune responses, we sought to examine the diversity of immune profiles within a representative cohort of boys with ASD. METHODS Peripheral blood mononuclear cells from male children with ASD (n = 50) and from typically developing age-matched male control subjects (n = 16) were stimulated with either lipopolysaccharide or phytohemagglutinin. Cytokine production was assessed after stimulation. The ASD study population was clustered into subgroups based on immune responses and assessed for behavioral outcomes. RESULTS Children with ASD who had a proinflammatory profile based on lipopolysaccharide stimulation were more developmentally impaired as assessed by the Mullen Scales of Early Learning. They also had greater impairments in social affect as measured by the Autism Diagnostic Observation Schedule. These children also displayed more frequent sleep disturbances and episodes of aggression. Similarly, children with ASD and a more activated T cell cytokine profile after phytohemagglutinin stimulation were more developmentally impaired as measured by the Mullen Scales of Early Learning. CONCLUSIONS Children with ASD may be phenotypically characterized based upon their immune profile. Those showing either an innate proinflammatory response or increased T cell activation/skewing display a more impaired behavioral profile than children with noninflamed or non-T cell activated immune profiles. These data suggest that there may be several possible immune subphenotypes within the ASD population that correlate with more severe behavioral impairments.
Collapse
Affiliation(s)
- Milo Careaga
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, California; MIND Institute, University of California, Davis, Davis, California
| | - Sally Rogers
- MIND Institute, University of California, Davis, Davis, California; Departments of Psychiatry, University of California, Davis, Davis, California
| | - Robin L Hansen
- MIND Institute, University of California, Davis, Davis, California; Pediatrics, University of California, Davis, Davis, California
| | - David G Amaral
- MIND Institute, University of California, Davis, Davis, California; Departments of Psychiatry, University of California, Davis, Davis, California
| | - Judy Van de Water
- MIND Institute, University of California, Davis, Davis, California; Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, Davis, California
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, California; MIND Institute, University of California, Davis, Davis, California.
| |
Collapse
|
49
|
Masi A, Glozier N, Dale R, Guastella AJ. The Immune System, Cytokines, and Biomarkers in Autism Spectrum Disorder. Neurosci Bull 2017; 33:194-204. [PMID: 28238116 PMCID: PMC5360854 DOI: 10.1007/s12264-017-0103-8] [Citation(s) in RCA: 151] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Accepted: 01/22/2017] [Indexed: 12/24/2022] Open
Abstract
Autism Spectrum Disorder (ASD) is a pervasive neurodevelopmental condition characterized by variable impairments in communication and social interaction as well as restricted interests and repetitive behaviors. Heterogeneity of presentation is a hallmark. Investigations of immune system problems in ASD, including aberrations in cytokine profiles and signaling, have been increasing in recent times and are the subject of ongoing interest. With the aim of establishing whether cytokines have utility as potential biomarkers that may define a subgroup of ASD, or function as an objective measure of response to treatment, this review summarizes the role of the immune system, discusses the relationship between the immune system, the brain, and behavior, and presents previously-identified immune system abnormalities in ASD, specifically addressing the role of cytokines in these aberrations. The roles and identification of biomarkers are also addressed, particularly with respect to cytokine profiles in ASD.
Collapse
Affiliation(s)
- Anne Masi
- Autism Clinic for Translational Research, Brain and Mind Centre, Central Clinical School, Sydney Medical School, University of Sydney, Sydney, New South Wales, 2050, Australia
| | - Nicholas Glozier
- Autism Clinic for Translational Research, Brain and Mind Centre, Central Clinical School, Sydney Medical School, University of Sydney, Sydney, New South Wales, 2050, Australia
| | - Russell Dale
- Childrens Hospital at Westmead Clinical School, University of Sydney, Sydney, New South Wales, 2145, Australia
| | - Adam J Guastella
- Autism Clinic for Translational Research, Brain and Mind Centre, Central Clinical School, Sydney Medical School, University of Sydney, Sydney, New South Wales, 2050, Australia.
| |
Collapse
|
50
|
Sivanesan S, Tan A, Jeyaraj R, Lam J, Gole M, Hardan A, Ashkan K, Rajadas J. Pharmaceuticals and Stem Cells in Autism Spectrum Disorders: Wishful Thinking? World Neurosurg 2017; 98:659-672. [DOI: 10.1016/j.wneu.2016.09.100] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Revised: 09/24/2016] [Accepted: 09/26/2016] [Indexed: 12/21/2022]
|