1
|
Kaminski VDL, Kulmann-Leal B, Tyska-Nunes GL, Beltrame BP, Riesgo RDS, Schüler-Faccini L, Roman T, Schuch JB, Chies JAB. Association between NKG2/KLR gene variants and epilepsy in Autism Spectrum Disorder. J Neuroimmunol 2023; 381:578132. [PMID: 37352688 DOI: 10.1016/j.jneuroim.2023.578132] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/31/2023] [Accepted: 06/15/2023] [Indexed: 06/25/2023]
Abstract
Autism Spectrum Disorder (ASD) is a set of neurodevelopmental disorders mainly characterized by repetitive, restrictive and stereotypical behaviors, and impaired communication skills. Several lines of evidence indicate that alterations of the immune system account for ASD development, including the presence of brain-reactive antibodies, abnormal T cell activation, altered cytokine levels in brain, cerebrospinal fluid and peripheral blood circulation, increased levels of circulating monocytes, and dysregulation in Natural Killer (NK) cells activity. Regarding NK cells, a lower cytotoxic activity, a higher level of activation and an increased number of these cells in individuals with ASD have been described. In 2019, a study showed that NK cells derived from patients with ASD show a characteristic pattern of NKG2C overexpression, highlighting the importance of the NK cell pathway in ASD. In fact, the study of genes related to NK cell activity has proven to be an excellent research target, both in terms of susceptibility as well as a marker for the different clinical manifestations observed in ASD individuals. Here, we evaluated the influence of KLRC2 gene deletion as well as KLRK1 rs1049174 and rs2255336 variants in a cohort of 185 children diagnosed with ASD and their respective biological parents in southern Brazil. Of note, this is the first study concerning genetic variants of the KLRC2 and KLRK1 genes in an ASD sample. The KLRC2 gene deletion (p = 0.001; pc = 0.009), KLRK1 rs1049174 (p = 0.005; pc = 0.045) and KLRK1 rs2255336 (p = 0.001; pc = 0.009) were associated with epilepsy in ASD patients. The results indicate that KLRC2 deletion, KLRK1 rs2255336, and KLRK1 rs1049174 could be involved in epilepsy manifestation in ASD patients, possibly impacting the NK dysregulation already described in ASD and epileptic patients.
Collapse
Affiliation(s)
- Valéria de Lima Kaminski
- Laboratory of Immunobiology and Immunogenetics, Graduate Program in Genetics and Molecular Biology - PPGBM, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil; Applied Immunology Laboratory, Graduate Program in Biotechnology, Institute of Science and Technology - ICT, Universidade Federal de São Paulo - UNIFESP, São José dos Campos - São Paulo, Brazil; Universidade Anhembi Morumbi, São José dos Campos - São Paulo, Brazil
| | - Bruna Kulmann-Leal
- Laboratory of Immunobiology and Immunogenetics, Graduate Program in Genetics and Molecular Biology - PPGBM, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - Guilherme Luís Tyska-Nunes
- Laboratory of Immunobiology and Immunogenetics, Graduate Program in Genetics and Molecular Biology - PPGBM, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - Brenda Pedron Beltrame
- Laboratory of Immunobiology and Immunogenetics, Graduate Program in Genetics and Molecular Biology - PPGBM, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - Rudimar Dos Santos Riesgo
- Child Neurology Unit, Hospital de Clínicas de Porto Alegre (HCPA), Universidade Federal do Rio Grande do Sul - UFRGS, Rua Ramiro Barcelos, 2350, Porto Alegre 90035-903, Brazil
| | - Lavinia Schüler-Faccini
- Laboratory of Immunobiology and Immunogenetics, Graduate Program in Genetics and Molecular Biology - PPGBM, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil; Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil; National Institute of Population Medical Genetics (INAGEMP), Porto Alegre, Brazil; Brazilian Teratogen Information Service (SIAT), Medical Genetics Service, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Tatiana Roman
- Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - Jaqueline Bohrer Schuch
- Graduate Program in Psychiatry and Behavioral Sciences, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - José Artur Bogo Chies
- Laboratory of Immunobiology and Immunogenetics, Graduate Program in Genetics and Molecular Biology - PPGBM, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil.
| |
Collapse
|
2
|
Kaminski VDL, Michita RT, Ellwanger JH, Veit TD, Schuch JB, Riesgo RDS, Roman T, Chies JAB. Exploring potential impacts of pregnancy-related maternal immune activation and extracellular vesicles on immune alterations observed in autism spectrum disorder. Heliyon 2023; 9:e15593. [PMID: 37305482 PMCID: PMC10256833 DOI: 10.1016/j.heliyon.2023.e15593] [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: 06/15/2022] [Revised: 04/03/2023] [Accepted: 04/14/2023] [Indexed: 06/13/2023] Open
Abstract
Autism Spectrum Disorder (ASD) is a set of neurodevelopmental disorders usually observed in early life, with impacts on behavioral and social skills. Incidence of ASD has been dramatically increasing worldwide, possibly due to increase in awareness/diagnosis as well as to genetic and environmental triggers. Currently, it is estimated that ∼1% of the world population presents ASD symptoms. In addition to its genetic background, environmental and immune-related factors also influence the ASD etiology. In this context, maternal immune activation (MIA) has recently been suggested as a component potentially involved in ASD development. In addition, extracellular vesicles (EVs) are abundant at the maternal-fetal interface and are actively involved in the immunoregulation required for a healthy pregnancy. Considering that alterations in concentration and content of EVs have also been associated with ASD, this article raises a debate about the potential roles of EVs in the processes surrounding MIA. This represents the major differential of the present review compared to other ASD studies. To support the suggested correlations and hypotheses, findings regarding the roles of EVs during pregnancy and potential influences on ASD are discussed, along with a review and update concerning the participation of infections, cytokine unbalances, overweight and obesity, maternal anti-fetal brain antibodies, maternal fever, gestational diabetes, preeclampsia, labor type and microbiota unbalances in MIA and ASD.
Collapse
Affiliation(s)
- Valéria de Lima Kaminski
- Laboratório de Imunobiologia e Imunogenética, Departamento de Genética, Universidade Federal do Rio Grande do Sul – UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
- Programa de Pós-Graduação em Genética e Biologia Molecular, Departamento de Genética, Universidade Federal do Rio Grande do Sul – UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
- Programa de Pós-Graduação em Biotecnologia, Laboratório de Imunologia Aplicada, Instituto de Ciência e Tecnologia - ICT, Universidade Federal de São Paulo - UNIFESP, São José dos Campos, São Paulo, Brazil
| | - Rafael Tomoya Michita
- Laboratório de Genética Molecular Humana, Universidade Luterana do Brasil - ULBRA, Canoas, Rio Grande do Sul, Brazil
| | - Joel Henrique Ellwanger
- Laboratório de Imunobiologia e Imunogenética, Departamento de Genética, Universidade Federal do Rio Grande do Sul – UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
- Programa de Pós-Graduação em Genética e Biologia Molecular, Departamento de Genética, Universidade Federal do Rio Grande do Sul – UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Tiago Degani Veit
- Instituto de Ciências Básicas da Saúde, Departmento de Microbiologia, Imunologia e Parasitologia, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Jaqueline Bohrer Schuch
- Centro de Pesquisa em Álcool e Drogas, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
- Programa de Pós-Graduação em Psiquiatria e Ciências do Comportamento, Universidade Federal do Rio Grande do Sul – UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Rudimar dos Santos Riesgo
- Child Neurology Unit, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Tatiana Roman
- Programa de Pós-Graduação em Genética e Biologia Molecular, Departamento de Genética, Universidade Federal do Rio Grande do Sul – UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - José Artur Bogo Chies
- Laboratório de Imunobiologia e Imunogenética, Departamento de Genética, Universidade Federal do Rio Grande do Sul – UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
- Programa de Pós-Graduação em Genética e Biologia Molecular, Departamento de Genética, Universidade Federal do Rio Grande do Sul – UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
3
|
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
|
4
|
Syu GD, Sutandy FXR, Chen K, Cheng Y, Chen CS, Shih JC. Autoantibody profiling of monoamine oxidase A knockout mice, an autism spectrum disorder model. Brain Behav Immun 2023; 107:193-200. [PMID: 36243286 DOI: 10.1016/j.bbi.2022.10.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 09/04/2022] [Accepted: 10/09/2022] [Indexed: 12/05/2022] Open
Abstract
Monoamine oxidase A (MAO A) is the critical enzyme to degrade serotonin in the brain and the knockout mouse exhibits hyperserotonemia and abnormalities that are observed in autism spectrum disorder (ASD). Thus, the MAO A knockout mouse is a valuable model for studying neurological and behavioral impairments in ASD. Based on the immune dysfunction hypothesis, dysregulated humoral immunity may cause neurological impairments. To address this hypothesis, we use high-density proteome microarray to profile the serum antibodies in both wild-type and MAO A knockout mice. The distingue autoantibody signatures were observed in the MAO A knockout and wild-type controls and showed 165 up-regulated and 232 down-regulated autoantibodies. The up-regulated autoantibodies were prone to target brain tissues while down-regulated ones were enriched in sex organs. The identified autoantibodies help bridge the gap between ASD mouse models and humoral immunity, not only yielding insights into the pathological mechanisms but also providing potential biomarkers for translational research in ASD.
Collapse
Affiliation(s)
- Guan-Da Syu
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, Taiwan; International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan; Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan.
| | - F X Reymond Sutandy
- Institute for Biochemistry II, Goethe University, Theodor-Stern-Kai 7, Frankfurt, Germany
| | - Kevin Chen
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, USA
| | - Yawei Cheng
- Department of Physical Medicine and Rehabilitation, National Yang-Ming University Hospital, Yilan, Taiwan; Institute of Neuroscience and Brain Research Center, National Yang-Ming University, Taipei, Taiwan; Department of Education and Research, Taipei City Hospital, Taipei, Taiwan
| | - Chien-Sheng Chen
- Department of Food Safety/Hygiene and Risk Management, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC.
| | - Jean C Shih
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, USA; USC-Taiwan Center for Translational Research, University of Southern California, Los Angeles, USA; Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, USA.
| |
Collapse
|
5
|
Bilgen Ulgar Ş, Ayaydın H, Çelik H, Koyuncu İ, Kirmit A. Evaluation of antineuronal antibodies and 8-OHdG in mothers of children with autism spectrum disorder: a case-control study. Int J Psychiatry Clin Pract 2022; 26:244-250. [PMID: 34689686 DOI: 10.1080/13651501.2021.1993925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVE The purpose of our study was to investigated the anti-Yo, anti-Hu, anti-Ri, anti-amphiphysin antibody levels and 8-OHdG in mothers of children with autism. METHODS This study included 60 participants, 33 of whom were healthy mothers of 3-12-year-old children diagnosed with autism spectrum disorder (ASD) and the 27 others who constituted the control group, were healthy mothers with age-matched healthy children. Two groups were examined for plasma anti-Yo, anti-Hu, anti-amphiphysin and anti-Ri antibodies and, 8-OHdG levels. The participants were asked to accomplish a sociodemographic data form. The severity of ASD symptoms was evaluated according to the Childhood Autism Rating Scale (CARS). RESULTS Anti-amphiphysin antibody levels and anti-Ri antibody positivity were significantly higher in the case group (p = 0.001; p = 0.027, respectively). The two groups did not significantly differ in terms of anti-Yo and anti-Hu antibody levels and in terms of 8-OHdG levels (p = 0.065; p = 0.099; p = 0.490, respectively). The two groups did not significantly differ in terms of sociodemographic data (p > 0.05). CONCLUSIONS According to the our study, maternal antineuronal antibodies, such as anti-amphiphysin and anti-Ri, may contribute to the risk of childhood autism. Studies with larger samples are needed.KEY POINTSMaternal factors associated with autism should be investigated in order to create early diagnosis and treatment opportunities for autism.Based on the importance of immunological and cerebellar pathologies in autism aetiology, we aimed to investigate antineuronal antibodies in mothers of children with autism.Maternal antineuronal antibodies, such as anti-amphiphysin and anti-Ri, may contribute to the risk of childhood autism.High anti-amphiphysin antibody levels in mothers of children with autism may also occur against the amphiphysin in the structure of the SrGAP3 gene, which is associated with autism.
Collapse
Affiliation(s)
- Şermin Bilgen Ulgar
- Department of Child and Adolescent Psychiatry, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Hamza Ayaydın
- Department of Child and Adolescent Psychiatry, School of Medicine, Harran University, Şanlıurfa, Turkey
| | - Hakim Çelik
- Department of Physiology, School of Medicine, Harran University, Şanlıurfa, Turkey
| | - İsmail Koyuncu
- Department of Biochemistry, School of Medicine, Harran University, Şanlıurfa, Turkey
| | - Adnan Kirmit
- Department of Biochemistry, School of Medicine, Harran University, Şanlıurfa, Turkey
| |
Collapse
|
6
|
Maltsev D, Stefanyshyn V. The efficacy of combined immunotherapy with Propes and Inflamafertin in adult patients with genetic deficiency of the folate cycle and selective deficiency of NK and NKT cells. Immunol Suppl 2022; 167:443-450. [PMID: 35866691 DOI: 10.1111/imm.13548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 07/15/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND The purpose of the present study is to evaluate the efficacy of combination immunotherapy with Propes and Inflamafertin in GDFC adults with NK and/or NKT cell deficiency. METHODS This single-center, retrospective, controlled, non-randomized clinical trial analyzed medical records of 212 adult GDFC patients aged 19 to 50 years (study group, SG). SG received Propes at a dose of 2 mL intramuscularly every other day at night for 3 consecutive months and Inflamafertin at a dose of 2 mL IM every other day at night for 3 consecutive months in rotation with Propes. The control group involved 34 patients with GDFC who followed the same age and gender distribution pattern but did not receive immunotherapy. RESULTS The number of NK cells reached the lower limit of normal in 95 out of 131 patients (72% of cases) with baseline deficiency of these lymphocytes. The average number of NK cells in the blood of SG patients almost doubled during the 3-month course of immunotherapy (p ˂ 0.05; Z ˂ Z0.05 ). However, it almost returned to initial levels 2 months following discontinuation of the immunotherapeutic agents (p ˃ 0.05; Z ˃ Z0.05 ). CONCLUSION Combination immunotherapy with Propes and Inflamafertin is an effective strategy for the treatment of immunodeficiency caused by GDFC in adult patients.
Collapse
Affiliation(s)
- Dmitry Maltsev
- Laboratory of Immunology and Molecular Biology, Research Institute of Experimental and Clinical Medicine, O.O. Bogomolets National Medical University, Kyiv, Ukraine
| | | |
Collapse
|
7
|
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
|
8
|
Kern JK, Geier DA, Mehta JA, Homme KG, Geier MR. Mercury as a hapten: A review of the role of toxicant-induced brain autoantibodies in autism and possible treatment considerations. J Trace Elem Med Biol 2020; 62:126504. [PMID: 32534375 DOI: 10.1016/j.jtemb.2020.126504] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/28/2020] [Accepted: 03/18/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND Mercury has many direct and well-recognized neurotoxic effects. However, its immune effects causing secondary neurotoxicity are less well-recognized. Mercury exposure can induce immunologic changes in the brain indicative of autoimmune dysfunction, including the production of highly specific brain autoantibodies. Mercury, and in particular, Thimerosal, can combine with a larger carrier, such as an endogenous protein, thereby acting as a hapten, and this new molecule can then elicit the production of antibodies. METHODS A comprehensive search using PubMed and Google Scholar for original studies and reviews related to autism, mercury, autoantibodies, autoimmune dysfunction, and haptens was undertaken. All articles providing relevant information from 1985 to date were examined. Twenty-three studies were identified showing autoantibodies in the brains of individuals diagnosed with autism and all were included and discussed in this review. RESULTS Research shows mercury exposure can result in an autoimmune reaction that may be causal or contributory to autism, especially in children with a family history of autoimmunity. The autoimmune pathogenesis in autism is demonstrated by the presence of brain autoantibodies (neuroantibodies), which include autoantibodies to: (1) human neuronal progenitor cells; (2) myelin basic protein (MBP); (3) neuron-axon filament protein (NAFP); (4) brain endothelial cells; (5) serotonin receptors; (6) glial fibrillary acidic protein (GFAP); (7) brain derived neurotrophic factor (BDNF); (8) myelin associated glycoprotein (MAG); and (9) various brain proteins in the cerebellum, hypothalamus, prefrontal cortex, cingulate gyrus, caudate putamen, cerebral cortex and caudate nucleus. CONCLUSION Recent evidence suggests a relationship between mercury exposure and brain autoantibodies in individuals diagnosed with autism. Moreover, brain autoantibody levels in autism are found to correlate with both autism severity and blood mercury levels. Treatments to reduce mercury levels and/or brain autoantibody formation should be considered in autism.
Collapse
Affiliation(s)
- Janet K Kern
- Institute of Chronic Illnesses, Inc., Silver Spring, MD, USA; CoMeD, Inc., Silver Spring, MD, USA; CONEM US Autism Research Group, Allen, TX, USA.
| | - David A Geier
- Institute of Chronic Illnesses, Inc., Silver Spring, MD, USA; CoMeD, Inc., Silver Spring, MD, USA
| | - Jyutika A Mehta
- Texas Woman's University, Department of Communication Sciences and Disorders, Denton, TX, USA
| | - Kristin G Homme
- CoMeD, Inc., Silver Spring, MD, USA; International Academy of Oral Medicine and Toxicology, Champions Gate, FL, USA
| | - Mark R Geier
- Institute of Chronic Illnesses, Inc., Silver Spring, MD, USA; CoMeD, Inc., Silver Spring, MD, USA
| |
Collapse
|
9
|
Critical Role of the Maternal Immune System in the Pathogenesis of Autism Spectrum Disorder. Biomedicines 2020; 8:biomedicines8120557. [PMID: 33271759 PMCID: PMC7760377 DOI: 10.3390/biomedicines8120557] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/26/2020] [Accepted: 11/29/2020] [Indexed: 12/27/2022] Open
Abstract
Autism spectrum disorders (ASD) are a group of neurodevelopmental disorders characterised by impairments in communication, social interaction, and the presence of restrictive and repetitive behaviours. Over the past decade, most of the research in ASD has focused on the contribution of genetics, with the identification of a variety of different genes and mutations. However, the vast heterogeneity in clinical presentations associated with this disorder suggests that environmental factors may be involved, acting as a “second hit” in already genetically susceptible individuals. To this regard, emerging evidence points towards a role for maternal immune system dysfunctions. This literature review considered evidence from epidemiological studies and aimed to discuss the pathological relevance of the maternal immune system in ASD by looking at the proposed mechanisms by which it alters the prenatal environment. In particular, this review focuses on the effects of maternal immune activation (MIA) by looking at foetal brain-reactive antibodies, cytokines and the microbiome. Despite the arguments presented here that strongly implicate MIA in the pathophysiology of ASD, further research is needed to fully understand the precise mechanisms by which they alter brain structure and behaviour. Overall, this review has not only shown the importance of the maternal immune system as a risk factor for ASD, but more importantly, has highlighted new promising pathways to target for the discovery of novel therapeutic interventions for the treatment of such a life-changing disorder.
Collapse
|
10
|
Carpita B, Marazziti D, Palego L, Giannaccini G, Betti L, Dell'Osso L. Microbiota, Immune System and Autism Spectrum Disorders: An Integrative Model towards Novel Treatment Options. Curr Med Chem 2020; 27:5119-5136. [PMID: 31448708 DOI: 10.2174/0929867326666190328151539] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 03/05/2019] [Accepted: 03/06/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Autism Spectrum Disorder (ASD) is a condition strongly associated with genetic predisposition and familial aggregation. Among ASD patients, different levels of symptoms severity are detectable, while the presence of intermediate autism phenotypes in close relatives of ASD probands is also known in literature. Recently, increasing attention has been paid to environmental factors that might play a role in modulating the relationship between genomic risk and development and severity of ASD. Within this framework, an increasing body of evidence has stressed a possible role of both gut microbiota and inflammation in the pathophysiology of neurodevelopment. The aim of this paper is to review findings about the link between microbiota dysbiosis, inflammation and ASD. METHODS Articles ranging from 1990 to 2018 were identified on PUBMED and Google Scholar databases, with keyword combinations as: microbiota, immune system, inflammation, ASD, autism, broad autism phenotype, adult. RESULTS Recent evidence suggests that microbiota alterations, immune system and neurodevelopment may be deeply intertwined, shaping each other during early life. However, results from both animal models and human samples are still heterogeneous, while few studies focused on adult patients and ASD intermediate phenotypes. CONCLUSION A better understanding of these pathways, within an integrative framework between central and peripheral systems, might not only shed more light on neural basis of ASD symptoms, clarifying brain pathophysiology, but it may also allow to develop new therapeutic strategies for these disorders, still poorly responsive to available treatments.
Collapse
Affiliation(s)
- Barbara Carpita
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, Via Roma, 6756100 Pisa, Italy
| | - Donatella Marazziti
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, Via Roma, 6756100 Pisa, Italy
| | - Lionella Palego
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, Via Roma, 6756100 Pisa, Italy
| | - Gino Giannaccini
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, Via Roma, 6756100 Pisa, Italy
| | - Laura Betti
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, Via Roma, 6756100 Pisa, Italy
| | - Liliana Dell'Osso
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, Via Roma, 6756100 Pisa, Italy
| |
Collapse
|
11
|
Vendrik KEW, Ooijevaar RE, de Jong PRC, Laman JD, van Oosten BW, van Hilten JJ, Ducarmon QR, Keller JJ, Kuijper EJ, Contarino MF. Fecal Microbiota Transplantation in Neurological Disorders. Front Cell Infect Microbiol 2020; 10:98. [PMID: 32266160 PMCID: PMC7105733 DOI: 10.3389/fcimb.2020.00098] [Citation(s) in RCA: 209] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 02/26/2020] [Indexed: 12/14/2022] Open
Abstract
Background: Several studies suggested an important role of the gut microbiota in the pathophysiology of neurological disorders, implying that alteration of the gut microbiota might serve as a treatment strategy. Fecal microbiota transplantation (FMT) is currently the most effective gut microbiota intervention and an accepted treatment for recurrent Clostridioides difficile infections. To evaluate indications of FMT for patients with neurological disorders, we summarized the available literature on FMT. In addition, we provide suggestions for future directions. Methods: In July 2019, five main databases were searched for studies and case descriptions on FMT in neurological disorders in humans or animal models. In addition, the ClinicalTrials.gov website was consulted for registered planned and ongoing trials. Results: Of 541 identified studies, 34 were included in the analysis. Clinical trials with FMT have been performed in patients with autism spectrum disorder and showed beneficial effects on neurological symptoms. For multiple sclerosis and Parkinson's disease, several animal studies suggested a positive effect of FMT, supported by some human case reports. For epilepsy, Tourette syndrome, and diabetic neuropathy some studies suggested a beneficial effect of FMT, but evidence was restricted to case reports and limited numbers of animal studies. For stroke, Alzheimer's disease and Guillain-Barré syndrome only studies with animal models were identified. These studies suggested a potential beneficial effect of healthy donor FMT. In contrast, one study with an animal model for stroke showed increased mortality after FMT. For Guillain-Barré only one study was identified. Whether positive findings from animal studies can be confirmed in the treatment of human diseases awaits to be seen. Several trials with FMT as treatment for the above mentioned neurological disorders are planned or ongoing, as well as for amyotrophic lateral sclerosis. Conclusions: Preliminary literature suggests that FMT may be a promising treatment option for several neurological disorders. However, available evidence is still scanty and some contrasting results were observed. A limited number of studies in humans have been performed or are ongoing, while for some disorders only animal experiments have been conducted. Large double-blinded randomized controlled trials are needed to further elucidate the effect of FMT in neurological disorders.
Collapse
Affiliation(s)
- Karuna E W Vendrik
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, Netherlands.,Netherlands Donor Feces Bank, Leiden University Medical Center, Leiden, Netherlands.,Centre for Infectious Disease Control, National Institute for Public Health and the Environment (Rijksinstituut voor Volksgezondheid en Milieu, RIVM), Bilthoven, Netherlands
| | - Rogier E Ooijevaar
- Netherlands Donor Feces Bank, Leiden University Medical Center, Leiden, Netherlands.,Department of Gastroenterology, Amsterdam University Medical Centers, VU University Medical Center, Amsterdam, Netherlands
| | - Pieter R C de Jong
- Department of Neurology, Leiden University Medical Center, Leiden, Netherlands
| | - Jon D Laman
- Department Biomedical Sciences of Cells & Systems, University Medical Center Groningen, Groningen, Netherlands
| | - Bob W van Oosten
- Department of Neurology, Amsterdam University Medical Centers, VU University Medical Center, Amsterdam, Netherlands
| | | | - Quinten R Ducarmon
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, Netherlands.,Center for Microbiome Analyses and Therapeutics, Leiden University Medical Center, Leiden, Netherlands
| | - Josbert J Keller
- Netherlands Donor Feces Bank, Leiden University Medical Center, Leiden, Netherlands.,Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, Netherlands.,Department of Gastroenterology, Haaglanden Medical Center, The Hague, Netherlands
| | - Eduard J Kuijper
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, Netherlands.,Netherlands Donor Feces Bank, Leiden University Medical Center, Leiden, Netherlands.,Centre for Infectious Disease Control, National Institute for Public Health and the Environment (Rijksinstituut voor Volksgezondheid en Milieu, RIVM), Bilthoven, Netherlands.,Center for Microbiome Analyses and Therapeutics, Leiden University Medical Center, Leiden, Netherlands
| | - Maria Fiorella Contarino
- Department of Neurology, Leiden University Medical Center, Leiden, Netherlands.,Department of Neurology, Haga Teaching Hospital, The Hague, Netherlands
| |
Collapse
|
12
|
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
|
13
|
Thom RP, Keary CJ, Palumbo ML, Ravichandran CT, Mullett JE, Hazen EP, Neumeyer AM, McDougle CJ. Beyond the brain: A multi-system inflammatory subtype of autism spectrum disorder. Psychopharmacology (Berl) 2019; 236:3045-3061. [PMID: 31139876 DOI: 10.1007/s00213-019-05280-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 05/09/2019] [Accepted: 05/14/2019] [Indexed: 12/16/2022]
Abstract
An immune-mediated subtype of autism spectrum disorder (ASD) has long been hypothesized. This article reviews evidence from family history studies of autoimmunity, immunogenetics, maternal immune activation, neuroinflammation, and systemic inflammation, which suggests immune dysfunction in ASD. Individuals with ASD have higher rates of co-morbid medical illness than the general population. Major medical co-morbidities associated with ASD are discussed by body system. Mechanisms by which FDA-approved and emerging treatments for ASD act upon the immune system are then reviewed. We conclude by proposing the hypothesis of an immune-mediated subtype of ASD which is characterized by systemic, multi-organ inflammation or immune dysregulation with shared mechanisms that drive both the behavioral and physical illnesses associated with ASD. Although gaps in evidence supporting this hypothesis remain, benefits of this conceptualization include framing future research questions that will help define a clinically meaningful subset of patients and focusing clinical interactions on early detection and treatment of high-risk medical illnesses as well as interfering behavioral signs and symptoms across the lifespan.
Collapse
Affiliation(s)
- Robyn P Thom
- Massachusetts General Hospital, 55 Fruit St, Boston, MA, 02114, USA.,Department of Psychiatry, Harvard Medical School, 25 Shattuck St, Boston, MA, 02115, USA
| | - Christopher J Keary
- Massachusetts General Hospital, 55 Fruit St, Boston, MA, 02114, USA.,Department of Psychiatry, Harvard Medical School, 25 Shattuck St, Boston, MA, 02115, USA.,Lurie Center for Autism, 1 Maguire Road, Lexington, MA, 02421, USA
| | - Michelle L Palumbo
- Massachusetts General Hospital, 55 Fruit St, Boston, MA, 02114, USA.,Lurie Center for Autism, 1 Maguire Road, Lexington, MA, 02421, USA.,Department of Pediatrics, Harvard Medical School, 25 Shattuck St, Boston, MA, 02115, USA
| | - Caitlin T Ravichandran
- Massachusetts General Hospital, 55 Fruit St, Boston, MA, 02114, USA.,Lurie Center for Autism, 1 Maguire Road, Lexington, MA, 02421, USA.,Department of Pediatrics, Harvard Medical School, 25 Shattuck St, Boston, MA, 02115, USA
| | - Jennifer E Mullett
- Massachusetts General Hospital, 55 Fruit St, Boston, MA, 02114, USA.,Lurie Center for Autism, 1 Maguire Road, Lexington, MA, 02421, USA
| | - Eric P Hazen
- Massachusetts General Hospital, 55 Fruit St, Boston, MA, 02114, USA.,Department of Psychiatry, Harvard Medical School, 25 Shattuck St, Boston, MA, 02115, USA
| | - Ann M Neumeyer
- Massachusetts General Hospital, 55 Fruit St, Boston, MA, 02114, USA.,Lurie Center for Autism, 1 Maguire Road, Lexington, MA, 02421, USA.,Department of Neurology, Harvard Medical School, 25 Shattuck St, Boston, MA, 02115, USA
| | - Christopher J McDougle
- Massachusetts General Hospital, 55 Fruit St, Boston, MA, 02114, USA. .,Department of Psychiatry, Harvard Medical School, 25 Shattuck St, Boston, MA, 02115, USA. .,Lurie Center for Autism, 1 Maguire Road, Lexington, MA, 02421, USA.
| |
Collapse
|
14
|
Rose D, Ashwood P. Rapid Communication: Plasma Interleukin-35 in Children with Autism. Brain Sci 2019; 9:E152. [PMID: 31252635 PMCID: PMC6680732 DOI: 10.3390/brainsci9070152] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 06/21/2019] [Accepted: 06/24/2019] [Indexed: 12/29/2022] Open
Abstract
In autism spectrum disorders (ASD) many individuals have co-morbid immune dysregulation that can lead to inflammation in the brain and periphery. The novel cytokine interleukin (IL)-35 has described anti-inflammatory properties; however, the plasma levels of IL-35 in children with ASD have never been investigated. The plasma levels of IL-35 were measured by an enzyme-linked immunosorbent assay in 30 children with ASD and 39 typically developing (TD) controls. In the current study, we found that plasma IL-35 levels were significantly decreased in children with ASD compared with TD children. Furthermore, lower IL-35 levels were associated with worse behaviors as assessed using the aberrant behavior checklist. These findings are in line with other observations of decreased regulatory cytokines such as transforming growth factor beta and IL-10 in ASD, and associations with severity of behaviors. In conclusion, regulating the expression of IL-35 may provide a new possible target for the treatment of immune issues in ASD to address an imbalance between pro- and anti-inflammatory signals that alter the behavioral phenotype.
Collapse
Affiliation(s)
- Destanie Rose
- Department of Medical Microbiology and Immunology, and The Medical Investigation of Neurodevelopmental Disorders Institute, University of California, Davis, CA 95817, USA
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, and The Medical Investigation of Neurodevelopmental Disorders Institute, University of California, Davis, CA 95817, USA.
| |
Collapse
|
15
|
Ahmad SF, Ansari MA, Nadeem A, Bakheet SA, Al-Ayadhi LY, Attia SM. Elevated IL-16 expression is associated with development of immune dysfunction in children with autism. Psychopharmacology (Berl) 2019; 236:831-838. [PMID: 30456538 DOI: 10.1007/s00213-018-5120-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 11/09/2018] [Indexed: 11/25/2022]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by impairments in communication skills and social behaviors. Several studies have suggested that neuroimmune dysfunction plays a significant role in the pathogenesis of ASD; however, its exact etiology is unknown. Interleukin-16 (IL-16), a chemoattractant, is associated with various inflammatory processes. However, its role in children with ASD is unclear. This study aimed to investigate whether IL-16 expression is associated with immune dysfunction in children with ASD. We examined IL-16 expression in CD4+, CD8+, CD14+, CCR3+, and CXCR7+ cells in typically developing (TD) controls and children with ASD using flow cytometry in peripheral blood mononuclear cells (PBMCs). We also investigated the expression of IL-1β+IL-16+, IL-6+IL-16+, and TNF-α+IL-16+ in TD controls and children with ASD. We further explored IL-16 mRNA and protein expression using RT-PCR and western blotting. CD4+IL-16+, CD8+IL-16+, CD14+IL-16+, CCR3+IL-16+, and CXCR7+IL-16+ cells increased significantly in children with ASD compared with TD controls. We also showed that expression of IL-1β+IL-16+, IL-6+IL-16+, and TNF-α+IL-16+ was elevated in children with ASD compared with TD controls. Moreover, IL-16 mRNA and protein expression was significantly induced in children with ASD compared with TD controls. These results suggest that IL-16 expression could play an essential role in immune alteration in children with ASD.
Collapse
Affiliation(s)
- Sheikh F Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia.
- College of Pharmacy, King Saud University, 2457, Riyadh, 11451, Saudi Arabia.
| | - Mushtaq A Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Saleh A Bakheet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Laila Y Al-Ayadhi
- Autism Research and Treatment Center, AL-Amodi Autism Research Chair, Department of Physiology, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia
- Department of Pharmacology and Toxicology, College of Pharmacy, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
16
|
Carpenter KLH, Major S, Tallman C, Chen LW, Franz L, Sun J, Kurtzberg J, Song A, Dawson G. White Matter Tract Changes Associated with Clinical Improvement in an Open-Label Trial Assessing Autologous Umbilical Cord Blood for Treatment of Young Children with Autism. Stem Cells Transl Med 2019; 8:138-147. [PMID: 30620122 PMCID: PMC6344899 DOI: 10.1002/sctm.18-0251] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 11/19/2018] [Indexed: 12/26/2022] Open
Abstract
Autism spectrum disorder (ASD) is a heterogeneous neurodevelopmental disorder characterized by social communication deficits and the presence of restricted interests and repetitive behaviors. We have previously reported significant improvements in behavior, including increased social functioning, improved communication abilities, and decreased clinical symptoms in children with ASD, following treatment with a single infusion of autologous cord blood in a phase I open‐label trial. In the current study, we aimed to understand whether these improvements were associated with concurrent changes in brain structural connectivity. Twenty‐five 2‐ to 6‐year‐old children with ASD participated in this trial. Clinical outcome measures included the Vineland Adaptive Behavior Scales‐II Socialization Subscale, Expressive One‐Word Picture Vocabulary Test‐4, and the Clinical Global Impression‐Improvement Scale. Structural connectivity was measured at baseline and at 6 months in a subset of 19 children with 25‐direction diffusion tensor imaging and deterministic tractography. Behavioral improvements were associated with increased white matter connectivity in frontal, temporal, and subcortical regions (hippocampus and basal ganglia) that have been previously shown to show anatomical, connectivity, and functional abnormalities in ASD. The current results suggest that improvements in social communication skills and a reduction in symptoms in children with ASD following treatment with autologous cord blood infusion were associated with increased structural connectivity in brain networks supporting social, communication, and language abilities. stem cells translational medicine2019;8:138&10
Collapse
Affiliation(s)
- Kimberly L H Carpenter
- Duke Center for Autism and Brain Development, Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, North Carolina, USA
| | - Samantha Major
- Duke Center for Autism and Brain Development, Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, North Carolina, USA
| | - Catherine Tallman
- Brain Imaging and Analysis Center, Duke University Medical Center, Duke University School of Medicine, Durham, North Carolina, USA
| | - Lyon W Chen
- Brain Imaging and Analysis Center, Duke University Medical Center, Duke University School of Medicine, Durham, North Carolina, USA
| | - Lauren Franz
- Duke Center for Autism and Brain Development, Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, North Carolina, USA.,Duke Global Health Institute, Duke University School of Medicine, Duke University, Durham, North Carolina, USA
| | - Jessica Sun
- Marcus Center for Cellular Cures, Duke University Medical Center, Duke University School of Medicine, Durham, North Carolina, USA
| | - Joanne Kurtzberg
- Marcus Center for Cellular Cures, Duke University Medical Center, Duke University School of Medicine, Durham, North Carolina, USA
| | - Allen Song
- Brain Imaging and Analysis Center, Duke University Medical Center, Duke University School of Medicine, Durham, North Carolina, USA
| | - Geraldine Dawson
- Duke Center for Autism and Brain Development, Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, North Carolina, USA.,Marcus Center for Cellular Cures, Duke University Medical Center, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
17
|
Beversdorf DQ, Stevens HE, Margolis KG, Van de Water J. Prenatal Stress and Maternal Immune Dysregulation in Autism Spectrum Disorders: Potential Points for Intervention. Curr Pharm Des 2019; 25:4331-4343. [PMID: 31742491 PMCID: PMC7100710 DOI: 10.2174/1381612825666191119093335] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 11/15/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Genetics is a major etiological contributor to autism spectrum disorder (ASD). Environmental factors, however, also appear to contribute. ASD pathophysiology due to gene x environment is also beginning to be explored. One reason to focus on environmental factors is that they may allow opportunities for intervention or prevention. METHODS AND RESULTS Herein, we review two such factors that have been associated with a significant proportion of ASD risk, prenatal stress exposure and maternal immune dysregulation. Maternal stress susceptibility appears to interact with prenatal stress exposure to affect offspring neurodevelopment. We also explore how maternal stress may interact with the microbiome in the neurodevelopmental setting. Additionally, understanding of the impact of maternal immune dysfunction on ASD has recently been advanced by recognition of specific fetal brain proteins targeted by maternal autoantibodies, and identification of unique mid-gestational maternal immune profiles. This might also be interrelated with maternal stress exposure. Animal models have been developed to explore pathophysiology targeting each of these factors. CONCLUSION We are beginning to understand the behavioral, pharmacopathological, and epigenetic effects related to these interactions, and we are beginning to explore potential mitigating factors. Continued growth in understanding of these mechanisms may ultimately allow for the identification of multiple potential targets for prevention or intervention for this subset of environmental-associated ASD cases.
Collapse
Affiliation(s)
- David Q. Beversdorf
- Departments of Radiology, Neurology, and Psychological Sciences, and The Thompson Center for Neurodevelopmental Disorders, University of Missouri, William and Nancy Thompson Endowed Chair in Radiology
| | - Hanna E. Stevens
- Departments of Psychiatry and Pediatrics, Iowa Neuroscience Institute, University of Iowa
| | - Kara Gross Margolis
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology and Nutrition, Morgan Stanley Children’s Hospital, Columbia University Medical Center
| | - Judy Van de Water
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, And the MIND Institute, University of California, Davis
| |
Collapse
|
18
|
Hughes HK, Mills Ko E, Rose D, Ashwood P. Immune Dysfunction and Autoimmunity as Pathological Mechanisms in Autism Spectrum Disorders. Front Cell Neurosci 2018; 12:405. [PMID: 30483058 PMCID: PMC6242891 DOI: 10.3389/fncel.2018.00405] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 10/19/2018] [Indexed: 12/21/2022] Open
Abstract
Autism spectrum disorders (ASD) are a group of heterogeneous neurological disorders that are highly variable and are clinically characterized by deficits in social interactions, communication, and stereotypical behaviors. Prevalence has risen from 1 in 10,000 in 1972 to 1 in 59 children in the United States in 2014. This rise in prevalence could be due in part to better diagnoses and awareness, however, these together cannot solely account for such a significant rise. While causative connections have not been proven in the majority of cases, many current studies focus on the combined effects of genetics and environment. Strikingly, a distinct picture of immune dysfunction has emerged and been supported by many independent studies over the past decade. Many players in the immune-ASD puzzle may be mechanistically contributing to pathogenesis of these disorders, including skewed cytokine responses, differences in total numbers and frequencies of immune cells and their subsets, neuroinflammation, and adaptive and innate immune dysfunction, as well as altered levels of immunoglobulin and the presence of autoantibodies which have been found in a substantial number of individuals with ASD. This review summarizes the latest research linking ASD, autoimmunity and immune dysfunction, and discusses evidence of a potential autoimmune component of ASD.
Collapse
Affiliation(s)
- Heather K. Hughes
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA, United States
- MIND Institute, UC Davis Medical Center, Sacramento, CA, United States
| | - Emily Mills Ko
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA, United States
- MIND Institute, UC Davis Medical Center, Sacramento, CA, United States
| | - Destanie Rose
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA, United States
- MIND Institute, UC Davis Medical Center, Sacramento, CA, United States
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA, United States
- MIND Institute, UC Davis Medical Center, Sacramento, CA, United States
| |
Collapse
|
19
|
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
|
20
|
Bennabi M, Gaman A, Delorme R, Boukouaci W, Manier C, Scheid I, Si Mohammed N, Bengoufa D, Charron D, Krishnamoorthy R, Leboyer M, Tamouza R. HLA-class II haplotypes and Autism Spectrum Disorders. Sci Rep 2018; 8:7639. [PMID: 29769579 PMCID: PMC5955937 DOI: 10.1038/s41598-018-25974-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 04/26/2018] [Indexed: 02/06/2023] Open
Abstract
Infections and autoimmunity are associated with autism spectrum disorders (ASD), with both strongly influenced by the genetic regulation of the human leukocyte antigen (HLA) system. The relationship between ASD and the HLA genetic diversity requires further investigation. Using a case control design, the distribution of HLA class II-DRB1 and DQB1 alleles, genotypes and haplotypes were investigated in ASD patients, versus healthy controls (HC). ASD patients meeting DSM-IV TR criteria and HC (474 and 350 respectively) were genotyped at medium resolution using a Luminex-based SSO technology. Comparisons of genotypes, allele frequencies associated with a haplotype analysis were performed. Results indicate: (i) the HLA-DRB1 *11-DQB1*07 haplotype was more prevalent in ASD patients, versus HC (Pc = 0.001), partially replicating previous data and possibly linking to gastro-intestinal (GI)-related pro-inflammatory processes, given that this haplotype associates with pediatric celiac disorders; (ii) the HLA-DRB1 *17-DQB1*02 haplotype was higher in HC, versus ASD patients (Pc = 0.002), indicating that this is a protective haplotype. Using the Autism Diagnostic Interview to assess clinical dimensions, higher scores on social (Pc = 0.006) and non-verbal functioning (Pc = 0.004) associated with the DRB1 *11 DQB1*07 haplotype. Our results support HLA involvement in ASD, with possible relevance to GI and gut-brain axis dysregulation.
Collapse
Affiliation(s)
- Meriem Bennabi
- INSERM, U1160, Hôpital Saint Louis, Paris, France.,INSERM, U955, Psychiatrie Translationnelle, Créteil, France.,Fondation FondaMental, Créteil, France
| | - Alexandru Gaman
- INSERM, U955, Psychiatrie Translationnelle, Créteil, France.,Fondation FondaMental, Créteil, France
| | - Richard Delorme
- Fondation FondaMental, Créteil, France.,DHU Protect, AP-HP, Service de psychiatrie de l'enfant et de l'adolescent, Hôpital Robert Debré, Paris, France.,Département de génétique humaine et fonctions cognitives, Institut Pasteur, Paris, France.,Université Paris Diderot, Sorbonne Paris-Cité, Paris, France
| | | | | | - Isabelle Scheid
- INSERM, U955, Psychiatrie Translationnelle, Créteil, France.,Fondation FondaMental, Créteil, France
| | | | - Djaouida Bengoufa
- Laboratoire Jean Dausset and LabEx Transplantex, Hôpital Saint Louis, Paris, France
| | - Dominique Charron
- INSERM, U1160, Hôpital Saint Louis, Paris, France.,Université Paris Diderot, Sorbonne Paris-Cité, Paris, France.,Laboratoire Jean Dausset and LabEx Transplantex, Hôpital Saint Louis, Paris, France
| | - Rajagopal Krishnamoorthy
- INSERM, U955, Psychiatrie Translationnelle, Créteil, France.,Fondation FondaMental, Créteil, France
| | - Marion Leboyer
- INSERM, U955, Psychiatrie Translationnelle, Créteil, France.,Fondation FondaMental, Créteil, France.,DHU PePSY, AP-HP, Pôle de Psychiatrie, Hôpitaux Universitaires Henri Mondor, Créteil, France.,Université Paris-Est-Créteil, Faculté de médecine, Créteil, France
| | - Ryad Tamouza
- INSERM, U1160, Hôpital Saint Louis, Paris, France. .,INSERM, U955, Psychiatrie Translationnelle, Créteil, France. .,Fondation FondaMental, Créteil, France. .,Université Paris Diderot, Sorbonne Paris-Cité, Paris, France. .,Laboratoire Jean Dausset and LabEx Transplantex, Hôpital Saint Louis, Paris, France. .,DHU PePSY, AP-HP, Pôle de Psychiatrie, Hôpitaux Universitaires Henri Mondor, Créteil, France.
| |
Collapse
|
21
|
Hamed NO, Al-Ayadhi L, Osman MA, Elkhawad AO, Qasem H, Al-Marshoud M, Merghani NM, El-Ansary A. Understanding the roles of glutamine synthetase, glutaminase, and glutamate decarboxylase autoantibodies in imbalanced excitatory/inhibitory neurotransmission as etiological mechanisms of autism. Psychiatry Clin Neurosci 2018; 72:362-373. [PMID: 29356297 DOI: 10.1111/pcn.12639] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 12/06/2017] [Accepted: 01/10/2018] [Indexed: 12/26/2022]
Abstract
AIM Autism is a heterogeneous neurological disorder that is characterized by impairments in communication and social interactions, repetitive behaviors, and sensory abnormalities. The etiology of autism remains unclear. Animal, genetic, and post-mortem studies suggest that an imbalance exists in the neuronal excitation and inhibition system in autism. The aim of this study was to determine whether alterations of the measured parameters in children with autism are significantly associated with the risk of a sensory dysfunction. METHODS The glutamine synthetase (GS), kidney-type glutaminase (GLS1), and glutamic acid decarboxylase autoantibody levels were analyzed in 38 autistic children and 33 age- and sex-matched controls using enzyme-linked immunosorbent assays. RESULTS The obtained data demonstrated significant alterations in glutamate and glutamine cycle enzymes, as represented by GS and GLS1, respectively. While the glutamic acid decarboxylase autoantibodies levels were remarkably increased, no significant difference was observed compared to the healthy control participants. CONCLUSION The obtained data indicate that GS and GLS1 are promising indicators of a neuronal excitation and inhibition system imbalance and that combined measured parameters are good predictive biomarkers of autism.
Collapse
Affiliation(s)
- Najat O Hamed
- Department of Medical Biochemistry, University of Medical Sciences and Technology, Khartoum, Sudan.,Department of Pharmacology, Almaarefa Colleges for Science & Technology (MCST), Riyadh, Saudi Arabia
| | - Laila Al-Ayadhi
- Department of Physiology, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia.,Autism Research and Treatment Center, King Khalid University Hospital, Riyadh, Saudi Arabia.,Shaik AL-Amodi Autism Research Chair, King Saud University, Riyadh, Saudi Arabia
| | - Mohamed A Osman
- Department of Medical Biochemistry, University of Medical Sciences and Technology, Khartoum, Sudan.,Department of Pharmacology, Faculty of Pharmacy, University of Medical Sciences and Technology, Sudan Medical and Scientific Research Institute, Khartoum, Sudan
| | | | - Hanan Qasem
- Autism Research and Treatment Center, King Khalid University Hospital, Riyadh, Saudi Arabia
| | - Majida Al-Marshoud
- Central Laboratory, Female Centre for Scientific and Medical Studies, King Saud University, Riyadh, Saudi Arabia
| | - Nada M Merghani
- Central Laboratory, Female Centre for Scientific and Medical Studies, King Saud University, Riyadh, Saudi Arabia
| | - Afaf El-Ansary
- Autism Research and Treatment Center, King Khalid University Hospital, Riyadh, Saudi Arabia.,Shaik AL-Amodi Autism Research Chair, King Saud University, Riyadh, Saudi Arabia.,Central Laboratory, Female Centre for Scientific and Medical Studies, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
22
|
Abnormal Microglia and Enhanced Inflammation-Related Gene Transcription in Mice with Conditional Deletion of Ctcf in Camk2a-Cre-Expressing Neurons. J Neurosci 2017; 38:200-219. [PMID: 29133437 DOI: 10.1523/jneurosci.0936-17.2017] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 10/21/2017] [Accepted: 11/09/2017] [Indexed: 01/22/2023] Open
Abstract
CCCTC-binding factor (CTCF) is an 11 zinc finger DNA-binding domain protein that regulates gene expression by modifying 3D chromatin structure. Human mutations in CTCF cause intellectual disability and autistic features. Knocking out Ctcf in mouse embryonic neurons is lethal by neonatal age, but the effects of CTCF deficiency in postnatal neurons are less well studied. We knocked out Ctcf postnatally in glutamatergic forebrain neurons under the control of Camk2a-Cre. CtcfloxP/loxP;Camk2a-Cre+ (Ctcf CKO) mice of both sexes were viable and exhibited profound deficits in spatial learning/memory, impaired motor coordination, and decreased sociability by 4 months of age. Ctcf CKO mice also had reduced dendritic spine density in the hippocampus and cerebral cortex. Microarray analysis of mRNA from Ctcf CKO mouse hippocampus identified increased transcription of inflammation-related genes linked to microglia. Separate microarray analysis of mRNA isolated specifically from Ctcf CKO mouse hippocampal neurons by ribosomal affinity purification identified upregulation of chemokine signaling genes, suggesting crosstalk between neurons and microglia in Ctcf CKO hippocampus. Finally, we found that microglia in Ctcf CKO mouse hippocampus had abnormal morphology by Sholl analysis and increased immunostaining for CD68, a marker of microglial activation. Our findings confirm that Ctcf KO in postnatal neurons causes a neurobehavioral phenotype in mice and provide novel evidence that CTCF depletion leads to overexpression of inflammation-related genes and microglial dysfunction.SIGNIFICANCE STATEMENT CCCTC-binding factor (CTCF) is a DNA-binding protein that organizes nuclear chromatin topology. Mutations in CTCF cause intellectual disability and autistic features in humans. CTCF deficiency in embryonic neurons is lethal in mice, but mice with postnatal CTCF depletion are less well studied. We find that mice lacking Ctcf in Camk2a-expressing neurons (Ctcf CKO mice) have spatial learning/memory deficits, impaired fine motor skills, subtly altered social interactions, and decreased dendritic spine density. We demonstrate that Ctcf CKO mice overexpress inflammation-related genes in the brain and have microglia with abnormal morphology that label positive for CD68, a marker of microglial activation. Our findings suggest that inflammation and dysfunctional neuron-microglia interactions are factors in the pathology of CTCF deficiency.
Collapse
|
23
|
The Putative Role of Environmental Mercury in the Pathogenesis and Pathophysiology of Autism Spectrum Disorders and Subtypes. Mol Neurobiol 2017; 55:4834-4856. [DOI: 10.1007/s12035-017-0692-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Accepted: 07/13/2017] [Indexed: 01/28/2023]
|
24
|
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
|
25
|
Edmiston E, Ashwood P, Van de Water J. Autoimmunity, Autoantibodies, and Autism Spectrum Disorder. Biol Psychiatry 2017; 81:383-390. [PMID: 28340985 PMCID: PMC5373490 DOI: 10.1016/j.biopsych.2016.08.031] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 07/27/2016] [Accepted: 08/24/2016] [Indexed: 12/15/2022]
Abstract
Auism spectrum disorder (ASD) now affects one in 68 births in the United States and is the fastest growing neurodevelopmental disability worldwide. Alarmingly, for the majority of cases, the causes of ASD are largely unknown, but it is becoming increasingly accepted that ASD is no longer defined simply as a behavioral disorder, but rather as a highly complex and heterogeneous biological disorder. Although research has focused on the identification of genetic abnormalities, emerging studies increasingly suggest that immune dysfunction is a viable risk factor contributing to the neurodevelopmental deficits observed in ASD. This review summarizes the investigations implicating autoimmunity and autoantibodies in ASD.
Collapse
Affiliation(s)
- Elizabeth Edmiston
- Division of Rheumatology/Allergy and Clinical Immunology, University of California, Davis, Davis, California; The M.I.N.D. Institute, University of California, Davis, Davis, California
| | - Paul Ashwood
- The M.I.N.D. Institute, University of California, Davis, Davis, California; NIEHS Center for Children's Environmental Health, University of California, Davis, Davis, California; Department of Medical Microbiology and Immunology, University of California, Davis, Davis, California
| | - Judy Van de Water
- Division of Rheumatology/Allergy and Clinical Immunology, University of California, Davis, Davis, California; The M.I.N.D. Institute, University of California, Davis, Davis, California; NIEHS Center for Children's Environmental Health, University of California, Davis, Davis, California.
| |
Collapse
|
26
|
Neymotin F, Nemzer LR. Linking autism and epilepsy. CHILDRENS HEALTH CARE 2016. [DOI: 10.1080/02739615.2014.979921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
27
|
Klyushnik TP, Androsova LV, Simashkova NV, Zozulya SA, Otman IN, Shushpanova OV, Brusov OS. Clinical and laboratory diagnosis of autism spectrum disorders in children. ACTA ACUST UNITED AC 2016. [DOI: 10.17116/labs20165222-27] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
28
|
Immune mediators in the brain and peripheral tissues in autism spectrum disorder. Nat Rev Neurosci 2015; 16:469-86. [PMID: 26189694 DOI: 10.1038/nrn3978] [Citation(s) in RCA: 328] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Increasing evidence points to a central role for immune dysregulation in autism spectrum disorder (ASD). Several ASD risk genes encode components of the immune system and many maternal immune system-related risk factors--including autoimmunity, infection and fetal reactive antibodies--are associated with ASD. In addition, there is evidence of ongoing immune dysregulation in individuals with ASD and in animal models of this disorder. Recently, several molecular signalling pathways--including pathways downstream of cytokines, the receptor MET, major histocompatibility complex class I molecules, microglia and complement factors--have been identified that link immune activation to ASD phenotypes. Together, these findings indicate that the immune system is a point of convergence for multiple ASD-related genetic and environmental risk factors.
Collapse
|
29
|
Jaiswal P, Mohanakumar KP, Rajamma U. Serotonin mediated immunoregulation and neural functions: Complicity in the aetiology of autism spectrum disorders. Neurosci Biobehav Rev 2015; 55:413-31. [PMID: 26021727 DOI: 10.1016/j.neubiorev.2015.05.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 05/11/2015] [Accepted: 05/12/2015] [Indexed: 12/19/2022]
Abstract
Serotonergic system has long been implicated in the aetiology of autism spectrum disorders (ASD), since platelet hyperserotonemia is consistently observed in a subset of autistic patients, who respond well to selective serotonin reuptake inhibitors. Apart from being a neurotransmitter, serotonin functions as a neurotrophic factor directing brain development and as an immunoregulator modulating immune responses. Serotonin transporter (SERT) regulates serotonin level in lymphoid tissues to ensure its proper functioning in innate and adaptive responses. Immunological molecules such as cytokines in turn regulate the transcription and activity of SERT. Dysregulation of serotonergic system could trigger signalling cascades that affect normal neural-immune interactions culminating in neurodevelopmental and neural connectivity defects precipitating behavioural abnormalities, or the disease phenotypes. Therefore, we suggest that a better understanding of the cross talk between serotonergic genes, immune systems and serotonergic neurotransmission will open wider avenues to develop pharmacological leads for addressing the core ASD behavioural deficits.
Collapse
Affiliation(s)
- Preeti Jaiswal
- Manovikas Biomedical Research and Diagnostic Centre, Manovikas Kendra, 482 Madudah, Plot I-24, Sector-J, EM Bypass, Kolkata 700 107, India
| | - Kochupurackal P Mohanakumar
- Division of Cell Biology & Physiology, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata 700 032, India
| | - Usha Rajamma
- Manovikas Biomedical Research and Diagnostic Centre, Manovikas Kendra, 482 Madudah, Plot I-24, Sector-J, EM Bypass, Kolkata 700 107, India.
| |
Collapse
|
30
|
Magid-Bernstein J, Mahajan K, Lincoln J, Ming X, Rohowsky-Kochan C. Case report: cytokine and CD4+ T-cell profiles of monozygotic twins with autism and divergent comorbidities and drug treatment. J Child Neurol 2015; 30:386-90. [PMID: 24736120 DOI: 10.1177/0883073814529821] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Autism spectrum disorders are neurodevelopmental disorders that are thought to be caused by a gene-by-environment interaction and in which various immune alterations are reported. We investigate CD4(+) T-cell cytokine profiles and subpopulations in 19-year-old monozygotic twins with autism and different comorbidities. CD4(+) T cells from the twin with epilepsy produce more interferon-gamma, less interleukin-17, and have an increased interferon-γ/interleukin-4 ratio. CD4(+) T cells from the twin with multiple sclerosis exhibit a cytokine profile similar to an age and gender-matched control and a higher percentage of T regulatory (Treg) cells. The twins' mother's T cells produce very high levels of both interleukin-17 and interferon-γ. Cytokine and CD4(+) T-cell abnormalities in the twins could contribute to or be a result of the manifestation of their divergent comorbidities. A proinflammatory, autoimmune-polarized cytokine profile is observed in this unique family with autism.
Collapse
Affiliation(s)
- Jessica Magid-Bernstein
- Department of Neurology and Neurosciences, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, NJ, USA
| | - Kedar Mahajan
- Department of Neurology, Thomas Jefferson University Hospitals, Philadelphia, PA, USA
| | - John Lincoln
- Department of Neurology, The University of Texas Medical School, Houston, TX, USA
| | - Xue Ming
- Department of Neurology and Neurosciences, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, NJ, USA
| | - Christine Rohowsky-Kochan
- Department of Neurology and Neurosciences, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, NJ, USA
| |
Collapse
|
31
|
A DeMarshall C, Sarkar A, G Nagele R. Serum Autoantibodies as Biomarkers for Parkinsons Disease: Background and Utility. AIMS MEDICAL SCIENCE 2015. [DOI: 10.3934/medsci.2015.4.316] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
32
|
Lisik MZ, Gutmajster E, Sieroń AL. Anti-neuronal antibodies in patients with fragile X syndrome: is there a role of autoimmunity in its pathogenesis? NEURODEGENER DIS 2014; 15:45-9. [PMID: 25500855 DOI: 10.1159/000369215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 10/20/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Fragile X syndrome (FXS) is a single-gene disorder with a broad spectrum of involvement, including cognitive and behavioural impairments of varying degrees with specific physical features and a strong association with autism. OBJECTIVES In this study, the frequency of serum anti-neural antibodies was investigated in FXS patients who did and those who did not manifest autism spectrum disorders (ASD) in comparison to typically developing controls. METHODS The study involved 23 males (mean age, 19.78 ± 6.56 years) who harboured a full mutation in the FMR1 gene. The control group comprised 19 healthy students (mean age 24.63 ± 1.89 years). Serum anti-neuronal antibodies were analyzed using Western blotting. RESULTS Serum anti-neuronal antibodies were present in 10/23 (43.48%) FXS males. CONCLUSION Serum anti-neuronal antibodies were found in a subgroup of FXS patients. Autistic symptoms in FXS may, in part, be caused by auto-immune factors. Further studies in larger patient and control groups are necessary to elucidate the aetiopathogenic role of anti-neuronal antibodies in FXS patients.
Collapse
Affiliation(s)
- Małgorzata Zofia Lisik
- Department of Molecular Biology and Genetics, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | | | | |
Collapse
|
33
|
Mead J, Ashwood P. Evidence supporting an altered immune response in ASD. Immunol Lett 2014; 163:49-55. [PMID: 25448709 DOI: 10.1016/j.imlet.2014.11.006] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 11/07/2014] [Accepted: 11/11/2014] [Indexed: 11/17/2022]
Abstract
Autism spectrum disorders (ASD) are neurodevelopmental disorders characterized by deficits in social interactions, communication, and increased stereotypical repetitive behaviors. The immune system plays an important role in neurodevelopment, regulating neuronal proliferation, synapse formation and plasticity, as well as removing apoptotic neurons. Immune dysfunction in ASD has been repeatedly described by many research groups across the globe. Symptoms of immune dysfunction in ASD include neuroinflammation, presence of autoantibodies, increased T cell responses, and enhanced innate NK cell and monocyte immune responses. Moreover these responses are frequently associated with more impairment in core ASD features including impaired social interactions, repetitive behaviors and communication. In mouse models replacing immune components in animals that exhibit autistic relevant features leads to improvement in behavior in these animals. Taken together this research suggests that the immune dysfunction often seen in ASD directly affects aspects of neurodevelopment and neurological processes leading to changes in behavior. Discussion of immune abnormalities in ASD will be the focus of this review.
Collapse
Affiliation(s)
- Jennifer Mead
- Department of Medical Microbiology and Immunology, UC Davis, CA, USA; The M.I.N.D. Institute, University of California at Davis, CA, USA
| | - Paul 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
|
34
|
Piras I, Haapanen L, Napolioni V, Sacco R, Van de Water J, Persico A. Anti-brain antibodies are associated with more severe cognitive and behavioral profiles in Italian children with Autism Spectrum Disorder. Brain Behav Immun 2014; 38:91-9. [PMID: 24389156 PMCID: PMC4111628 DOI: 10.1016/j.bbi.2013.12.020] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 12/10/2013] [Accepted: 12/24/2013] [Indexed: 12/22/2022] Open
Abstract
Circulating 45 and 62kDa antibodies targeting the cerebellum were previously associated with Autism Spectrum Disorder (ASD), lower adaptive/cognitive function and aberrant behaviors. Moreover, 37, 39 and 73kDa maternal antibodies (mAb) targeting the fetal brain were previously correlated with broad autism spectrum, irritability, abnormal brain enlargement and impaired expressive language. The present study aims towards clinically characterizing individuals with brain-targeted IgG and/or exposed to maternal antibrain antibodies in a large sample of Italian autistic children (N=355), their unaffected siblings (N=142) and mothers (N=333). The presence of patient- and mother-produced anti-brain antibodies does not confer increased risk of autism within the same sibship. However, the 45 and 62kDa antibodies are correlated with autism severity: the 45kDa Ab is associated with cognitive impairment and lower scores at the Vineland Adaptive Behavior Scales, the 62kDa Ab with motor stereotypies, while both correlate with larger head circumference (all P<0.05). On the other hand, maternal 37, 39 and 73kDa antibrain antibodies, either alone or in combination, are correlated with impaired verbal and non-verbal language development, neurodevelopmental delay and sleep/wake cycle disturbances in their autistic children (P<0.05). Presence of the 62kDa autoAb in the child is significantly associated with presence of the 39 and/or 73kDa antibodies in his/her mother. Our results confirm and extend previous observations in an ethnically distinct sample, providing further evidence of a pathomorphic role for anti-brain antibodies in autism while demonstrating their familial clustering.
Collapse
Affiliation(s)
- I.S. Piras
- Unit of Child and Adolescent NeuroPsychiatry, Laboratory of Molecular Psychiatry and Neurogenetics, University “Campus Bio-Medico”, Rome, Italy
| | - L. Haapanen
- Department of Internal Medicine, University of California, Davis, Davis, CA, USA,University of California, Davis M.I.N.D. Institute, Davis, CA, USA,Children’s Center for Environmental Health, University of California, Davis, Davis, CA, USA
| | - V. Napolioni
- Unit of Child and Adolescent NeuroPsychiatry, Laboratory of Molecular Psychiatry and Neurogenetics, University “Campus Bio-Medico”, Rome, Italy
| | - R. Sacco
- Unit of Child and Adolescent NeuroPsychiatry, Laboratory of Molecular Psychiatry and Neurogenetics, University “Campus Bio-Medico”, Rome, Italy,Department of Experimental Neurosciences, I.R.C.C.S. “Fondazione Santa Lucia”, Rome, Italy
| | - J. Van de Water
- Department of Internal Medicine, University of California, Davis, Davis, CA, USA,University of California, Davis M.I.N.D. Institute, Davis, CA, USA,Children’s Center for Environmental Health, University of California, Davis, Davis, CA, USA
| | - A.M. Persico
- Unit of Child and Adolescent NeuroPsychiatry, Laboratory of Molecular Psychiatry and Neurogenetics, University “Campus Bio-Medico”, Rome, Italy,Department of Experimental Neurosciences, I.R.C.C.S. “Fondazione Santa Lucia”, Rome, Italy,Mafalda Luce Center for Pervasive Developmental Disorders, Milan, Italy,Corresponding author at: Unit of Child and Adolescent NeuroPsychiatry, Laboratory of Molecular Psychiatry and Neurogenetics, University “Campus Bio-Medico”, Via Àlvaro del Portillo 21, Rome, Italy. Tel.: +39 06225419155. (A.M. Persico)
| |
Collapse
|
35
|
Mazur-Kolecka B, Cohen IL, Gonzalez M, Jenkins EC, Kaczmarski W, Brown WT, Flory M, Frackowiak J. Autoantibodies against neuronal progenitors in sera from children with autism. Brain Dev 2014; 36:322-9. [PMID: 23838310 DOI: 10.1016/j.braindev.2013.04.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 01/09/2013] [Accepted: 04/30/2013] [Indexed: 12/13/2022]
Abstract
The pathological role of autoantibodies in development of CNS disorders is a new idea with growing interest among neuroscientists. The involvement of autoimmune response in the pathogenesis of autism spectrum disorders (ASD) has been suggested by the presence of multiple brain-specific autoantibodies in children with ASD and in their mothers. The possibility of the effect of autoimmunity on neurogenesis and postnatal brain plasticity has not been determined. The presence of autoantibodies against human neuronal progenitor cells (NPCs) stimulated for neuronal differentiation in culture was tested in sera from children with autism (n=20) and age-matched controls (n=18) by immunoblotting and immunocytochemistry. Immunoreactivity against multiple NPCs proteins of molecular sizes of approximately 55 kDa, 105 kDa, 150 kDa, and 210 kDa in sera from individuals with autism had a higher incidence and was stronger than in control sera which immunoreacted mainly with a 150 kDa protein. The sera from children with autism immunoreacted the strongest with NPCs expressing neuronal markers Tuj1 and doublecortin, but not astrocyte marker GFAP. The epitopes recognized by antibodies from sera were not human-specific because they detected also NPCs in situ in murine hippocampus. The autoimmune reactions against NPCs suggest an impaired tolerance to neural antigens in autism. These autoantibodies may be symptomatic for autism and furthermore, their presence suggests that autoimmunity may affect postnatal neuronal plasticity particularly after impairment of blood-brain barrier. Future studies will determine the diagnostic value of the presence of autoantibodies in autism and the therapeutic value of prevention of autoimmunity in autism.
Collapse
Affiliation(s)
- Bozena Mazur-Kolecka
- Department of Developmental Neurobiology, New York State Institute for Basic Research in Developmental Disabilities, USA.
| | | | | | | | - Wojciech Kaczmarski
- Department of Developmental Neurobiology, New York State Institute for Basic Research in Developmental Disabilities, USA
| | - W Ted Brown
- Department of Human Genetics, NYS IBRDD, USA
| | - Michael Flory
- Laboratory of Research Design and Analysis, NYS IBRDD, USA
| | - Janusz Frackowiak
- Department of Developmental Neurobiology, New York State Institute for Basic Research in Developmental Disabilities, USA
| |
Collapse
|
36
|
Cannell JJ, Grant WB. What is the role of vitamin D in autism? DERMATO-ENDOCRINOLOGY 2014; 5:199-204. [PMID: 24494055 PMCID: PMC3897590 DOI: 10.4161/derm.24356] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Revised: 03/03/2013] [Accepted: 03/18/2013] [Indexed: 12/24/2022]
Abstract
A growing body of literature suggests that higher serum 25-hydroxyvitamin D [25(OH)D] concentrations, either in utero or in early life, may reduce the risk of autism. For example, an ecological study in the companion paper inversely correlated solar UV-B doses in the United States with prevalence of autism among those aged 6–17 y. That study proposed that vitamin D deficiency during pregnancy could account for this finding, although the findings are also consistent with childhood vitamin D deficiency contributing to the condition. Also, in a recent study, children with autism had lower serum 25(OH)D concentrations than did control subjects (19 vs. 33 ng/ml), despite parents of each group reporting the same amount of sun exposure. The same study found highly significant inverse correlations between 25(OH)D and autism rating scales and between 25(OH)D and levels of an antineuronal antibody. This finding indicates that higher serum 25(OH)D concentrations may reduce the symptoms of established autism. Because activated vitamin D, a secosteroid, upregulates DNA-repair genes, vitamin D deficiency during development may inhibit the repair of de novo DNA mutations in fetuses and infants and thus contribute to risk of autism. Vitamin D might also reduce the risk or severity of autism through its anti-inflammatory actions, antiautoimmune effects, increasing seizure threshold, increasing T-regulatory cells, protecting the mitochondria, and upregulating glutathione, which scavenges oxidative by-products and chelates (captures and excretes) heavy metals. Vitamin D deficiency during pregnancy and childhood is a widespread and growing epidemic.
Collapse
Affiliation(s)
| | - William B Grant
- Sunlight, Nutrition, and Health Research Center; San Francisco, CA USA
| |
Collapse
|
37
|
Goldani AAS, Downs SR, Widjaja F, Lawton B, Hendren RL. Biomarkers in autism. Front Psychiatry 2014; 5:100. [PMID: 25161627 PMCID: PMC4129499 DOI: 10.3389/fpsyt.2014.00100] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 07/22/2014] [Indexed: 12/20/2022] Open
Abstract
Autism spectrum disorders (ASDs) are complex, heterogeneous disorders caused by an interaction between genetic vulnerability and environmental factors. In an effort to better target the underlying roots of ASD for diagnosis and treatment, efforts to identify reliable biomarkers in genetics, neuroimaging, gene expression, and measures of the body's metabolism are growing. For this article, we review the published studies of potential biomarkers in autism and conclude that while there is increasing promise of finding biomarkers that can help us target treatment, there are none with enough evidence to support routine clinical use unless medical illness is suspected. Promising biomarkers include those for mitochondrial function, oxidative stress, and immune function. Genetic clusters are also suggesting the potential for useful biomarkers.
Collapse
Affiliation(s)
| | - Susan R Downs
- Department of Psychiatry, University of California San Francisco , San Francisco, CA , USA
| | - Felicia Widjaja
- Department of Psychiatry, University of California San Francisco , San Francisco, CA , USA
| | - Brittany Lawton
- Department of Psychiatry, University of California San Francisco , San Francisco, CA , USA
| | - Robert L Hendren
- Department of Psychiatry, University of California San Francisco , San Francisco, CA , USA
| |
Collapse
|
38
|
Noriega DB, Savelkoul HFJ. Immune dysregulation in autism spectrum disorder. Eur J Pediatr 2014; 173:33-43. [PMID: 24297668 DOI: 10.1007/s00431-013-2183-4] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 10/09/2013] [Indexed: 12/25/2022]
Abstract
UNLABELLED Autism spectrum disorder (ASD) is a common and severe neuro-developmental disorder in early childhood which is defined by social and communication deficits and repetitive and stereotypic behaviours. The aetiology of ASD remains poorly understood. Susceptibility to development of ASD has significant environmental components, in addition to the profound genetic heritability. Few genes have been associated to the risk for ASD development. There is substantial evidence implicating chronic neurological inflammation and immune dysregulation leading to upregulation of inflammatory cytokines in the ASD brain, probably due to altered blood-brain barrier function. The immune system is characterized by excessive and skewed cytokine responses, modulated T cell reactivity, decreased regulation and production of immunosuppressive cytokines, modified NK function and increased autoantibody production. CONCLUSION The perinatal environment generates vulnerability to chronic neuro-inflammation in the brain associated with profound modulation and dysregulation in the immune system leading to the rapid development of ASD in genetically susceptible children.
Collapse
Affiliation(s)
- Daniela Briceno Noriega
- Cell Biology and Immunology Group, Wageningen University, P.O. Box 338, 6700 AH, Wageningen, The Netherlands
| | | |
Collapse
|
39
|
Abstract
Autism has been classically defined by its behavioral symptoms. Traditional medical research has focused on genetic or intrinsic brain-based causes of autism. While both of these are important, additional research has focused on the underlying disordered biochemistry seen in many individuals with autism. Many of these biomedical factors are amenable to treatment. This article will review the main pathophysiologic factors seen in individuals with autism spectrum disorders.
Collapse
|
40
|
Rossi CC, Fuentes J, Van de Water J, Amaral DG. Brief Report: Antibodies Reacting to Brain Tissue in Basque Spanish Children with Autism Spectrum Disorder and Their Mothers. J Autism Dev Disord 2013; 44:10.1007/s10803-013-1859-y. [PMID: 24022729 PMCID: PMC3980136 DOI: 10.1007/s10803-013-1859-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Previous investigations found that a subset of children with autism spectrum disorder (ASD) in California possessed plasma autoantibodies that reacted intensely with brain interneurons or other neural profiles. Moreover, for several cohorts of American women, maternal autoantibody reactivity to specific fetal brain proteins was highly specific to mothers of children with ASD. We sought to determine whether children and their mothers from a regionally specific cohort from the Basque Country of Spain demonstrated similar reactivity. Some children's plasma reacted to interneurons, beaded axons or other neural profiles with no difference in the occurrence of these antibodies in children with or without ASD. Findings on the maternal antibodies confirmed previous research; plasma reactivity to fetal brain a combination of proteins at 37 and 73 kDa or 39 and 73 kDa was found exclusively in mothers of children with ASD.
Collapse
Affiliation(s)
- Christy C. Rossi
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Sacramento, CA, USA, The MIND Institute, University of California, Davis, 2825 50th Street, Sacramento, CA 95817, USA, Department of Psychology, University of Denver, Denver, CO, USA
| | - Joaquin Fuentes
- Child and Adolescent Psychiatry Unit, Policlinica Guipuzkoa and Gautena Autism Society, Donostia/San Sebastián, Spain
| | - Judy Van de Water
- The MIND Institute, University of California, Davis, 2825 50th Street, Sacramento, CA 95817, USA, Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, Sacramento, CA, USA, NIEHS Center for Children’s Environmental Health, University of California, Davis, Sacramento, CA, USA
| | - David G. Amaral
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Sacramento, CA, USA, The MIND Institute, University of California, Davis, 2825 50th Street, Sacramento, CA 95817, USA, Center for Neuroscience and California National Primate Research Center, University of California, Davis, Sacramento, CA, USA
| |
Collapse
|
41
|
Gesundheit B, Rosenzweig JP, Naor D, Lerer B, Zachor DA, Procházka V, Melamed M, Kristt DA, Steinberg A, Shulman C, Hwang P, Koren G, Walfisch A, Passweg JR, Snowden JA, Tamouza R, Leboyer M, Farge-Bancel D, Ashwood P. Immunological and autoimmune considerations of Autism Spectrum Disorders. J Autoimmun 2013; 44:1-7. [PMID: 23867105 DOI: 10.1016/j.jaut.2013.05.005] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 05/26/2013] [Accepted: 05/30/2013] [Indexed: 12/22/2022]
Abstract
Autism Spectrum Disorders (ASD) are a group of heterogeneous neurodevelopmental conditions presenting in early childhood with a prevalence ranging from 0.7% to 2.64%. Social interaction and communication skills are impaired and children often present with unusual repetitive behavior. The condition persists for life with major implications for the individual, the family and the entire health care system. While the etiology of ASD remains unknown, various clues suggest a possible association with altered immune responses and ASD. Inflammation in the brain and CNS has been reported by several groups with notable microglia activation and increased cytokine production in postmortem brain specimens of young and old individuals with ASD. Moreover several laboratories have isolated distinctive brain and CNS reactive antibodies from individuals with ASD. Large population based epidemiological studies have established a correlation between ASD and a family history of autoimmune diseases, associations with MHC complex haplotypes, and abnormal levels of various inflammatory cytokines and immunological markers in the blood. In addition, there is evidence that antibodies that are only present in some mothers of children with ASD bind to fetal brain proteins and may be a marker or risk factor for ASD. Studies involving the injection of these ASD specific maternal serum antibodies into pregnant mice during gestation, or gestational exposure of Rhesus monkeys to IgG subclass of these antibodies, have consistently elicited behavioral changes in offspring that have relevance to ASD. We will summarize the various types of studies associating ASD with the immune system, critically evaluate the quality of these studies, and attempt to integrate them in a way that clarifies the areas of immune and autoimmune phenomena in ASD research that will be important indicators for future research.
Collapse
|
42
|
Zhang Y, Gao D, Kluetzman K, Mendoza A, Bolivar VJ, Reilly A, Jolly JK, Lawrence DA. The maternal autoimmune environment affects the social behavior of offspring. J Neuroimmunol 2013; 258:51-60. [DOI: 10.1016/j.jneuroim.2013.02.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Revised: 02/23/2013] [Accepted: 02/26/2013] [Indexed: 12/28/2022]
|
43
|
Nordahl CW, Braunschweig D, Iosif AM, Lee A, Rogers S, Ashwood P, Amaral DG, Van de Water J. Maternal autoantibodies are associated with abnormal brain enlargement in a subgroup of children with autism spectrum disorder. Brain Behav Immun 2013; 30:61-5. [PMID: 23395715 PMCID: PMC3641177 DOI: 10.1016/j.bbi.2013.01.084] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 12/05/2012] [Accepted: 01/28/2013] [Indexed: 11/27/2022] Open
Abstract
Autism spectrum disorder (ASD) is very heterogeneous and multiple subtypes and etiologies likely exist. The maternal immune system has been implicated in the pathogenesis of some forms of ASD. Previous studies have identified the presence of specific maternal IgG autoantibodies with reactivity to fetal brain proteins at 37 and 73kDa in up to 12% of mothers of children with ASD. The current study evaluates the presence of these autoantibodies in an independent cohort of mothers of 181 preschool-aged male children (131 ASD, 50 typically developing (TD) controls). We also investigated whether ASD children born to mothers with these autism-specific maternal IgG autoantibodies exhibit a distinct neural phenotype by evaluating total brain volume using structural magnetic resonance imaging (MRI). Of the 131 ASD children, 10 (7.6%) were born to mothers with the 37/73kDa IgG autoantibodies (ASD-IgG). The mothers of the remaining ASD children and all TD controls were negative for these paired autoantibodies. While both ASD groups exhibited abnormal brain enlargement that is commonly observed in this age range, the ASD-IgG group exhibited a more extreme 12.1% abnormal brain enlargement relative to the TD controls. In contrast, the remaining ASD children exhibited a smaller 4.4% abnormal brain enlargement relative to TD controls. Lobar and tissue type analyses revealed that the frontal lobe is selectively enlarged in the ASD-IgG group and that both gray and white matter are similarly affected. These results suggest that maternal autoantibodies associated with autism spectrum disorder may impact brain development leading to abnormal enlargement.
Collapse
Affiliation(s)
- Christine Wu Nordahl
- The M.I.N.D. Institute, University of California at Davis, School of Medicine, Sacramento, CA 95817, USA.
| | - Daniel Braunschweig
- Department of Internal Medicine, University of California at Davis, School of Medicine, Davis, CA 95616
| | - Ana-Maria Iosif
- Department of Public Health Sciences, Division of Biostatistics, University of California at Davis, Davis, CA 95616
| | - Aaron Lee
- Department of Psychiatry and Behavioral Sciences, University of California at Davis, School of Medicine, Sacramento, CA 95817
| | - Sally Rogers
- The M.I.N.D Institute, University of California at Davis, School of Medicine, Sacramento, CA 95817,Department of Psychiatry and Behavioral Sciences, University of California at Davis, School of Medicine, Sacramento, CA 95817
| | - Paul Ashwood
- The M.I.N.D Institute, University of California at Davis, School of Medicine, Sacramento, CA 95817,Department of Internal Medicine, University of California at Davis, School of Medicine, Davis, CA 95616
| | - David G. Amaral
- The M.I.N.D Institute, University of California at Davis, School of Medicine, Sacramento, CA 95817,Department of Psychiatry and Behavioral Sciences, University of California at Davis, School of Medicine, Sacramento, CA 95817
| | - Judy Van de Water
- The M.I.N.D Institute, University of California at Davis, School of Medicine, Sacramento, CA 95817,Department of Internal Medicine, University of California at Davis, School of Medicine, Davis, CA 95616
| |
Collapse
|
44
|
Blossom SJ, Cooney CA, Melnyk SB, Rau JL, Swearingen CJ, Wessinger WD. Metabolic changes and DNA hypomethylation in cerebellum are associated with behavioral alterations in mice exposed to trichloroethylene postnatally. Toxicol Appl Pharmacol 2013; 269:263-9. [PMID: 23566951 DOI: 10.1016/j.taap.2013.03.025] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 03/16/2013] [Accepted: 03/18/2013] [Indexed: 02/07/2023]
Abstract
Previous studies demonstrated that low-level postnatal and early life exposure to the environmental contaminant, trichloroethylene (TCE), in the drinking water of MRL+/+ mice altered glutathione redox homeostasis and increased biomarkers of oxidative stress indicating a more oxidized state. Plasma metabolites along the interrelated transmethylation pathway were also altered indicating impaired methylation capacity. Here we extend these findings to further characterize the impact of TCE exposure in mice exposed to water only or two doses of TCE in the drinking water (0, 2, and 28mg/kg/day) postnatally from birth until 6weeks of age on redox homeostasis and biomarkers of oxidative stress in the cerebellum. In addition, pathway intermediates involved in methyl metabolism and global DNA methylation patterns were examined in cerebellar tissue. Because the cerebellum is functionally important for coordinating motor activity, including exploratory and social approach behaviors, these parameters were evaluated in the present study. Mice exposed to 28mg/kg/day TCE exhibited increased locomotor activity over time as compared with control mice. In the novel object exploration test, these mice were more likely to enter the zone with the novel object as compared to control mice. Similar results were obtained in a second test when an unfamiliar mouse was introduced into the testing arena. The results show for the first time that postnatal exposure to TCE causes key metabolic changes in the cerebellum that may contribute to global DNA methylation deficits and behavioral alterations in TCE-exposed mice.
Collapse
Affiliation(s)
- Sarah J Blossom
- Department of Pediatrics, University of Arkansas for Medical Sciences, College of Medicine, Arkansas Children's Hospital Research Institute, 13 Children's Way, Little Rock, AR 72202, USA.
| | | | | | | | | | | |
Collapse
|
45
|
Zhang Y, Bolivar VJ, Lawrence DA. Maternal exposure to mercury chloride during pregnancy and lactation affects the immunity and social behavior of offspring. Toxicol Sci 2013; 133:101-11. [PMID: 23392568 DOI: 10.1093/toxsci/kft023] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Developmental HgCl2 exposures of F1 offspring (H-2(q/s)) from unsociable SJL/J (H-2(s)) dams with high susceptibility to Hg-induced autoimmunity (SFvF1) and from highly sociable FVB/NJ (FVB; H-2(q)) dams with lower susceptibility to Hg-induced autoimmunity (FvSF1) were investigated. Hg exposure increased the serum IgG levels of all offspring at postnatal day 21 (pnd21) and of SJL/J dams but not of FVB dams. Serum IgG anti-brain antibody (Ab) levels of pnd21 SFvF1 offspring and SJL dams were higher than those of the FvSF1 offspring and FVB dams, but Hg only increased the titers of the FVB dams and their offspring. Hg significantly elevated the presence of IgG in all brain regions of the pnd21 SFvF1 offspring, and the SFvF1 offspring had greater amounts of IgG in the brain than the FvSF1 offspring, which had Hg-induced increases in only two brain regions. Cytokine levels were elevated in the brain regions of Hg-treated pnd21 SFvF1 but not of FvSF1 offspring, and SFvF1 females had more brain regions expressing cytokines than the males. At pnd70, the serum IgG, serum antibrain Abs, amounts of brain IgG, and brain cytokine levels of all of the Hg-treated offspring were equivalent to those of their appropriate controls, suggesting that developmental Hg exposure did not induce chronic immunological effects. However, the social behaviors of Hg-exposed SFvF1 offspring at pnd70 were significantly impaired, and SFvF1 females displayed greater decline in social behaviors than males, suggesting that the higher neuroinflammation of SFvF1 females earlier in life is associated with the altered behavior. Thus, developmental Hg exposure induces long-lasting effects on social behavior of offspring, which is dependent on sex and genetics and the induction of neuroinflammation.
Collapse
Affiliation(s)
- Yubin Zhang
- Wadsworth Center, New York State Department of Health and The State University of New York at Albany School of Public Health, Albany, New York 12201, USA
| | | | | |
Collapse
|
46
|
Celecoxib as adjunctive treatment to risperidone in children with autistic disorder: a randomized, double-blind, placebo-controlled trial. Psychopharmacology (Berl) 2013; 225:51-9. [PMID: 22782459 DOI: 10.1007/s00213-012-2796-8] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 06/23/2012] [Indexed: 10/28/2022]
Abstract
RATIONAL Autism is associated with activation of the inflammatory response system. OBJECTIVE This study aims to assess the efficacy of a cyclooxygenase-2 inhibitor, celecoxib, as adjunctive therapy in the treatment of autism METHODS In a 10-week randomized double-blind placebo-controlled study, 40 outpatient children with a Diagnostic and Statistical Manual of Mental Disorders, fourth edition, text revision clinical diagnosis of autism were randomly allocated to celecoxib plus risperidone or placebo plus risperidone. The dose of risperidone and celecoxib were titrated up to 3 and 300 mg/day, respectively. Patients were assessed at baseline and after 2, 4, 6, and 10 weeks of starting medication using the Aberrant Behavior Checklist-Community (ABC-C) Rating Scale. Primary outcome measure was the change in irritability subscale of ABC-C. RESULTS Significant time × treatment interaction was observed for Irritability (F (1.658, 63.021) = 13.580, P < 0.001), Lethargy/Social Withdrawal (F (1.948, 74.032) = 16.811, P < 0.001), and Stereotypic Behavior (F(1.742, 66.198) = 12.104, P < 0.001), but not for Hyperactivity/Noncompliance (F (2.564, 97.424) = 1.469, P = 0.232), and Inappropriate Speech subscales (F (1.607, 61.075) = 0.173, P = 0.794). By week 10, patients in the celecoxib group showed significantly greater improvement in the Irritability (P < 0.001), Lethargy/Social Withdrawal (P < 0.001), and Stereotypic Behavior (P < 0.00) but not in Hyperactivity/Noncompliance (P = 0.202) and Inappropriate Speech (P = 0.802) subscales than the placebo group. Complete response was achieved by four (20 %) patients in the placebo group and 11 (55 %) patients in the celecoxib group (χ (2) (1) = 5.227, P = 0.022). Frequency of side effects was similar between the two groups. CONCLUSIONS Combination of risperidone and celecoxib was superior to risperidone alone in treating irritability, social withdrawal, and stereotypy of children with autism. (Registration, www.irct.ir ; IRCT138711091556N2).
Collapse
|
47
|
Coury DL, Ashwood P, Fasano A, Fuchs G, Geraghty M, Kaul A, Mawe G, Patterson P, Jones NE. Gastrointestinal conditions in children with autism spectrum disorder: developing a research agenda. Pediatrics 2012; 130 Suppl 2:S160-8. [PMID: 23118247 DOI: 10.1542/peds.2012-0900n] [Citation(s) in RCA: 152] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Daniel L Coury
- Developmental/Behavioral Pediatrics, Nationwide Children's Hospital, Columbus, OH 43205-2696, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Mostafa GA, Shehab AA, Al-Ayadhi LY. The link between some alleles on human leukocyte antigen system and autism in children. J Neuroimmunol 2012; 255:70-4. [PMID: 23110937 DOI: 10.1016/j.jneuroim.2012.10.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Revised: 09/29/2012] [Accepted: 10/03/2012] [Indexed: 01/17/2023]
Abstract
The reason behind the initiation of autoimmunity to brain in some patients with autism is not well understood. There is an association between some autoimmune disorders and specific alleles of human leukocyte antigen (HLA) system. Thus, we examined the frequency of some HLA-DRB1 alleles in 100 autistic children and 100 healthy matched-children by differential hybridization with sequence-specific oligonucleotide probes. The risk of association between acquisition or absence of these alleles and autism and also a history of autoimmune diseases in autistic relatives was studied. Autistic children had significantly higher frequency of HLA-DRB1*11 allele than controls (P<0.001). In contrast, autistic children had significantly lower frequency of HLA-DRB1*03 allele than controls (P<0.001). Acquisition of HLA-DRB1*011 and absence of HLA-DRB1*3 had significant risk for association with autism (odds ratio: 3.21 and 0.17, respectively; 95% CI: 1.65-6.31 and 0.06-0.45, respectively). HLA-DRB1*11 had a significant risk for association with a family history of autoimmunity in autistic children (odds ratio: 5.67; 95% CI: 2.07-16.3). In conclusions, the link of some HLA alleles to autism and to family history of autoimmunity indicates the possible contributing role of these alleles to autoimmunity in some autistic children. Despite a relatively small sample size, we are the first to report a probable protective association of HLA-DRB1*03 allele with autism. It warrants a replication study of a larger sample to validate the HLA-DRB1 genetic association with autism. This is important to determine whether therapeutic modulations of the immune function are legitimate avenues for novel therapy in selected cases of autism.
Collapse
Affiliation(s)
- Gehan A Mostafa
- Department of Pediatrics, Faculty of Medicine, Ain Shams University, Cairo, Egypt; Autism Research and Treatment Center, Al-Amodi Autism Research Chair, Department of Physiology, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia.
| | | | | |
Collapse
|
49
|
Braunschweig D, Duncanson P, Boyce R, Hansen R, Ashwood P, Pessah IN, Hertz-Picciotto I, Van de Water J. Behavioral correlates of maternal antibody status among children with autism. J Autism Dev Disord 2012; 42:1435-45. [PMID: 22012245 DOI: 10.1007/s10803-011-1378-7] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Autism spectrum disorders (ASDs) affect approximately 1 in 110 children in the United States. This report profiles fetal-brain reactive autoantibodies of a large cohort of mothers of children with autism and controls, yielding significant associations between the presence of IgG reactivity to fetal brain proteins at 37 and 73 kDa and a childhood diagnosis of full autism (p = 0.0005), which also correlated with lower expressive language scores (p = 0.005). Additionally, we report on reactivity to proteins at 39 and 73 kDa, which correlated with the broader diagnosis of ASD (p = 0.0007) and increased irritability on the Aberrant Behavioral Checklist (p = 0.05). This study provides evidence of multiple patterns of reactivity to fetal brain proteins by maternal antibodies associated with ASD and specific childhood behavioral outcomes.
Collapse
Affiliation(s)
- Daniel Braunschweig
- Division of Rheumatology/Allergy and Clinical Immunology, University of California, Davis, 451 E Health Sciences Drive, Suite 6510 GBSF, Davis, CA 95616, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Torres AR, Westover JB, Gibbons C, Johnson RC, Ward DC. Activating killer-cell immunoglobulin-like receptors (KIR) and their cognate HLA ligands are significantly increased in autism. Brain Behav Immun 2012; 26:1122-7. [PMID: 22884899 PMCID: PMC3469320 DOI: 10.1016/j.bbi.2012.07.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Revised: 07/16/2012] [Accepted: 07/19/2012] [Indexed: 01/18/2023] Open
Abstract
Killer-cell immunoglobulin-like receptor (KIR) proteins are expressed on natural killer (NK) cells and appear important in innate and adaptive immunity. There are about 14 KIR genes on chromosome 19q13.4, composed of those that inhibit and those that activate NK cell killing. Haplotypes have different combinations of these genes meaning that not all genes are present in a subject. There are two main classes of cognate human leukocyte antigen (HLA) ligands (HLA-Bw4 and HLA-C1/C2) that bind to the inhibitory/activating receptors. As a general rule, the inhibitory state is maintained except when virally infected or tumor cells are encountered; however, both increased activation and inhibition states have been associated with susceptibility and protection against numerous disease states including cancer, arthritis, and psoriasis. Utilizing DNA from 158 Caucasian subjects with autism and 176 KIR control subjects we show for the first time a highly significant increase in four activating KIR genes (2DS5, 3DS1, 2DS1 and 2DS4) as measured by chi square values and odds ratios. In addition, our data suggests a highly significant increase in the activating KIR gene 2DS1 and its cognate HLA-C2 ligand (2DS1+C2; p = 0.00003 [Odds ratio = 2.87]). This information ties together two major immune gene complexes, the human leukocyte complex and the leukocyte receptor complex, and may partially explain immune abnormalities observed in many subjects with autism.
Collapse
Affiliation(s)
- Anthony R Torres
- Center for Persons with Disabilities, Utah State University, Logan, UT 84322-6804, USA.
| | | | | | | | | |
Collapse
|