1
|
Su J, Gupta R, Van Hoof S, Kreye J, Prüss H, Spielman B, Brimberg L, Volpe BT, Huerta PT, Diamond B. Heterogeneity of anti-Caspr2 antibodies: specificity and pathogenicity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.16.633238. [PMID: 39896527 PMCID: PMC11785012 DOI: 10.1101/2025.01.16.633238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Maternal anti-Caspr2 (Contactin-associated protein-like 2) antibodies have been associated with increased risk for autism spectrum disorder (ASD). Previous studies have shown that in utero exposure to anti-Caspr2 antibodies results in a phenotype with ASD-like features in male mice. Here we ask whether four newly generated antibodies against Caspr2 are pathogenic to the developing fetal brain and whether they function through similar means. Our results show that the novel anti-Caspr2 antibodies recognize different epitopes of Caspr2. In utero exposure to these antibodies elicits differential ASD-like phenotypes in male offspring, tested in the social interaction, open field, and light-dark tasks. These results demonstrate variability in the antigenic specificity and pathogenicity of anti-Caspr2 antibodies which may have clinical implications.
Collapse
|
2
|
Mamun AA, Geng P, Wang S, Shao C, Xiao J. IUPHAR review: Targeted therapies of signaling pathways based on the gut microbiome in autism spectrum disorders: Mechanistic and therapeutic applications. Pharmacol Res 2025; 211:107559. [PMID: 39733842 DOI: 10.1016/j.phrs.2024.107559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 12/22/2024] [Accepted: 12/23/2024] [Indexed: 12/31/2024]
Abstract
Autism spectrum disorders (ASD) are complex neurodevelopmental disorders characterized by impairments in social interaction, communication and repetitive activities. Gut microbiota significantly influences behavior and neurodevelopment by regulating the gut-brain axis. This review explores gut microbiota-influenced treatments for ASD, focusing on their therapeutic applications and mechanistic insights. In addition, this review discusses the interactions between gut microbiota and the immune, metabolic and neuroendocrine systems, focusing on crucial microbial metabolites including short-chain fatty acids (SCFAs) and several neurotransmitters. Furthermore, the review explores various therapy methods including fecal microbiota transplantation, dietary modifications, probiotics and prebiotics and evaluates their safety and efficacy in reducing ASD symptoms. The discussion shows the potential of customized microbiome-based therapeutics and the integration of multi-omics methods to understand the underlying mechanisms. Moreover, the review explores the intricate relationship between gut microbiota and ASD, aiming to develop innovative therapies that utilize the gut microbiome to improve the clinical outcomes of ASD patients. Microbial metabolites such as neurotransmitter precursors, tryptophan metabolites and SCFAs affect brain development and behavior. Symptoms of ASD are linked to changes in these metabolites. Dysbiosis in the gut microbiome may impact neuroinflammatory processes linked to autism, negatively affecting immune signaling pathways. Research indicates that probiotics and prebiotics can improve gut microbiota and alleviate symptoms in ASD patients. Fecal microbiota transplantation may also improve behavioral symptoms and restore gut microbiota balance. The review emphasizes the need for further research on gut microbiota modification as a potential therapeutic approach for ASD, highlighting its potential in clinical settings.
Collapse
Affiliation(s)
- Abdullah Al Mamun
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People's Hospital, Lishui, Zhejiang 323000, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Peiwu Geng
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People's Hospital, Lishui, Zhejiang 323000, China
| | - Shuanghu Wang
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People's Hospital, Lishui, Zhejiang 323000, China
| | - Chuxiao Shao
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People's Hospital, Lishui, Zhejiang 323000, China.
| | - Jian Xiao
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People's Hospital, Lishui, Zhejiang 323000, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Department of Wound Healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| |
Collapse
|
3
|
Mohebalizadeh M, Babapour G, Maleki Aghdam M, Mohammadi T, Jafari R, Shafiei-Irannejad V. Role of Maternal Immune Factors in Neuroimmunology of Brain Development. Mol Neurobiol 2024; 61:9993-10005. [PMID: 38057641 DOI: 10.1007/s12035-023-03749-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 10/27/2023] [Indexed: 12/08/2023]
Abstract
Inflammation during pregnancy may occur due to various factors. This condition, in which maternal immune system activation occurs, can affect fetal brain development and be related to neurodevelopmental diseases. MIA interacts with the fetus's brain development through maternal antibodies, cytokines, chemokines, and microglial cells. Antibodies are associated with the development of the nervous system by two mechanisms: direct binding to brain inflammatory factors and binding to brain antigens. Cytokines and chemokines have an active presence in inflammatory processes. Additionally, glial cells, defenders of the nervous system, play an essential role in synaptic modulation and neurogenesis. Maternal infections during pregnancy are the most critical factors related to MIA; however, several studies show the relation between these infections and neurodevelopmental diseases. Infection with specific viruses, such as Zika, cytomegalovirus, influenza A, and SARS-CoV-2, has revealed effects on neurodevelopment and the onset of diseases such as schizophrenia and autism. We review the relationship between maternal infections during pregnancy and their impact on neurodevelopmental processes.
Collapse
Affiliation(s)
- Mehdi Mohebalizadeh
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Urmia, Iran
- Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Urmia, Iran
| | - Golsa Babapour
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Mahdi Maleki Aghdam
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Urmia, Iran
| | - Tooba Mohammadi
- Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Urmia, Iran
| | - Reza Jafari
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Vahid Shafiei-Irannejad
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
4
|
Chen S, Wang J, Xiaofang Chen, Zhang Y, Hong Y, Zhuang W, Huang X, Kang J, Ou P, Huang L. Chinese acupuncture: A potential treatment for autism rat model via improving synaptic function. Heliyon 2024; 10:e37130. [PMID: 39286195 PMCID: PMC11402746 DOI: 10.1016/j.heliyon.2024.e37130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 08/22/2024] [Accepted: 08/28/2024] [Indexed: 09/19/2024] Open
Abstract
Purpose Autistic symptom improvement can be observed in children treated with acupuncture, but the mechanism is still being explored. In the present study, we used scalp acupuncture to treat autism rat model, and then their improvement in the abnormal behaviors and specific mechanisms behind were revealed by detecting animal behaviors, analyzing the RNA sequencing of the prefrontal cortex (PFC), and observing the ultrastructure of PFC neurons under the transmission electron microscope. Methods On gestational day 12.5, Wistar rats were given valproic acid (VPA) by intraperitoneal injection, and their offspring were considered to be reliable rat models of autism. They were randomized to VPA or VPA-acupuncture group (n = 8). Offspring of Wistar pregnant rats that were simultaneously injected with saline were randomly selected as the wild-type group (WT). VPA_acupuncture group rats received acupuncture intervention at 23 days of age for 4 weeks, and the other two groups followed without intervention. After the intervention, all experimental rats underwent behavioral tests. Immediately afterward, they were euthanized by cervical dislocation, and their prefrontal cortex was isolated for RNA sequencing and transmission electron microscopy. Results The main results are as follows: 1. Animal behavioural tests: VPA group rats showed more anxiety-like behaviour and repetitive, stereotyped behaviour than WT group rats. While VPA group rats showed less spatial exploration ability, activity level, social interaction, and social novelty preference than WT group rats. It was gratifying to observe that acupuncture indeed improved these abnormal behaviors of autism rat model. 2. RNA-sequencing: The three groups of rats differed in the expression and enrichment pathways of multiple genes related to synaptic function, neural signal transduction, immune-inflammatory responses and circadian rhythm regulation. Our experiments indicated that acupuncture can alleviate the major symptoms of ASD by improving these neurological abnormalities. 3. Under the transmission electron microscopy, several lysosomes and mitochondrial structural abnormalities were observed in the prefrontal neurons of VPA group rats, which were manifested as atrophy of the mitochondrial membrane, blurring or disappearance of the mitochondrial cristae, and even vacuolization. Moreover, the number of synapses and synaptic vesicles was relatively small. Conversely, the mitochondrial structure of rats in the WT group and VPA_acupuncture was normal, and the number of synapses and synaptic vesicles was relatively large. Conclusion Acupuncture effectively improved the abnormal behaviors of autism rat model and the ultrastructure of the PFC neurons, which might worked by improving their abnormal synaptic function, synaptic plasticity promoting neuronal signal transduction and regulating immune-inflammatory responses.
Collapse
Affiliation(s)
- Sijie Chen
- Fujian Maternity and Child Health Hospital, Fuzhou, 350108, Fujian, China
| | - Juan Wang
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350108, Fujian, China
| | - Xiaofang Chen
- Jinjiang Maternal and Child Health Hospital, Quanzhou, 362299, Fujian, China
| | - Yingying Zhang
- Fujian Maternity and Child Health Hospital, Fuzhou, 350108, Fujian, China
| | - Yu Hong
- Fujian Maternity and Child Health Hospital, Fuzhou, 350108, Fujian, China
| | - Wanyu Zhuang
- Fujian Maternity and Child Health Hospital, Fuzhou, 350108, Fujian, China
| | - Xinxin Huang
- Fujian Maternity and Child Health Hospital, Fuzhou, 350108, Fujian, China
| | - Jie Kang
- Department of TCM Syndrome Research Base, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Ping Ou
- Fujian Maternity and Child Health Hospital, Fuzhou, 350108, Fujian, China
| | - Longsheng Huang
- Fujian Maternity and Child Health Hospital, Fuzhou, 350108, Fujian, China
| |
Collapse
|
5
|
Zhang P, Wang X, Xu Y, Zhao X, Zhang X, Zhao Z, Wang H, Xiong Z. Association between interpregnancy interval and risk of autism spectrum disorder: a systematic review and Bayesian network meta-analysis. Eur J Pediatr 2024; 183:1209-1221. [PMID: 38085281 DOI: 10.1007/s00431-023-05364-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 11/29/2023] [Accepted: 12/01/2023] [Indexed: 03/20/2024]
Abstract
Although the risk of autism spectrum disorder (ASD) has been reported to be associated with interpregnancy intervals (IPIs), their association remains debatable due to inconsistent findings in existing studies. Therefore, the present study aimed to explore their association. PubMed, Embase, Web of Science, and the Cochrane Library were systematically retrieved up to May 25, 2022. An updated search was performed on May 25, 2023, to encompass recent studies. The quality of the included studies was assessed using the Newcastle-Ottawa Scale (NOS). Our primary outcome measures were expressed as adjusted odds ratios (ORs). Given various control measures for IPI and diverse IPI thresholds in the included studies, a Bayesian network meta-analysis was performed. Eight studies were included, involving 24,865 children with ASD and 2,890,289 children without ASD. Compared to an IPI of 24 to 35 months, various IPIs were significantly associated with a higher risk of ASD (IPIs < 6 months: OR = 1.63, 95% CI 1.53-1.74, n = 5; IPIs of 6-11 months: OR = 1.50, 95% CI 1.42-1.59, n = 4; IPIs of 12-23 months: OR = 1.19, 95% CI 1.12-1.23, n = 10; IPIs of 36-59 months: OR = 0.96, 95% CI 0.94-0.99, n = 2; IPIs of 60-119 months: OR = 1.15, 95% CI 1.10-1.20, n = 4; IPIs > 120 months: OR = 1.57, 95% CI 1.43-1.72, n = 4). After adjusting confounding variables, our analysis delineated a U-shaped restricted cubic spline curve, underscoring that both substantially short (< 24 months) and excessively long IPIs (> 72 months) are significantly correlated with an increased risk of ASD. Conclusion: Our analysis indicates that both shorter and longer IPIs might predispose children to a higher risk of ASD. Optimal childbearing health and neurodevelopmental outcomes appear to be associated with a moderate IPI, specifically between 36 and 60 months. What is Known: • An association between autism spectrum disorder (ASD) and interpregnancy intervals (IPIs) has been speculated in some reports. • This association remains debatable due to inconsistent findings in available studies. What is New: • Our study delineated a U-shaped restricted cubic spline curve, suggesting that both shorter and longer IPIs predispose children to a higher risk of ASD. • Optimal childbearing health and neurodevelopmental outcomes appear to be associated with a moderate IPI, specifically between 36 and 60 months.
Collapse
Affiliation(s)
- Ping Zhang
- Department of Child Health Care, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430070, China
| | - Xiaoyan Wang
- Department of Child Health Care, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430070, China
| | - Yufen Xu
- Department of Child Health Care, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430070, China
| | - Xiaoming Zhao
- Department of Child Health Care, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430070, China
| | - Xuan Zhang
- Department of Child Health Care, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430070, China
| | - Zhiwei Zhao
- Department of Child Health Care, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430070, China
| | - Hong Wang
- Department of Child Health Care, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430070, China.
| | - Zhonggui Xiong
- Department of Child Health Care, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430070, China.
| |
Collapse
|
6
|
Chen CC, Lin CH, Lin MC. Maternal autoimmune disease and risk of offspring autism spectrum disorder - a nationwide population-based cohort study. Front Psychiatry 2023; 14:1254453. [PMID: 38025447 PMCID: PMC10654781 DOI: 10.3389/fpsyt.2023.1254453] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Autism spectrum disorder (ASD) is a group of neurodevelopmental disorders which cause long term social and behavior impairment, and its prevalence is on the rise. Studies about the association between maternal autoimmune diseases and offspring ASD have controversial results. The aim of this study was to investigate whether maternal autoimmune diseases increase the risk of ASD in offspring from a population-based perspective. Methods The data sources were Taiwan's National Health Insurance Research Database (NHIRD) and Taiwan's Maternal and Child Health Database (MCHD), which were integrated and used to identify newborns whose mothers were diagnosed with autoimmune disease. Newborns were matched by maternal age, neonatal gender, and date of birth with controls whose mothers were without autoimmune disease using a ratio of 1:4 between 2004 and 2019. Data on diagnoses of autoimmune disease and autism spectrum disorders were retrieved from NHIRD. Patients who had at least 3 outpatient visits or at least 1 admission with a diagnosis of autoimmune disease and autism spectrum disorders were defined as incidence cases. The risks of ASD in offspring were compared between mothers with or without autoimmune disorders. Results We identified 20,865 newborns whose mothers had been diagnosed with autoimmune disease before pregnancy and matched them at a ratio of 1:4 with a total of 83,460 newborn whose mothers were without autoimmune disease, by maternal age, neonatal gender, and date of birth. They were randomly selected as the control group. The cumulative incidence rates of autism spectrum disorders (ASD) were significantly higher among the offspring of mothers with autoimmune diseases. After adjusting for cofactors, the risk of ASD remained significantly higher in children whose mother had autoimmune diseases. Regarding to specific maternal autoimmune disease, Sjögren's syndrome and rheumatoid arthritis were both associated with elevated risks of ASD in offspring. Conclusion Mother with autoimmune disease might be associated with increasing the risk of autism spectrum disorder in offspring.
Collapse
Affiliation(s)
- Ching-Chu Chen
- Children’s Medical Center, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Ching-Heng Lin
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Ming-Chih Lin
- Children’s Medical Center, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
- Department of Food and Nutrition, Providence University, Taichung, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| |
Collapse
|
7
|
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: 4.0] [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
|
8
|
Bagnall-Moreau C, Spielman B, Brimberg L. Maternal brain reactive antibodies profile in autism spectrum disorder: an update. Transl Psychiatry 2023; 13:37. [PMID: 36737600 PMCID: PMC9898547 DOI: 10.1038/s41398-023-02335-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 01/20/2023] [Accepted: 01/24/2023] [Indexed: 02/05/2023] Open
Abstract
Autism spectrum disorder (ASD) is a heterogeneous neurodevelopmental disorder with multifactorial etiologies involving both genetic and environmental factors. In the past two decades it has become clear that in utero exposure to toxins, inflammation, microbiome, and antibodies (Abs), may play a role in the etiology of ASD. Maternal brain-reactive Abs, present in 10-20% of mothers of a child with ASD, pose a potential risk to the developing brain because they can gain access to the brain during gestation, altering brain development during a critical period. Different maternal anti-brain Abs have been associated with ASD and have been suggested to bind extracellular or intracellular neuronal antigens. Clinical data from various cohorts support the increase in prevalence of such maternal brain-reactive Abs in mothers of a child with ASD compared to mothers of a typically developing child. Animal models of both non-human primates and rodents have provided compelling evidence supporting a pathogenic role of these Abs. In this review we summarize the data from clinical and animal models addressing the role of pathogenic maternal Abs in ASD. We propose that maternal brain-reactive Abs are an overlooked and promising field of research, representing a modifiable risk factor that may account for up to 20% of cases of ASD. More studies are needed to better characterize the Abs that contribute to the risk of having a child with ASD, to understand whether we can we predict such cases of ASD, and to better pinpoint the antigenic specificity of these Abs and their mechanisms of pathogenicity.
Collapse
Affiliation(s)
- Ciara Bagnall-Moreau
- grid.250903.d0000 0000 9566 0634Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, New York, NY USA
| | - Benjamin Spielman
- grid.250903.d0000 0000 9566 0634Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, New York, NY USA ,grid.512756.20000 0004 0370 4759Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY USA
| | - Lior Brimberg
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, New York, NY, USA. .,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA.
| |
Collapse
|
9
|
Mazón-Cabrera R, Liesenborgs J, Brône B, Vandormael P, Somers V. Novel maternal autoantibodies in autism spectrum disorder: Implications for screening and diagnosis. Front Neurosci 2023; 17:1067833. [PMID: 36816132 PMCID: PMC9932693 DOI: 10.3389/fnins.2023.1067833] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 01/09/2023] [Indexed: 02/05/2023] Open
Abstract
Introduction Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder for which early recognition is a major challenge. Autoantibodies against fetal brain antigens have been found in the blood of mothers of children with ASD (m-ASD) and can be transferred to the fetus where they can impact neurodevelopment by binding to fetal brain proteins. This study aims to identify novel maternal autoantibodies reactive against human fetal brain antigens, and explore their use as biomarkers for ASD screening and diagnosis. Methods A custom-made human fetal brain cDNA phage display library was constructed, and screened for antibody reactivity in m-ASD samples from the Simons Simplex Collection (SSC) of the Simons Foundation Autism Research Initiative (SFARI). Antibody reactivity against 6 identified antigens was determined in plasma samples of 238 m-ASD and 90 mothers with typically developing children (m-TD). Results We identified antibodies to 6 novel University Hasselt (UH)-ASD antigens, including three novel m-ASD autoantigens, i.e., ribosomal protein L23 (RPL23), glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and calmodulin-regulated spectrin-associated protein 3 (CAMSAP3). Antibody reactivity against a panel of four of these targets was found in 16% of m-ASD samples, compared to 4% in m-TD samples (p = 0.0049). Discussion Maternal antibodies against 4 UH-ASD antigens could therefore provide a novel tool to support the diagnosis of ASD in a subset of individuals.
Collapse
Affiliation(s)
- Rut Mazón-Cabrera
- Department of Immunology and Infection, Biomedical Research Institute, UHasselt, Transnational University Limburg, Diepenbeek, Belgium
| | - Jori Liesenborgs
- Expertise Centre for Digital Media, UHasselt, Transnational University Limburg, Diepenbeek, Belgium
| | - Bert Brône
- Department of Neurosciences, Biomedical Research Institute, UHasselt, Transnational University Limburg, Diepenbeek, Belgium
| | - Patrick Vandormael
- Department of Immunology and Infection, Biomedical Research Institute, UHasselt, Transnational University Limburg, Diepenbeek, Belgium
| | - Veerle Somers
- Department of Immunology and Infection, Biomedical Research Institute, UHasselt, Transnational University Limburg, Diepenbeek, Belgium,*Correspondence: Veerle Somers,
| |
Collapse
|
10
|
Pregestational Exposure to T. gondii Produces Maternal Antibodies That Recognize Fetal Brain Mimotopes and Induces Neurochemical and Behavioral Dysfunction in the Offspring. Cells 2022; 11:cells11233819. [PMID: 36497079 PMCID: PMC9741080 DOI: 10.3390/cells11233819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/23/2022] [Accepted: 11/25/2022] [Indexed: 11/30/2022] Open
Abstract
The activation of the maternal immune system by a prenatal infection is considered a risk factor for developing psychiatric disorders in the offspring. Toxoplasma gondii is one of the pathogenic infections associated with schizophrenia. Recent studies have shown an association between high levels of IgG anti-T. gondii from mothers and their neonates, with a higher risk of developing schizophrenia. The absence of the parasite and the levels of IgGs found in the early stages of life suggest a transplacental transfer of the anti-T. gondii IgG antibodies, which could bind fetal brain structures by molecular mimicry and induce alterations in neurodevelopment. This study aimed to determine the maternal pathogenic antibodies formation that led to behavioral impairment on the progeny of rats immunized with T. gondii. Female rats were immunized prior to gestation with T. gondii lysate (3 times/once per week). The anti-T. gondii IgG levels were determined in the serum of pregestational exposed females' previous mating. After this, locomotor activity, cognitive and social tests were performed. Cortical neurotransmitter levels for dopamine and glutamate were evaluated at 60 PND in the progeny of rats immunized before gestation (Pregestational group). The maternal pathogenic antibodies were evidenced by their binding to fetal brain mimotopes in the Pregestational group and the reactivity of the serum containing anti-T. gondii IgG was tested in control fetal brains (non-immunized). These results showed that the Pregestational group presented impairment in short and long-term memory, hypoactivity and alteration in social behavior, which was also associated with a decrease in cortical glutamate and dopamine levels. We also found the IgG antibodies bound to brain mimotopes in fetuses from females immunized with T. gondii, as well as observing a strong reactivity of the serum females immunized for fetal brain structures of fetuses from unimmunized mothers. Our results suggest that the exposure to T. gondii before gestation produced maternal pathogenic antibodies that can recognize fetal brain mimotopes and lead to neurochemical and behavioral alterations in the offspring.
Collapse
|
11
|
Al Dera H. Cellular and molecular mechanisms underlying autism spectrum disorders and associated comorbidities: A pathophysiological review. Biomed Pharmacother 2022; 148:112688. [PMID: 35149383 DOI: 10.1016/j.biopha.2022.112688] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/25/2022] [Accepted: 02/01/2022] [Indexed: 12/31/2022] Open
Abstract
Autism spectrum disorders (ASD) are a group of neurodevelopmental disorders that develop in early life due to interaction between several genetic and environmental factors and lead to alterations in brain function and structure. During the last decades, several mechanisms have been placed to explain the pathogenesis of autism. Unfortunately, these are reported in several studies and reviews which make it difficult to follow by the reader. In addition, some recent molecular mechanisms related to ASD have been unrevealed. This paper revises and highlights the major common molecular mechanisms responsible for the clinical symptoms seen in people with ASD, including the roles of common genetic factors and disorders, neuroinflammation, GABAergic signaling, and alterations in Ca+2 signaling. Besides, it covers the major molecular mechanisms and signaling pathways involved in initiating the epileptic seizure, including the alterations in the GABAergic and glutamate signaling, vitamin and mineral deficiency, disorders of metabolism, and autoimmunity. Finally, this review also discusses sleep disorder patterns and the molecular mechanisms underlying them.
Collapse
Affiliation(s)
- Hussain Al Dera
- Department of Basic Medical Sciences, College of Medicine at King Saud, Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Saudi Arabia; King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia.
| |
Collapse
|
12
|
McLellan J, Kim DHJ, Bruce M, Ramirez-Celis A, Van de Water J. Maternal Immune Dysregulation and Autism-Understanding the Role of Cytokines, Chemokines and Autoantibodies. Front Psychiatry 2022; 13:834910. [PMID: 35722542 PMCID: PMC9201050 DOI: 10.3389/fpsyt.2022.834910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 04/20/2022] [Indexed: 11/29/2022] Open
Abstract
Autism spectrum disorder (ASD) is acknowledged as a highly heterogeneous, behaviorally defined neurodevelopmental disorder with multiple etiologies. In addition to its high heritability, we have come to recognize a role for maternal immune system dysregulation as a prominent risk factor for the development of ASD in the child. Examples of these risk factors include altered cytokine/chemokine activity and the presence of autoantibodies in mothers that are reactive to proteins in the developing brain. In addition to large clinical studies, the development of pre-clinical models enables the ability to evaluate the cellular and molecular underpinnings of immune-related pathology. For example, the novel animal models of maternal autoantibody-related (MAR) ASD described herein will serve as a preclinical platform for the future testing of targeted therapeutics for one 'type' of ASD. Identification of the cellular targets will advance precision medicine efforts toward tailored therapeutics and prevention. This minireview highlights emerging evidence for the role of maternal immune dysregulation as a potential biomarker, as well as a pathologically relevant mechanism for the development of ASD in offspring. Further, we will discuss the current limitations of these models as well as potential avenues for future research.
Collapse
Affiliation(s)
- Janna McLellan
- Division of Rheumatology, Department of Internal Medicine, Allergy, and Clinical Immunology, University of California, Davis, Davis, CA, United States
| | - Danielle H J Kim
- Division of Rheumatology, Department of Internal Medicine, Allergy, and Clinical Immunology, University of California, Davis, Davis, CA, United States
| | - Matthew Bruce
- Division of Rheumatology, Department of Internal Medicine, Allergy, and Clinical Immunology, University of California, Davis, Davis, CA, United States
| | - Alexandra Ramirez-Celis
- Division of Rheumatology, Department of Internal Medicine, Allergy, and Clinical Immunology, University of California, Davis, Davis, CA, United States
| | - Judy Van de Water
- Division of Rheumatology, Department of Internal Medicine, Allergy, and Clinical Immunology, University of California, Davis, Davis, CA, United States.,MIND Institute, University of California, Davis, Davis, CA, United States
| |
Collapse
|
13
|
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: 9.3] [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
|
14
|
Panisi C, Guerini FR, Abruzzo PM, Balzola F, Biava PM, Bolotta A, Brunero M, Burgio E, Chiara A, Clerici M, Croce L, Ferreri C, Giovannini N, Ghezzo A, Grossi E, Keller R, Manzotti A, Marini M, Migliore L, Moderato L, Moscone D, Mussap M, Parmeggiani A, Pasin V, Perotti M, Piras C, Saresella M, Stoccoro A, Toso T, Vacca RA, Vagni D, Vendemmia S, Villa L, Politi P, Fanos V. Autism Spectrum Disorder from the Womb to Adulthood: Suggestions for a Paradigm Shift. J Pers Med 2021; 11:70. [PMID: 33504019 PMCID: PMC7912683 DOI: 10.3390/jpm11020070] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 01/10/2021] [Accepted: 01/19/2021] [Indexed: 02/07/2023] Open
Abstract
The wide spectrum of unique needs and strengths of Autism Spectrum Disorders (ASD) is a challenge for the worldwide healthcare system. With the plethora of information from research, a common thread is required to conceptualize an exhaustive pathogenetic paradigm. The epidemiological and clinical findings in ASD cannot be explained by the traditional linear genetic model, hence the need to move towards a more fluid conception, integrating genetics, environment, and epigenetics as a whole. The embryo-fetal period and the first two years of life (the so-called 'First 1000 Days') are the crucial time window for neurodevelopment. In particular, the interplay and the vicious loop between immune activation, gut dysbiosis, and mitochondrial impairment/oxidative stress significantly affects neurodevelopment during pregnancy and undermines the health of ASD people throughout life. Consequently, the most effective intervention in ASD is expected by primary prevention aimed at pregnancy and at early control of the main effector molecular pathways. We will reason here on a comprehensive and exhaustive pathogenetic paradigm in ASD, viewed not just as a theoretical issue, but as a tool to provide suggestions for effective preventive strategies and personalized, dynamic (from womb to adulthood), systemic, and interdisciplinary healthcare approach.
Collapse
Affiliation(s)
- Cristina Panisi
- Fondazione Istituto Sacra Famiglia ONLUS, Cesano Boscone, 20090 Milan, Italy;
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy;
| | - Franca Rosa Guerini
- IRCCS Fondazione Don Carlo Gnocchi, ONLUS, 20148 Milan, Italy; (M.C.); (M.S.)
| | | | - Federico Balzola
- Division of Gastroenterology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, University of Turin, 10126 Turin, Italy;
| | - Pier Mario Biava
- Scientific Institute of Research and Care Multimedica, 20138 Milan, Italy;
| | - Alessandra Bolotta
- DIMES, School of Medicine, University of Bologna, 40126 Bologna, Italy; (P.M.A.); (A.B.); (A.G.)
| | - Marco Brunero
- Department of Pediatric Surgery, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Ernesto Burgio
- ECERI—European Cancer and Environment Research Institute, Square de Meeus 38-40, 1000 Bruxelles, Belgium;
| | - Alberto Chiara
- Dipartimento Materno Infantile ASST, 27100 Pavia, Italy;
| | - Mario Clerici
- IRCCS Fondazione Don Carlo Gnocchi, ONLUS, 20148 Milan, Italy; (M.C.); (M.S.)
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy
| | - Luigi Croce
- Centro Domino per l’Autismo, Universita’ Cattolica Brescia, 20139 Milan, Italy;
| | - Carla Ferreri
- National Research Council of Italy, Institute of Organic Synthesis and Photoreactivity (ISOF), 40129 Bologna, Italy;
| | - Niccolò Giovannini
- Department of Obstetrics and Gynecology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Alessandro Ghezzo
- DIMES, School of Medicine, University of Bologna, 40126 Bologna, Italy; (P.M.A.); (A.B.); (A.G.)
| | - Enzo Grossi
- Autism Research Unit, Villa Santa Maria Foundation, 22038 Tavernerio, Italy;
| | - Roberto Keller
- Adult Autism Centre DSM ASL Città di Torino, 10138 Turin, Italy;
| | - Andrea Manzotti
- RAISE Lab, Foundation COME Collaboration, 65121 Pescara, Italy;
| | - Marina Marini
- DIMES, School of Medicine, University of Bologna, 40126 Bologna, Italy; (P.M.A.); (A.B.); (A.G.)
| | - Lucia Migliore
- Medical Genetics Laboratories, Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy; (L.M.); (A.S.)
| | - Lucio Moderato
- Fondazione Istituto Sacra Famiglia ONLUS, Cesano Boscone, 20090 Milan, Italy;
| | - Davide Moscone
- Associazione Spazio Asperger ONLUS, Centro Clinico CuoreMenteLab, 00141 Rome, Italy;
| | - Michele Mussap
- Neonatal Intensive Care Unit, Department of Surgical Sciences, Puericulture Institute and Neonatal Section, Azienda Ospedaliera Universitaria, 09100 Cagliari, Italy; (M.M.); (V.F.)
| | - Antonia Parmeggiani
- Child Neurology and Psychiatry Unit, IRCCS ISNB, S. Orsola-Malpighi Hospital, Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy;
| | - Valentina Pasin
- Milan Institute for health Care and Advanced Learning, 20124 Milano, Italy;
| | | | - Cristina Piras
- Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy;
| | - Marina Saresella
- IRCCS Fondazione Don Carlo Gnocchi, ONLUS, 20148 Milan, Italy; (M.C.); (M.S.)
| | - Andrea Stoccoro
- Medical Genetics Laboratories, Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy; (L.M.); (A.S.)
| | - Tiziana Toso
- Unione Italiana Lotta alla Distrofia Muscolare UILDM, 35100 Padova, Italy;
| | - Rosa Anna Vacca
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council of Italy, 70126 Bari, Italy;
| | - David Vagni
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy, 98164 Messina, Italy;
| | | | - Laura Villa
- Scientific Institute, IRCCS Eugenio Medea, Via Don Luigi Monza 20, 23842 Bosisio Parini, Italy;
| | - Pierluigi Politi
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy;
| | - Vassilios Fanos
- Neonatal Intensive Care Unit, Department of Surgical Sciences, Puericulture Institute and Neonatal Section, Azienda Ospedaliera Universitaria, 09100 Cagliari, Italy; (M.M.); (V.F.)
- Neonatal Intensive Care Unit, Azienda Ospedaliera Universitaria, 09042 Cagliari, Italy
| |
Collapse
|
15
|
Mesleh AG, Abdulla SA, El-Agnaf O. Paving the Way toward Personalized Medicine: Current Advances and Challenges in Multi-OMICS Approach in Autism Spectrum Disorder for Biomarkers Discovery and Patient Stratification. J Pers Med 2021; 11:jpm11010041. [PMID: 33450950 PMCID: PMC7828397 DOI: 10.3390/jpm11010041] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/07/2021] [Accepted: 01/08/2021] [Indexed: 02/06/2023] Open
Abstract
Autism spectrum disorder (ASD) is a multifactorial neurodevelopmental disorder characterized by impairments in two main areas: social/communication skills and repetitive behavioral patterns. The prevalence of ASD has increased in the past two decades, however, it is not known whether the evident rise in ASD prevalence is due to changes in diagnostic criteria or an actual increase in ASD cases. Due to the complexity and heterogeneity of ASD, symptoms vary in severity and may be accompanied by comorbidities such as epilepsy, attention deficit hyperactivity disorder (ADHD), and gastrointestinal (GI) disorders. Identifying biomarkers of ASD is not only crucial to understanding the biological characteristics of the disorder, but also as a detection tool for its early screening. Hence, this review gives an insight into the main areas of ASD biomarker research that show promising findings. Finally, it covers success stories that highlight the importance of precision medicine and the current challenges in ASD biomarker discovery studies.
Collapse
Affiliation(s)
- Areej G. Mesleh
- Division of Genomics and Precision Medicine (GPM), College of Health & Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Doha 34110, Qatar;
| | - Sara A. Abdulla
- Neurological Disorder Center, Qatar Biomedical Research Institute (QBRI), HBKU, Doha 34110, Qatar
- Correspondence: (S.A.A.); (O.E.-A.)
| | - Omar El-Agnaf
- Division of Genomics and Precision Medicine (GPM), College of Health & Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Doha 34110, Qatar;
- Neurological Disorder Center, Qatar Biomedical Research Institute (QBRI), HBKU, Doha 34110, Qatar
- Correspondence: (S.A.A.); (O.E.-A.)
| |
Collapse
|
16
|
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: 4.0] [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
|
17
|
Jones KL, Pride MC, Edmiston E, Yang M, Silverman JL, Crawley JN, Van de Water J. Autism-specific maternal autoantibodies produce behavioral abnormalities in an endogenous antigen-driven mouse model of autism. Mol Psychiatry 2020; 25:2994-3009. [PMID: 29955164 PMCID: PMC6310680 DOI: 10.1038/s41380-018-0126-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 05/25/2018] [Accepted: 06/05/2018] [Indexed: 01/05/2023]
Abstract
Immune dysregulation has been noted consistently in individuals with autism spectrum disorder (ASD) and their families, including the presence of autoantibodies reactive to fetal brain proteins in nearly a quarter of mothers of children with ASD versus <1% in mothers of typically developing children. Our lab recently identified the peptide epitope sequences on seven antigenic proteins targeted by these maternal autoantibodies. Through immunization with these peptide epitopes, we have successfully created an endogenous, antigen-driven mouse model that ensures a constant exposure to the salient autoantibodies throughout gestation in C57BL/6J mice. This exposure more naturally mimics what is observed in mothers of children with ASD. Male and female offspring were tested using a comprehensive sequence of behavioral assays, as well as measures of health and development highly relevant to ASD. We found that MAR-ASD male and female offspring had significant alterations in development and social interactions during dyadic play. Although 3-chambered social approach was not significantly different, fewer social interactions with an estrous female were noted in the adult male MAR-ASD animals, as well as reduced vocalizations emitted in response to social cues with robust repetitive self-grooming behaviors relative to saline treated controls. The generation of MAR-ASD-specific epitope autoantibodies in female mice prior to breeding created a model that demonstrates for the first time that ASD-specific antigen-induced maternal autoantibodies produced alterations in a constellation of ASD-relevant behaviors.
Collapse
Affiliation(s)
- Karen L. Jones
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, University of California, Davis, CA, USA,MIND Institute, University of California, Davis, CA, USA
| | - Michael C. Pride
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA
| | - Elizabeth Edmiston
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, University of California, Davis, CA, USA
| | - Mu Yang
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA,Institute for Genomic Medicine, Columbia University Medical Center, New York, NY, USA
| | - Jill L. Silverman
- MIND Institute, University of California, Davis, CA, USA,Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA
| | - Jacqueline N. Crawley
- MIND Institute, University of California, Davis, CA, USA,Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA
| | - Judy Van de Water
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, University of California, Davis, CA, USA. .,MIND Institute, University of California, Davis, CA, USA.
| |
Collapse
|
18
|
In utero exposure to endogenous maternal polyclonal anti-Caspr2 antibody leads to behavioral abnormalities resembling autism spectrum disorder in male mice. Sci Rep 2020; 10:14446. [PMID: 32879327 PMCID: PMC7468145 DOI: 10.1038/s41598-020-71201-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 07/29/2020] [Indexed: 01/17/2023] Open
Abstract
The concept that exposure in utero to maternal anti-brain antibodies contributes to the development of autism spectrum disorders (ASD) has been entertained for over a decade. We determined that antibodies targeting Caspr2 are present at high frequency in mothers with brain-reactive serology and a child with ASD, and further demonstrated that exposure in utero to a monoclonal anti-Caspr2 antibody, derived from a mother of an ASD child, led to an-ASD like phenotype in male offspring. Now we propose a new model to study the effects of in utero exposure to anti-Caspr2 antibody. Dams immunized with the extracellular portion of Caspr2 express anti-Caspr2 antibodies throughout gestation to better mimic the human condition. Male but not female mice born to dams harboring polyclonal anti-Caspr2 antibodies showed abnormal cortical development, decreased dendritic complexity of excitatory neurons and reduced numbers of inhibitory neurons in the hippocampus, as well as repetitive behaviors and impairments in novelty interest in the social preference test as adults. These data supporting the pathogenicity of anti-Caspr2 antibodies are consistent with the concept that anti-brain antibodies present in women during gestation can alter fetal brain development, and confirm that males are peculiarly susceptible.
Collapse
|
19
|
Marks K, Coutinho E, Vincent A. Maternal-Autoantibody-Related (MAR) Autism: Identifying Neuronal Antigens and Approaching Prospects for Intervention. J Clin Med 2020; 9:jcm9082564. [PMID: 32784803 PMCID: PMC7465310 DOI: 10.3390/jcm9082564] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/31/2020] [Accepted: 08/03/2020] [Indexed: 02/06/2023] Open
Abstract
Recent studies indicate the existence of a maternal-autoantibody-related subtype of autism spectrum disorder (ASD). To date, a large number of studies have focused on describing patterns of brain-reactive serum antibodies in maternal-autoantibody-related (MAR) autism and some have described attempts to define the antigenic targets. This article describes evidence on MAR autism and the various autoantibodies that have been implicated. Among other possibilities, antibodies to neuronal surface protein Contactin Associated Protein 2 (CASPR2) have been found more frequently in mothers of children with neurodevelopmental disorders or autism, and two independent experimental studies have shown pathogenicity in mice. The N-methyl-D-aspartate receptor (NMDAR) is another possible target for maternal antibodies as demonstrated in mice. Here, we discuss the growing evidence, discuss issues regarding biomarker definition, and summarise the therapeutic approaches that might be used to reduce or prevent the transfer of pathogenic maternal antibodies.
Collapse
Affiliation(s)
- Katya Marks
- Medical Sciences Division, John Radcliffe Hospital, University of Oxford, OX3 9DU Oxford, UK;
| | - Ester Coutinho
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King’s College London, SE5 9RT London, UK;
- Nuffield Department of Clinical Neurosciences and Weatherall Institute for Molecular Medicine, University of Oxford, OX3 9DS Oxford, UK
| | - Angela Vincent
- Medical Research Council Centre for Neurodevelopmental Disorders, King’s College London, SE1 1UL London, UK
- Correspondence: ; Tel.: +44-781-722-4849 or +44-186-555-9636
| |
Collapse
|
20
|
Uddin MN, Yao Y, Mondal T, Matala R, Manley K, Lin Q, Lawrence DA. Immunity and autoantibodies of a mouse strain with autistic-like behavior. Brain Behav Immun Health 2020; 4:100069. [PMID: 34589851 PMCID: PMC8474232 DOI: 10.1016/j.bbih.2020.100069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 12/20/2022] Open
Abstract
Female and male mice of the BTBR T + Itpr3 tf /J (BTBR) strain have behaviors that resemble autism spectrum disorder. In comparison to C57BL/6 (B6) mice, BTBR mice have elevated humoral immunity, in that they have naturally high serum IgG levels and generate high levels of IgG antibodies, including autoantibodies to brain antigens. This study focused on the specificities of autoantibodies and the immune cells and their transcription factors that might be responsible for the autoantibodies. BTBR IgG autoantibodies bind to neurons better than microglia and with highest titer to nuclear antigens. Two of the antigens identified were alpha-enolase (ENO1) and dihydrolipoyllysine-residue succinyltransferase component of 2-oxoglutarate dehydrogenase complex, mitochondrial (DLST). Surprisingly based on IgG levels, the blood and spleens of BTBR mice have more CD4+ and CD8+ T cells, but fewer B cells than B6 mice. The high levels of autoantibodies in BTBR relates to their splenic T follicular helper (Tfh) cell levels, which likely are responsible for the higher number of plasma cells in BTBR mice than B6 mice. BTBR mice have increased gene expression of interleukin-21 receptor (I l -21 r) and Paired Box 5 (Pax5), which are known to aid B cell differentiation to plasma cells, and an increased Lysine Demethylase 6B (Kdm6b)/DNA Methyltransferase 1 (Dnmt1) ratio, which increases gene expression. Identification of gene expression and immune activities of BTBR mice may aid understanding of mechanisms associated with autism since neuroimmune network interactions have been posited and induction of autoantibodies may drive the neuroinflammation associated with autism.
Collapse
Key Words
- ASD, autism spectrum disorder
- Ab, antibody
- Ag, antigen
- Alpha-enolase
- Autism
- Autoantibody
- BM, bone marrow
- BTBR
- Dlst, dihydrolipoyllysine-residue succinyltransferase component of 2-oxoglutarate dehydrogenase complex, mitochondrial
- Dnmt1
- Dnmt1, DNA Methyltransferase 1
- Eno1, alpha-enolase
- IL-21r
- IL21R, interleukin-21 receptor
- Kdm6b
- Kdm6b, Lysine Demethylase 6B
- Pax5
- Pax5, Paired Box 5
- Plasma cell
- T follicular helper cell
- Tfh, T follicular helper cell
Collapse
Affiliation(s)
- Mohammad Nizam Uddin
- Wadsworth Center/New York State Department of Health, RNA Epitranscriptomics & Proteomics Resource, SUNY at Albany, Albany, NY, USA
| | - Yunyi Yao
- Wadsworth Center/New York State Department of Health, RNA Epitranscriptomics & Proteomics Resource, SUNY at Albany, Albany, NY, USA
| | - Tapan Mondal
- Wadsworth Center/New York State Department of Health, RNA Epitranscriptomics & Proteomics Resource, SUNY at Albany, Albany, NY, USA
| | - Rosemary Matala
- University at Albany School of Public Health, Rensselaer, NY, USA
| | - Kevin Manley
- Wadsworth Center/New York State Department of Health, RNA Epitranscriptomics & Proteomics Resource, SUNY at Albany, Albany, NY, USA
| | - Qishan Lin
- RNA Epitranscriptomics & Proteomics Resource, SUNY at Albany, Albany, NY, USA
| | - David A Lawrence
- Wadsworth Center/New York State Department of Health, RNA Epitranscriptomics & Proteomics Resource, SUNY at Albany, Albany, NY, USA.,University at Albany School of Public Health, Rensselaer, NY, USA
| |
Collapse
|
21
|
Ramirez-Celis A, Edmiston E, Schauer J, Vu T, Van de Water J. Peptides of neuron specific enolase as potential ASD biomarkers: From discovery to epitope mapping. Brain Behav Immun 2020; 84:200-208. [PMID: 31812776 PMCID: PMC7010557 DOI: 10.1016/j.bbi.2019.12.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/23/2019] [Accepted: 12/02/2019] [Indexed: 11/18/2022] Open
Abstract
Autism spectrum disorder (ASD) is an important health issue and affects 1 in 59 children in the US. Prior studies determined that maternal autoantibody related (MAR) autism is thought to be associated with ~23% of ASD cases. We previously identified seven MAR-specific autoantigens including CRMP1, CRMP2, GDA, LDHA, LDHB, STIP1, and YBX1. We subsequently described the epitope peptide sequences recognized by maternal autoantibodies for each of the seven ASD-specific autoantigens. The aim of the current study was to expand upon our previous work and identify additional antigens recognized by the ASD-specific maternal autoantibodies, as well as to map the unique ASD-specific epitopes using microarray technology. Fetal Rhesus macaque brain tissues were separated by molecular weight and a fraction containing bands between 37 and 45 kDa was analyzed using 2-D gel electrophoresis, followed by peptide mass mapping using MALDI-TOF MS and TOF/TOF tandem MS/MS. Using this methodology, Neuron specific enolase (NSE) was identified as a target autoantigen and selected for epitope mapping. The full NSE sequence was translated into 15-mer peptides with an overlap of 14 amino acids onto microarray slides and probed with maternal plasma from mothers with an ASD child and from mothers with a Typically Developing child (TD) (ASD = 27 and TD = 21). The resulting data were analyzed by T-test. We found 16 ASD-specific NSE-peptide sequences for which four sequences were statistically significant (p < 0.05) using both the t-test and SAM t-test: DVAASEFYRDGKYDL (p = 0.047; SAM score 1.49), IEDPFDQDDWAAWSK (p = 0.049; SAM score 1.49), ERLAKYNQLMRIEEE (p = 0.045; SAM score 1.57), and RLAKYNQLMRIEEEL (p = 0.017; SAM score 1.82). We further identified 5 sequences that were recognized by both ASD and TD antibodies suggesting a large immunodominant epitope (DYPVVSIEDPFDQDDWAAW). While maternal autoantibodies against the NSE protein are present both in mothers with ASD and mothers of TD children, there are several ASD-specific epitopes that can potentially be used as MAR ASD biomarkers. Further, studies including analysis of NSE as a target protein in combination with the previously identified MAR ASD autoantigens are currently underway.
Collapse
Affiliation(s)
- Alexandra Ramirez-Celis
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, University of California, One Shields Avenue, Davis, CA 95616, USA.
| | - Elizabeth Edmiston
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, University of California, One Shields Avenue, Davis, CA 95616, USA
| | - Joseph Schauer
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, University of California, One Shields Avenue, Davis, CA 95616, USA.
| | - Tam Vu
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, University of California, One Shields Avenue, Davis, CA 95616, USA.
| | - Judy Van de Water
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, University of California, One Shields Avenue, Davis, CA 95616, USA; UC Davis MIND Institute, 2825 50th St, Sacramento, CA 95817, USA.
| |
Collapse
|
22
|
Li C, Liu Y, Fang H, Chen Y, Weng J, Zhai M, Xiao T, Ke X. Study on Aberrant Eating Behaviors, Food Intolerance, and Stereotyped Behaviors in Autism Spectrum Disorder. Front Psychiatry 2020; 11:493695. [PMID: 33240114 PMCID: PMC7678488 DOI: 10.3389/fpsyt.2020.493695] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 10/02/2020] [Indexed: 01/29/2023] Open
Abstract
Objective: To investigate the aberrant eating behaviors (EBs), gastrointestinal (GI) symptoms, and food intolerance in children with autism spectrum disorder (ASD) and their association with clinical core symptoms of ASD. Method: A total of 94 preschool children with ASD treated at the Child Mental Health Research Center of Nanjing Brain Hospital between October 2016 and April 2018 were enrolled. In addition, 90 children with typical development (TD) in the community during the same period were recruited. The conditions of aberrant EBs and GI symptoms in children were investigated using questionnaire surveys. Serum specific IgG antibodies against 14 kinds of food were detected using enzyme-linked immunosorbent assays (ELISAs). Results: The detection rate of aberrant EBs in the ASD group was significantly higher than that in the TD group (67.39 vs. 34.94%), and the rate of GI symptoms was also higher in the ASD group than that in the TD group (80.22 vs. 42.11%). Detection of food intolerance in children with ASD showed that the positive rate was 89.89% and that the majority of children had multiple food intolerances. The correlation analysis results showed that the severity of aberrant EBs positively correlated with stereotyped behavior of children with ASD (r = 0.21, P = 0.04) and that food-specific IgG antibodies concentrations positively correlated with high-level stereotyped behavior in children with ASD (r = 0.23, P = 0.03). Conclusion: ASD with aberrant EBs or high food-specific IgG antibodies concentrations had more severe stereotyped behavior, which may have implications for exploring the immune mechanism of ASD. Clinical Trial Registration: ChiCTR-RPC-16008139.
Collapse
Affiliation(s)
- Chunyan Li
- Children's Mental Health Research Center, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Yang Liu
- Children's Mental Health Research Center, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Hui Fang
- Children's Mental Health Research Center, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Yu Chen
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Jiao Weng
- Children's Mental Health Research Center, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Mengyao Zhai
- Children's Mental Health Research Center, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Ting Xiao
- Children's Mental Health Research Center, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaoyan Ke
- Children's Mental Health Research Center, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
23
|
Dudova I, Horackova K, Hrdlicka M, Balastik M. Can Maternal Autoantibodies Play an Etiological Role in ASD Development? Neuropsychiatr Dis Treat 2020; 16:1391-1398. [PMID: 32581542 PMCID: PMC7276202 DOI: 10.2147/ndt.s239504] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 04/10/2020] [Indexed: 12/20/2022] Open
Abstract
Autism spectrum disorder (ASD) is a heterogeneous condition with multiple etiologies and risk factors - both genetic and environmental. Recent data demonstrate that the immune system plays an important role in prenatal brain development. Deregulation of the immune system during embryonic development can lead to neurodevelopmental changes resulting in ASD. One of the potential etiologic factors in the development of ASD has been identified as the presence of maternal autoantibodies targeting fetal brain proteins. The type of ASD associated with the presence of maternal autoantibodies has been referred to as maternal antibodies related to ASD (MAR ASD). The link between maternal autoantibodies and ASD has been demonstrated in both clinical studies and animal models, but the exact mechanism of their action in the pathogenesis of ASD has not been clarified yet. Several protein targets of ASD-related maternal autoantibodies have been identified. Here, we discuss the role of microtubule-associated proteins of the collapsin response mediator protein (CRMP) family in neurodevelopment and ASD. CRMPs have been shown to integrate multiple signaling cascades regulating neuron growth, guidance or migration. Their targeting by maternal autoantibodies could change CRMP levels or distribution in the developing nervous system, leading to defects in axon growth/guidance, cortical migration, or dendritic projection, which could play an etiological role in ASD development. In addition, we discuss the future possibilities of MAR ASD treatment.
Collapse
Affiliation(s)
- Iva Dudova
- Department of Child Psychiatry, Charles University Second Faculty of Medicine, Prague, Czech Republic
| | - Klara Horackova
- Department of Psychiatry, Charles University First Faculty of Medicine, Prague, Czech Republic
| | - Michal Hrdlicka
- Department of Child Psychiatry, Charles University Second Faculty of Medicine, Prague, Czech Republic
| | - Martin Balastik
- Laboratory of Molecular Neurobiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
24
|
Afroz KF, Alviña K. Maternal elevated salt consumption and the development of autism spectrum disorder in the offspring. J Neuroinflammation 2019; 16:265. [PMID: 31837704 PMCID: PMC6911292 DOI: 10.1186/s12974-019-1666-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 11/27/2019] [Indexed: 01/15/2023] Open
Abstract
Autism spectrum disorder (ASD) is a prevalent neurodevelopmental condition with no known etiology or cure. Several possible contributing factors, both genetic and environmental, are being actively investigated. Amongst these, maternal immune dysregulation has been identified as potentially involved in promoting ASD in the offspring. Indeed, ASD-like behaviors have been observed in studies using the maternal immune activation mouse model. Furthermore, recent studies have shed light on maternal dietary habits and their impact on the gut microbiome as factors possibly facilitating ASD. However, most of these studies have been limited to the effects of high fat and/or high sugar. More recent data, however, have shown that elevated salt consumption has a significant effect on the immune system and gut microbiome, often resulting in gut dysbiosis and induction of pro-inflammatory pathways. Specifically, high salt alters the gut microbiome and induces the differentiation of T helper-17 cells that produce pro-inflammatory cytokines such as interleukin-17 and interleukin-23. Moreover, elevated salt can also reduce the differentiation of regulatory T cells that help maintaining a balanced immune system. While in the innate immune system, high salt can cause over activation of M1 pro-inflammatory macrophages and downregulation of M2 regulatory macrophages. These changes to the immune system are alarming because excessive consumption of salt is a documented worldwide problem. Thus, in this review, we discuss recent findings on high salt intake, gut microbiome, and immune system dysregulation while proposing a hypothesis to link maternal overconsumption of salt and children’s ASD.
Collapse
Affiliation(s)
- Kazi Farhana Afroz
- Department of Biological Sciences, Texas Tech University, 2901 Main St. Room #05, Biology Building, Lubbock, TX, 79409, USA
| | - Karina Alviña
- Department of Biological Sciences, Texas Tech University, 2901 Main St. Room #05, Biology Building, Lubbock, TX, 79409, USA. .,Department of Neuroscience, University of Florida, 1149 Newell Drive, Room L1-100, Gainesville, FL, 32611, USA.
| |
Collapse
|
25
|
Álvarez-Lindo N, Baleriola J, de Los Ríos V, Suárez T, de la Rosa EJ. RAG-2 deficiency results in fewer phosphorylated histone H2AX foci, but increased retinal ganglion cell death and altered axonal growth. Sci Rep 2019; 9:18486. [PMID: 31811168 PMCID: PMC6898044 DOI: 10.1038/s41598-019-54873-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 11/19/2019] [Indexed: 02/06/2023] Open
Abstract
DNA double-strand breaks (DSBs), selectively visualized as γ-H2AX+ foci, occur during the development of the central nervous system, including the retina, although their origin and biological significance are poorly understood. Mutant mice with DSB repair mechanism defects exhibit increased numbers of γ-H2AX+ foci, increased cell death during neural development, and alterations in axonogenesis in the embryonic retina. The aim of this study was to identify putative sources of DSBs. One of the identified DSBs sources is LINE-1 retrotransposition. While we did not detect changes in LINE-1 DNA content during the early period of cell death associated with retinal neurogenesis, retinal development was altered in mice lacking RAG-2, a component of the RAG-1,2-complex responsible for initiating somatic recombination in lymphocytes. Although γ-H2AX+ foci were less abundant in the rag2−/− mouse retina, retinal ganglion cell death was increased and axonal growth and navigation were impaired in the RAG-2 deficient mice, a phenotype shared with mutant mice with defective DNA repair mechanisms. These findings demonstrate that RAG-2 is necessary for proper retinal development, and suggest that both DSB generation and repair are genuine processes intrinsic to neural development.
Collapse
Affiliation(s)
- Noemí Álvarez-Lindo
- 3D Lab: Development, Differentiation & Degeneration, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CIB/CSIC), Madrid, Spain
| | - Jimena Baleriola
- 3D Lab: Development, Differentiation & Degeneration, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CIB/CSIC), Madrid, Spain.,Laboratory of local translation in neurons and glia, Achucarro Basque Center for Neuroscience; Department of Cell Biology and Histology, University of the Basque Country, Leioa; and Ikerbasque Foundation, Bilbao, Bizkaia, Spain
| | - Vivian de Los Ríos
- Proteomics and Genomics, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CIB/CSIC), Madrid, Spain
| | - Teresa Suárez
- 3D Lab: Development, Differentiation & Degeneration, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CIB/CSIC), Madrid, Spain
| | - Enrique J de la Rosa
- 3D Lab: Development, Differentiation & Degeneration, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CIB/CSIC), Madrid, Spain.
| |
Collapse
|
26
|
Frye RE, Vassall S, Kaur G, Lewis C, Karim M, Rossignol D. Emerging biomarkers in autism spectrum disorder: a systematic review. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:792. [PMID: 32042808 DOI: 10.21037/atm.2019.11.53] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Autism spectrum disorder (ASD) affects approximately 2% of children in the United States (US) yet its etiology is unclear and effective treatments are lacking. Therapeutic interventions are most effective if started early in life, yet diagnosis often remains delayed, partly because the diagnosis of ASD is based on identifying abnormal behaviors that may not emerge until the disorder is well established. Biomarkers that identify children at risk during the pre-symptomatic period, assist with early diagnosis, confirm behavioral observations, stratify patients into subgroups, and predict therapeutic response would be a great advance. Here we underwent a systematic review of the literature on ASD to identify promising biomarkers and rated the biomarkers in regards to a Level of Evidence and Grade of Recommendation using the Oxford Centre for Evidence-Based Medicine scale. Biomarkers identified by our review included physiological biomarkers that identify neuroimmune and metabolic abnormalities, neurological biomarkers including abnormalities in brain structure, function and neurophysiology, subtle behavioral biomarkers including atypical development of visual attention, genetic biomarkers and gastrointestinal biomarkers. Biomarkers of ASD may be found prior to birth and after diagnosis and some may predict response to specific treatments. Many promising biomarkers have been developed for ASD. However, many biomarkers are preliminary and need to be validated and their role in the diagnosis and treatment of ASD needs to be defined. It is likely that biomarkers will need to be combined to be effective to identify ASD early and guide treatment.
Collapse
Affiliation(s)
- Richard E Frye
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA.,Deparment of Child Health, University of Arizona College of Medicine, Phoenix, AZ, USA
| | - Sarah Vassall
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA
| | - Gurjot Kaur
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA
| | - Christina Lewis
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA
| | - Mohammand Karim
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA.,Deparment of Child Health, University of Arizona College of Medicine, Phoenix, AZ, USA
| | | |
Collapse
|
27
|
Bolandparvaz A, Harriman R, Alvarez K, Lilova K, Zang Z, Lam A, Edmiston E, Navrotsky A, Vapniarsky N, Van De Water J, Lewis JS. Towards a nanoparticle-based prophylactic for maternal autoantibody-related autism. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2019; 21:102067. [PMID: 31349087 PMCID: PMC7197945 DOI: 10.1016/j.nano.2019.102067] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 06/21/2019] [Accepted: 07/12/2019] [Indexed: 12/17/2022]
Abstract
Recently, the causative agents of Maternal Autoantibody-Related (MAR) autism, pathological autoantibodies and their epitopic targets (e.g. lactate dehydrogenase B [LDH B] peptide), have been identified. Herein, we report on the development of Systems for Nanoparticle-based Autoantibody Reception and Entrapment (SNAREs), which we hypothesized could scavenge disease-propagating MAR autoantibodies from the maternal blood. To demonstrate this functionality, we synthesized 15 nm dextran iron oxide nanoparticles surface-modified with citric acid, methoxy PEG(10 kDa) amine, and LDH B peptide (33.8 μg peptide/cm2). In vitro, we demonstrated significantly lower macrophage uptake for SNAREs compared to control NPs. The hallmark result of this study was the efficacy of the SNAREs to remove 90% of LDH B autoantibody from patient-derived serum. Further, in vitro cytotoxicity testing and a maximal tolerated dose study in mice demonstrated the safety of the SNARE formulation. This work establishes the feasibility of SNAREs as the first-ever prophylactic against MAR autism.
Collapse
Affiliation(s)
- Amir Bolandparvaz
- University of California, Davis, Department of Biomedical Engineering, Davis, CA, USA
| | - Rian Harriman
- University of California, Davis, Department of Biomedical Engineering, Davis, CA, USA
| | - Kenneth Alvarez
- University of California, Davis, Department of Biomedical Engineering, Davis, CA, USA
| | - Kristina Lilova
- University of California, Davis, Peter A. Rock Thermochemistry Laboratory and NEAT, Davis, CA, USA
| | - Zexi Zang
- University of California, Davis, Department of Biomedical Engineering, Davis, CA, USA
| | - Andy Lam
- University of California, Davis, Peter A. Rock Thermochemistry Laboratory and NEAT, Davis, CA, USA
| | - Elizabeth Edmiston
- University of California, Davis, Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, Davis, CA, USA
| | - Alexandra Navrotsky
- University of California, Davis, Peter A. Rock Thermochemistry Laboratory and NEAT, Davis, CA, USA
| | - Natalia Vapniarsky
- University of California, Davis, Department of Pathology Microbiology and Immunology, Davis, CA, USA
| | - Judy Van De Water
- University of California, Davis, Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, Davis, CA, USA; University of California, Davis, M.I.N.D. (Medical Investigation of Neurodevelopmental Disorders), Davis, CA, USA
| | - Jamal S Lewis
- University of California, Davis, Department of Biomedical Engineering, Davis, CA, USA.
| |
Collapse
|
28
|
Pröbstel AK, Zamvil SS. Do maternal anti-N-methyl-D-aspartate receptor antibodies promote development of neuropsychiatric disease in children? Ann Neurol 2019; 86:653-655. [PMID: 31531881 PMCID: PMC6856826 DOI: 10.1002/ana.25584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 08/18/2019] [Indexed: 01/05/2023]
Affiliation(s)
- Anne-Katrin Pröbstel
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA.,Neurologic Clinic and Policlinic, Departments of Medicine and Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Scott S Zamvil
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA.,Program in Immunology, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
29
|
Jurek B, Chayka M, Kreye J, Lang K, Kraus L, Fidzinski P, Kornau HC, Dao LM, Wenke NK, Long M, Rivalan M, Winter Y, Leubner J, Herken J, Mayer S, Mueller S, Boehm-Sturm P, Dirnagl U, Schmitz D, Kölch M, Prüss H. Human gestational N-methyl-d-aspartate receptor autoantibodies impair neonatal murine brain function. Ann Neurol 2019; 86:656-670. [PMID: 31325344 DOI: 10.1002/ana.25552] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 07/12/2019] [Accepted: 07/15/2019] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Maternal autoantibodies are a risk factor for impaired brain development in offspring. Antibodies (ABs) against the NR1 (GluN1) subunit of the N-methyl-d-aspartate receptor (NMDAR) are among the most frequently diagnosed anti-neuronal surface ABs, yet little is known about effects on fetal development during pregnancy. METHODS We established a murine model of in utero exposure to human recombinant NR1 and isotype-matched nonreactive control ABs. Pregnant C57BL/6J mice were intraperitoneally injected on embryonic days 13 and 17 each with 240μg of human monoclonal ABs. Offspring were investigated for acute and chronic effects on NMDAR function, brain development, and behavior. RESULTS Transferred NR1 ABs enriched in the fetus and bound to synaptic structures in the fetal brain. Density of NMDAR was considerably reduced (up to -49.2%) and electrophysiological properties were altered, reflected by decreased amplitudes of spontaneous excitatory postsynaptic currents in young neonates (-34.4%). NR1 AB-treated animals displayed increased early postnatal mortality (+27.2%), impaired neurodevelopmental reflexes, altered blood pH, and reduced bodyweight. During adolescence and adulthood, animals showed hyperactivity (+27.8% median activity over 14 days), lower anxiety, and impaired sensorimotor gating. NR1 ABs caused long-lasting neuropathological effects also in aged mice (10 months), such as reduced volumes of cerebellum, midbrain, and brainstem. INTERPRETATION The data collectively support a model in which asymptomatic mothers can harbor low-level pathogenic human NR1 ABs that are diaplacentally transferred, causing neurotoxic effects on neonatal development. Thus, AB-mediated network changes may represent a potentially treatable neurodevelopmental congenital brain disorder contributing to lifelong neuropsychiatric morbidity in affected children. ANN NEUROL 2019;86:656-670.
Collapse
Affiliation(s)
- Betty Jurek
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany
| | - Mariya Chayka
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany
| | - Jakob Kreye
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany
| | - Katharina Lang
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany
| | - Larissa Kraus
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Clinical and Experimental Epileptology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Pawel Fidzinski
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Clinical and Experimental Epileptology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Hans-Christian Kornau
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany.,Neuroscience Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Le-Minh Dao
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany
| | - Nina K Wenke
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany
| | - Melissa Long
- Neurocure Cluster of Excellence, Animal Outcome Core Facility, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Marion Rivalan
- Neurocure Cluster of Excellence, Animal Outcome Core Facility, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - York Winter
- Neurocure Cluster of Excellence, Animal Outcome Core Facility, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jonas Leubner
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Julia Herken
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Simone Mayer
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research and Department of Neurology, University of California, San Francisco, San Francisco, CA
| | - Susanne Mueller
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Neurocure Cluster of Excellence, Core Facility 7T Experimental MRIs, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Center for Stroke Research, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Philipp Boehm-Sturm
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Neurocure Cluster of Excellence, Core Facility 7T Experimental MRIs, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Center for Stroke Research, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Ulrich Dirnagl
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany.,Center for Stroke Research, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Dietmar Schmitz
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany.,Neuroscience Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Einstein Center for Neurosciences, Berlin, Germany
| | - Michael Kölch
- Department for Child and Adolescent Psychiatry, Neurology, Psychosomatics, and Psychotherapy, Universitätsmedizin Rostock, Rostock, Germany
| | - Harald Prüss
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany.,Department of Neurology, Center for Autoimmune Encephalitis and Paraneoplastic Neurological Syndromes, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
30
|
Zheng W, Hu Y, Chen D, Li Y, Wang S. [Improvement of a mouse model of valproic acid-induced autism]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2019; 39:718-723. [PMID: 31270052 DOI: 10.12122/j.issn.1673-4254.2019.06.14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To establish an improved mouse model of valproic acid (VPA)-induced autism that better mimics human autism. METHODS We established mouse models of autism in female C57 mice by intraperitoneal injection of sodium valproate either at a single dose (600 mg/kg) on day 12.5 after conception (conventional group) or in two doses of 300 mg/kg each on days 10 and 12 after conception (modified group), and the control mice were injected with saline only on day 12.5. The responses of the mice to VPA injection, the uterus, mortality rate, and abortion rate were compared among the 3 groups. The morphology and development of the offspring mice were assessed, and their behavioral ontogeny was evaluated using 3- chambered social test, social test, juvenil play test, and open field test. RESULTS The mortality and abortion rates were significantly lower in the modified model group than in the conventional group (P < 0.01). Compared with those in the control group, the offspring mice in both the conventional group and the modified group showed developmental disorders (P < 0.05). The mortality rate of the newborn mice was significantly lower in the modified group than in the conventional group with a rate of curvy tail of up to 100% (P < 0.001). The offspring mice in both the modified group and conventional group exhibited autism-like behavioral abnormalities, including social disorder and repetitive stereotyped behavior (P < 0.05). CONCLUSIONS The mouse model of autism established using the modified method better mimics human autism with reduced mortality and abortion rates of the pregnant mice and also decreased mortality rate of the newborn mice.
Collapse
Affiliation(s)
- Wenxia Zheng
- College of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yuling Hu
- College of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Di Chen
- College of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yingbo Li
- College of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Shali Wang
- College of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
31
|
Mazón-Cabrera R, Vandormael P, Somers V. Antigenic Targets of Patient and Maternal Autoantibodies in Autism Spectrum Disorder. Front Immunol 2019; 10:1474. [PMID: 31379804 PMCID: PMC6659315 DOI: 10.3389/fimmu.2019.01474] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 06/13/2019] [Indexed: 12/13/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder whose behavioral symptoms become apparent in early childhood. The underlying pathophysiological mechanisms are only partially understood and the clinical manifestations are heterogeneous in nature, which poses a major challenge for diagnosis, prognosis and intervention. In the last years, an important role of a dysregulated immune system in ASD has emerged, but the mechanisms connecting this to a disruption of brain development are still largely unknown. Although ASD is not considered as a typical autoimmune disease, self-reactive antibodies or autoantibodies against a wide variety of targets have been found in a subset of ASD patients. In addition, autoantibodies reactive to fetal brain proteins have also been described in the prenatal stage of neurodevelopment, where they can be transferred from the mother to the fetus by transplacental transport. In this review, we give an extensive overview of the antibodies described in ASD according to their target antigens, their different origins, and timing of exposure during neurodevelopment.
Collapse
Affiliation(s)
| | | | - Veerle Somers
- Biomedical Research Institute, Faculty of Medicine and Life Science, Hasselt University, Diepenbeek, Belgium
| |
Collapse
|
32
|
Gata-Garcia A, Diamond B. Maternal Antibody and ASD: Clinical Data and Animal Models. Front Immunol 2019; 10:1129. [PMID: 31191521 PMCID: PMC6547809 DOI: 10.3389/fimmu.2019.01129] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 05/03/2019] [Indexed: 12/26/2022] Open
Abstract
Over the past several decades there has been an increasing interest in the role of environmental factors in the etiology of neuropsychiatric and neurodevelopmental disorders. Epidemiologic studies have shifted from an exclusive focus on the identification of genetic risk alleles for such disorders to recognizing and understanding the contribution of xenobiotic exposures, infections, and the maternal immune system during the prenatal and early post-natal periods. In this review we discuss the growing literature regarding the effects of maternal brain-reactive antibodies on fetal brain development and their contribution to the development of neuropsychiatric and neurodevelopmental disorders. Autoimmune diseases primarily affect women and are more prevalent in mothers of children with neurodevelopmental disorders. For example, mothers of children with Autism Spectrum Disorder (ASD) are significantly more likely to have an autoimmune disease than women of neurotypically developing children. Moreover, they are four to five times more likely to harbor brain-reactive antibodies than unselected women of childbearing age. Many of these women exhibit no apparent clinical consequence of harboring these antibodies, presumably because the antibodies never access brain tissue. Nevertheless, these maternal brain-reactive antibodies can access the fetal brain, and some may be capable of altering brain development when present during pregnancy. Several animal models have provided evidence that in utero exposure to maternal brain-reactive antibodies can permanently alter brain anatomy and cause persistent behavioral or cognitive phenotypes. Although this evidence supports a contribution of maternal brain-reactive antibodies to neurodevelopmental disorders, an interplay between antibodies, genetics, and other environmental factors is likely to determine the specific neurodevelopmental phenotypes and their severity. Additional modulating factors likely also include the microbiome, sex chromosomes, and gonadal hormones. These interactions may help to explain the sex-bias observed in neurodevelopmental disorders. Studies on this topic provide a unique opportunity to learn how to identify and protect at risk pregnancies while also deciphering critical pathways in neurodevelopment.
Collapse
Affiliation(s)
- Adriana Gata-Garcia
- Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Manhasset, NY, United States.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | - Betty Diamond
- Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Manhasset, NY, United States
| |
Collapse
|
33
|
Yang Z, Zhao Y, Li Q, Shao Y, Yu X, Cong W, Jia X, Qu W, Cheng L, Xue P, Zhou Z, He M, Zhang Y. Developmental exposure to mercury chloride impairs social behavior in male offspring dependent on genetic background and maternal autoimmune environment. Toxicol Appl Pharmacol 2019; 370:1-13. [PMID: 30862457 DOI: 10.1016/j.taap.2019.03.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 02/03/2019] [Accepted: 03/08/2019] [Indexed: 10/27/2022]
Abstract
To date, the connection between inorganic mercury (Hg) and social behavior remains incompletely understood. The aim of this study was to investigate the influence of maternal autoimmunity by inorganic Hg (Hg2+) exposure on social behavior of offspring. Wild-type (WT) and immunoglobulin deficient (Ig-/-) B10.S dams fertilized by male WT B10.S or SJL mice were treated with 50 μM Hg chloride (HgCl2). Non-pregnant female WT B10.S mice were used to investigate factors regulating HgCl2-induced autoimmunity to brain. HgCl2 selectively impaired social behavior in male offspring, but not female offspring from WT B10.S dams × male SJL, in that only male offspring displayed reduced time distribution with the stranger mouse, decreased sniffing to the stranger mouse and increased self-grooming. HgCl2 did not disrupt social behavior of male or female offspring from WT B10.S dams × male WT B10.S or Ig-/- B10.S dams × male SJL. The offspring from WT and Ig-/- B10.S dams × male SJL had equivalent autoimmunity to brain antigens during HgCl2 exposure, indicating that maternal, but not offspring-derived anti-brain antibodies (Ab) impaired social behavior of the offspring. Non-pregnant WT B10.S mice treated with HgCl2 had increased anti-brain Ab dependent on increase in CD4 T cell activation and IFNγ signaling to macrophages. IFNγ interaction with macrophages drove B cells and plasma cells to produce IgG. Therefore, HgCl2 selectively impaired social behavior in males with certain genetic background via maternally derived anti-brain Ab production, thus providing a novel insight into our current understanding of Hg toxicity.
Collapse
Affiliation(s)
- Zhengli Yang
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai 200032, China
| | - Yifan Zhao
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai 200032, China
| | - Qian Li
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai 200032, China
| | - Yiming Shao
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai 200032, China
| | - Xinchun Yu
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai 200032, China
| | - Wei Cong
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
| | - Xiaodong Jia
- Shanghai Municipal Center for Disease Control and Prevention, Shanghai 200336, China
| | - Weidong Qu
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai 200032, China
| | - Longzhen Cheng
- Department of Biology, Southern University of Science and Technology, Shenzhen 518055, China
| | - Peng Xue
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai 200032, China
| | - Zhijun Zhou
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai 200032, China
| | - Miao He
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
| | - Yubin Zhang
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai 200032, China.
| |
Collapse
|
34
|
Bölte S, Girdler S, Marschik PB. The contribution of environmental exposure to the etiology of autism spectrum disorder. Cell Mol Life Sci 2019; 76:1275-1297. [PMID: 30570672 PMCID: PMC6420889 DOI: 10.1007/s00018-018-2988-4] [Citation(s) in RCA: 285] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 11/14/2018] [Accepted: 12/04/2018] [Indexed: 01/04/2023]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental condition of heterogeneous etiology. While it is widely recognized that genetic and environmental factors and their interactions contribute to autism phenotypes, their precise causal mechanisms remain poorly understood. This article reviews our current understanding of environmental risk factors of ASD and their presumed adverse physiological mechanisms. It comprehensively maps the significance of parental age, teratogenic compounds, perinatal risks, medication, smoking and alcohol use, nutrition, vaccination, toxic exposures, as well as the role of extreme psychosocial factors. Further, we consider the role of potential protective factors such as folate and fatty acid intake. Evidence indicates an increased offspring vulnerability to ASD through advanced maternal and paternal age, valproate intake, toxic chemical exposure, maternal diabetes, enhanced steroidogenic activity, immune activation, and possibly altered zinc-copper cycles and treatment with selective serotonin reuptake inhibitors. Epidemiological studies demonstrate no evidence for vaccination posing an autism risk. It is concluded that future research needs to consider categorical autism, broader autism phenotypes, as well as autistic traits, and examine more homogenous autism variants by subgroup stratification. Our understanding of autism etiology could be advanced by research aimed at disentangling the causal and non-causal environmental effects, both founding and moderating, and gene-environment interplay using twin studies, longitudinal and experimental designs. The specificity of many environmental risks for ASD remains unknown and control of multiple confounders has been limited. Further understanding of the critical windows of neurodevelopmental vulnerability and investigating the fit of multiple hit and cumulative risk models are likely promising approaches in enhancing the understanding of role of environmental factors in the etiology of ASD.
Collapse
Affiliation(s)
- Sven Bölte
- Department of Women's and Children's Health, Karolinska Institutet & Child and Adolescent Psychiatry, Stockholm Health Care Services, Center of Neurodevelopmental Disorders (KIND), Centre for Psychiatry Research, Stockholm County Council, Stockholm, Sweden.
- Curtin Autism Research Group, School of Occupational Therapy, Social Work and Speech Pathology, Curtin University, Perth, WA, Australia.
| | - Sonya Girdler
- Curtin Autism Research Group, School of Occupational Therapy, Social Work and Speech Pathology, Curtin University, Perth, WA, Australia
| | - Peter B Marschik
- Department of Women's and Children's Health, Karolinska Institutet & Child and Adolescent Psychiatry, Stockholm Health Care Services, Center of Neurodevelopmental Disorders (KIND), Centre for Psychiatry Research, Stockholm County Council, Stockholm, Sweden
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
- iDN-interdisciplinary Developmental Neuroscience, Department of Phoniatrics, Medical University of Graz, Graz, Austria
| |
Collapse
|
35
|
Puljevic M, Danilowicz‐Szymanowicz L, Molon G, Puljevic D, Raczak G, Canali G, Velagic V, Pezo‐Nikolic B, Milicic D. TWARMI pilot trial: The value and optimal criteria of microvolt T-wave alternans in the diagnosis of reversible myocardial ischemia in patients without structural cardiac disease. Ann Noninvasive Electrocardiol 2019; 24:e12610. [PMID: 30383318 PMCID: PMC6931817 DOI: 10.1111/anec.12610] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 09/19/2018] [Accepted: 10/08/2018] [Indexed: 12/13/2022] Open
Abstract
AIMS Microvolt T-wave alternans (MTWA) testing is a beat-to-beat fluctuation in the amplitude of T wave. We investigated whether: (a) MTWA can be new non-invasive tool for detection of reversible ischemia in patients with suspected CAD without structural heart disease, (b) MTWA can detect ischemia earlier and with greater test accuracy compared with exercise ECG ST-segment testing, and (c) threshold value of MTWA and heart rate at which the alternans is estimated can be different compared to standard values. METHODS A total of 101 patients with suspected stable coronary disease, but without structural heart disease, were included. Echocardiography, exercise ECG test, MTWA with classical and modified threshold alternans values, and coronary angiography were performed. RESULTS About 33.3% patients had a false-positive result on exercise ECG test. The sensitivity of exercise ECG ST-segment test in the detection of coronary artery disease was 97.8%, and the specificity was 42.5% (DOR 33.89). In a group of angiographically positive patients, standard MTWA accurately identified 60% of patients, while 40% had a false-negative result. About 91.8% patients with negative angiography result were accurately identified with 8.2% false positives. The sensitivity of MTWA was 59.61% and specificity 91.83%. Best ratio of sensitivity and specificity (86.53% and 95.91%, DOR 151.06) had modified criteria for positive MTWA (MTWA >1.5 µV at heart rate 115-125/min). CONCLUSIONS This study showed that MTWA can be the new non-invasive tool for the detection of reversible ischemia in patients with suspected CAD without structural heart disease. Also, MTWA can detect ischemia earlier and with greater accuracy compared with exercise ECG testing.
Collapse
Affiliation(s)
- Mislav Puljevic
- University of Zagreb, School of Medicine, University Hospital Center ZagrebZagrebCroatia
| | | | - Giulio Molon
- Cardiology DepartmentS. Cuore HospitalVeronaItaly
| | - Davor Puljevic
- University of Zagreb, School of Medicine, University Hospital Center ZagrebZagrebCroatia
| | - Grzegorz Raczak
- Department of Cardiology and ElectrotherapyMedical University of GdanskGdanskPoland
| | - Guido Canali
- Cardiology DepartmentS. Cuore HospitalVeronaItaly
| | - Vedran Velagic
- University of Zagreb, School of Medicine, University Hospital Center ZagrebZagrebCroatia
| | - Borka Pezo‐Nikolic
- University of Zagreb, School of Medicine, University Hospital Center ZagrebZagrebCroatia
| | - Davor Milicic
- University of Zagreb, School of Medicine, University Hospital Center ZagrebZagrebCroatia
| |
Collapse
|
36
|
Liu Q, Chen MX, Sun L, Wallis CU, Zhou JS, Ao LJ, Li Q, Sham PC. Rational use of mesenchymal stem cells in the treatment of autism spectrum disorders. World J Stem Cells 2019; 11:55-72. [PMID: 30842805 PMCID: PMC6397804 DOI: 10.4252/wjsc.v11.i2.55] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 12/30/2018] [Accepted: 01/23/2019] [Indexed: 02/06/2023] Open
Abstract
Autism and autism spectrum disorders (ASD) refer to a range of conditions characterized by impaired social and communication skills and repetitive behaviors caused by different combinations of genetic and environmental influences. Although the pathophysiology underlying ASD is still unclear, recent evidence suggests that immune dysregulation and neuroinflammation play a role in the etiology of ASD. In particular, there is direct evidence supporting a role for maternal immune activation during prenatal life in neurodevelopmental conditions. Currently, the available options of behavioral therapies and pharmacological and supportive nutritional treatments in ASD are only symptomatic. Given the disturbing rise in the incidence of ASD, and the fact that there is no effective pharmacological therapy for ASD, there is an urgent need for new therapeutic options. Mesenchymal stem cells (MSCs) possess immunomodulatory properties that make them relevant to several diseases associated with inflammation and tissue damage. The paracrine regenerative mechanisms of MSCs are also suggested to be therapeutically beneficial for ASD. Thus the underlying pathology in ASD, including immune system dysregulation and inflammation, represent potential targets for MSC therapy. This review will focus on immune dysfunction in the pathogenesis of ASD and will further discuss the therapeutic potential for MSCs in mediating ASD-related immunological disorders.
Collapse
Affiliation(s)
- Qiang Liu
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China
| | - Mo-Xian Chen
- School of Rehabilitation, Kunming Medical University, Kunming 650500, Yunnan Province, China
| | - Lin Sun
- Department of Psychology, Weifang Medical University, Weifang 261053, Shandong Province, China
| | - Chloe U Wallis
- Medical Sciences Division, University of Oxford, Oxford OX3 9DU, United Kingdom
| | - Jian-Song Zhou
- Mental Health Institute of the Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Li-Juan Ao
- School of Rehabilitation, Kunming Medical University, Kunming 650500, Yunnan Province, China
| | - Qi Li
- Department of Psychiatry, the University of Hong Kong, Hong Kong, China
| | - Pak C Sham
- Department of Psychiatry, the University of Hong Kong, Hong Kong, China
- State Key Laboratory of Brain and Cognitive Sciences, Center for Genomic Sciences, the University of Hong Kong, Hong Kong, China
| |
Collapse
|
37
|
Jones KL, Van de Water J. Maternal autoantibody related autism: mechanisms and pathways. Mol Psychiatry 2019; 24:252-265. [PMID: 29934547 PMCID: PMC6784837 DOI: 10.1038/s41380-018-0099-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 04/12/2018] [Accepted: 04/23/2018] [Indexed: 12/16/2022]
Abstract
It has been estimated that autism spectrum disorder (ASD) now affects 1 in 59 children in the United States. Although the cause(s) of ASD remain largely unknown, it is becoming increasingly apparent that ASD can no longer be defined simply as a behavioral disorder, but is in effect a rather complex and highly heterogeneous biological disorder. Up until recently the brain was thought to be "immune privileged." However, it is now known that the immune system plays critical roles in the development and functioning of the brain throughout life. Recent evidence from multiple investigators has illustrated the deleterious role that dysregulation of the maternal immune system during gestation can play in the manifestation of changes in neurodevelopment, resulting in the development of neurobehavioral disorders such as ASD. One potential etiologic pathway through which the maternal immune system can interfere with neurodevelopment is through maternal autoantibodies that recognize proteins in the developing fetal brain. This mechanism of pathogenesis is now thought to lead to a subphenotype of ASD that has been termed maternal autoantibody related (MAR) ASD. This review provides an overview of the current research implicating the presence of brain-reactive maternal autoantibodies as a risk factor for MAR ASD.
Collapse
Affiliation(s)
- Karen L. Jones
- Rheumatology/Allergy and Clinical Immunology, University of California, 451 E. Health Sciences Drive, Suite 6510 GBSF, Davis, CA 95616, USA,The M.I.N.D. Institute, University of California, Davis, CA 95616, USA
| | - Judy Van de Water
- Rheumatology/Allergy and Clinical Immunology, University of California, 451 E. Health Sciences Drive, Suite 6510 GBSF, Davis, CA, 95616, USA. .,The M.I.N.D. Institute, University of California, Davis, CA, 95616, USA. .,NIEHS Center for Children's Environmental Health, University of California, Davis, CA, 95616, USA.
| |
Collapse
|
38
|
Azhari A, Azizan F, Esposito G. A systematic review of gut-immune-brain mechanisms in Autism Spectrum Disorder. Dev Psychobiol 2018; 61:752-771. [PMID: 30523646 DOI: 10.1002/dev.21803] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 09/10/2018] [Accepted: 10/09/2018] [Indexed: 12/13/2022]
Abstract
Despite decades of research, the etiological origins of Autism Spectrum Disorder (ASD) remain elusive. Recently, the mechanisms of ASD have encompassed emerging theories involving the gastrointestinal, immune, and nervous systems. While each of these perspectives presents its own set of supporting evidence, the field requires an integration of these modular concepts and an overarching view of how these subsystems intersect. In this systematic review, we have synthesized relevant evidences from the existing literature, evaluating them in an interdependent manner and in doing so, outlining their possible connections. Specifically, we first discussed gastrointestinal and immuno-inflammation pathways in-depth, exploring the relationships between microbial composition, bacterial metabolites, gut mucosa, and immune system constituents. Accounting for temporal differences in the mechanisms involved in neurodevelopment, prenatal and postnatal phases were further elucidated, where the former focused on maternal immune activation (MIA) and fetal development, while the latter addressed the role of immune dysregulation in contributing to atypical neurodevelopment. As autism remains, foremost, a neurodevelopmental disorder, this review presents an integration of disparate modules into a "Gut-Immune-Brain" paradigm. Existing gaps in the literature have been highlighted, and possible avenues for future research with an integrated physiological perspective underlying ASD have also been suggested.
Collapse
Affiliation(s)
- Atiqah Azhari
- Psychology Program, School of Social Sciences, Nanyang Technological University, Singapore, Singapore
| | - Farouq Azizan
- Psychology Program, School of Social Sciences, Nanyang Technological University, Singapore, Singapore
| | - Gianluca Esposito
- Psychology Program, School of Social Sciences, Nanyang Technological University, Singapore, Singapore.,Department of Psychology and Cognitive Science, University of Trento, Rovereto, TN, Italy
| |
Collapse
|
39
|
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: 156] [Impact Index Per Article: 22.3] [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
|
40
|
Dadalko OI, Travers BG. Evidence for Brainstem Contributions to Autism Spectrum Disorders. Front Integr Neurosci 2018; 12:47. [PMID: 30337860 PMCID: PMC6180283 DOI: 10.3389/fnint.2018.00047] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 09/18/2018] [Indexed: 12/27/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental condition that affects one in 59 children in the United States. Although there is a mounting body of knowledge of cortical and cerebellar contributions to ASD, our knowledge about the early developing brainstem in ASD is only beginning to accumulate. Understanding how brainstem neurotransmission is implicated in ASD is important because many of this condition’s sensory and motor symptoms are consistent with brainstem pathology. Therefore, the purpose of this review was to integrate epidemiological, behavioral, histological, neuroimaging, and animal evidence of brainstem contributions to ASD. Because ASD is a neurodevelopmental condition, we examined the available data through a lens of hierarchical brain development. The review of the literature suggests that developmental alterations of the brainstem could have potential cascading effects on cortical and cerebellar formation, ultimately leading to ASD symptoms. This view is supported by human epidemiology findings and data from animal models of ASD, showing that perturbed development of the brainstem substructures, particularly during the peak formation of the brainstem’s monoaminergic centers, may relate to ASD or ASD-like behaviors. Furthermore, we review evidence from human histology, psychophysiology, and neuroimaging suggesting that brainstem development and maturation may be atypical in ASD and may be related to key ASD symptoms, such as atypical sensorimotor features and social responsiveness. From this review there emerges the need of future research to validate early detection of the brainstem-based somatosensory and psychophysiological behaviors that emerge in infancy, and to examine the brainstem across the life span, while accounting for age. In all, there is preliminary evidence for brainstem involvement in ASD, but a better understanding of the brainstem’s role would likely pave the way for earlier diagnosis and treatment of ASD.
Collapse
Affiliation(s)
- Olga I Dadalko
- Motor and Brain Development Lab, Waisman Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Brittany G Travers
- Motor and Brain Development Lab, Occupational Therapy Program in the Department of Kinesiology, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
41
|
Saint-Martin M, Joubert B, Pellier-Monnin V, Pascual O, Noraz N, Honnorat J. Contactin-associated protein-like 2, a protein of the neurexin family involved in several human diseases. Eur J Neurosci 2018; 48:1906-1923. [PMID: 30028556 DOI: 10.1111/ejn.14081] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 06/08/2018] [Accepted: 07/02/2018] [Indexed: 12/11/2022]
Abstract
Contactin-associated protein-like 2 (CASPR2) is a cell adhesion protein of the neurexin family. Proteins of this family have been shown to play a role in the development of the nervous system, in synaptic functions, and in neurological diseases. Over recent years, CASPR2 function has gained an increasing interest as demonstrated by the growing number of publications. Here, we gather published data to comprehensively review CASPR2 functions within the nervous system in relation to CASPR2-related diseases in humans. On the one hand, studies on Cntnap2 (coding for CASPR2) knockout mice revealed its role during development, especially, in setting-up the inhibitory network. Consistent with this result, mutations in the CNTNAP2 gene coding for CASPR2 in human have been identified in neurodevelopmental disorders such as autism, intellectual disability, and epilepsy. On the other hand, CASPR2 was shown to play a role beyond development, in the localization of voltage-gated potassium channel (VGKC) complex that is composed of TAG-1, Kv1.1, and Kv1.2. This complex was found in several subcellular compartments essential for action potential propagation: the node of Ranvier, the axon initial segment, and the synapse. In line with a role of CASPR2 in the mature nervous system, neurological autoimmune diseases have been described in patients without neurodevelopmental disorders but with antibodies directed against CASPR2. These autoimmune diseases were of two types: central with memory disorders and temporal lobe seizures, or peripheral with muscular hyperactivity. Overall, we review the up-to-date knowledge on CASPR2 function and pinpoint confused or lacking information that will need further investigation.
Collapse
Affiliation(s)
- Margaux Saint-Martin
- Institut NeuroMyoGene INSERM U1217/CNRS UMR 5310, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Bastien Joubert
- Institut NeuroMyoGene INSERM U1217/CNRS UMR 5310, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France.,French Reference Center on Paraneoplastic Neurological Syndrome, Hospices Civils de Lyon, Hôpital Neurologique, Bron, France
| | - Véronique Pellier-Monnin
- Institut NeuroMyoGene INSERM U1217/CNRS UMR 5310, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Olivier Pascual
- Institut NeuroMyoGene INSERM U1217/CNRS UMR 5310, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Nelly Noraz
- Institut NeuroMyoGene INSERM U1217/CNRS UMR 5310, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Jérôme Honnorat
- Institut NeuroMyoGene INSERM U1217/CNRS UMR 5310, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France.,French Reference Center on Paraneoplastic Neurological Syndrome, Hospices Civils de Lyon, Hôpital Neurologique, Bron, France
| |
Collapse
|
42
|
Mader S, Brimberg L, Soltys JN, Bennett JL, Diamond B. Mutations of Recombinant Aquaporin-4 Antibody in the Fc Domain Can Impair Complement-Dependent Cellular Cytotoxicity and Transplacental Transport. Front Immunol 2018; 9:1599. [PMID: 30057582 PMCID: PMC6053506 DOI: 10.3389/fimmu.2018.01599] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 06/27/2018] [Indexed: 11/13/2022] Open
Abstract
Maternal antibodies provide protection for the developing fetus. Transplacental transport of pathogenic autoantibodies might pose a risk for the developing fetus. The transport of antibodies across the placenta to the fetal circulation occurs through the neonatal Fc salvage receptor (FcRn). During gestation, maternal autoantibodies are able to penetrate the embryonic brain before a functional intact blood-brain barrier is established. Brain-reactive antibodies to the water channel protein aquaporin-4 (AQP4) are a hallmark finding in neuromyelitis optica (NMO), a neurological disease that predominantly affects women, many of whom are of childbearing age. AQP4-IgG mediate astrocytic injury in a complement-dependent fashion. Recent studies suggest these antibodies contribute to impaired pregnancy outcome. The aim of the study was to investigate the transplacental transport as well as FcRn binding of a monoclonal AQP4-IgG cloned from an NMO patient (wild-type antibody) compared to five different mutated Fc domain of this antibody containing single amino acid substitutions in the Fc region. All of the Fc-mutated antibodies lack complement-dependent cytotoxicity. Four of the five Fc-mutated antibodies showed limited transplacental transport in vivo. Three mutated Fc with impaired transplacental transport showed persistent binding to rodent FcRn at pH 6 but also at pH 7.2, suggesting that limited transplacental transport could be due to diminished release from FcRn. One mutated Fc with modestly limited transplacental transport showed diminished binding to FcRn at pH 6. This study suggests that mutated Fc with intact transplacental transport may be used to study antibody effector functions and Fc with limited transport may be used as a carrier to deliver therapies to pregnant woman, while sparing the developing fetus.
Collapse
Affiliation(s)
- Simone Mader
- The Feinstein Institute for Medical Research, The Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases, Northwell Health System, Manhasset, NY, United States
| | - Lior Brimberg
- The Feinstein Institute for Medical Research, The Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases, Northwell Health System, Manhasset, NY, United States
| | - John N Soltys
- Medical Scientist Training and Neuroscience Graduate Training Programs, University of Colorado Denver School of Medicine, Aurora, IL, United States
| | - Jeffrey L Bennett
- Department of Neurology, Program in Neuroscience, University of Colorado Denver School of Medicine, Aurora, IL, United States.,Department of Ophthalmology, Program in Neuroscience, University of Colorado Denver School of Medicine, Aurora, IL, United States
| | - Betty Diamond
- The Feinstein Institute for Medical Research, The Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases, Northwell Health System, Manhasset, NY, United States
| |
Collapse
|
43
|
Larroya-García A, Navas-Carrillo D, Orenes-Piñero E. Impact of gut microbiota on neurological diseases: Diet composition and novel treatments. Crit Rev Food Sci Nutr 2018; 59:3102-3116. [PMID: 29870270 DOI: 10.1080/10408398.2018.1484340] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Gut microbiota has significant effects on the structure and function of the enteric and central nervous system including human behaviour and brain regulation. Herein, we analyze the role of this intestinal ecosystem, the effects of dietary changes and the administration of nutritional supplements, such as probiotics, prebiotics, or fecal transplantation in neuropsychiatric disorders. Numerous factors have been highlighted to influence gut microbiota composition, including genetics, health status, mode of birth delivery and environment. However, diet composition and nutritional status has been repeatedly shown to be one of the most critical modifiable factors of this ecosystem. A comprehensively analysis of the microbiome-intestine-brain axis has been performed, including the impact of intestinal bacteria in alterations in the nervous, immune and endocrine systems and their metabolites. Finally, we discuss the latest literature examining the effects of diet composition, nutritional status and microbiota alterations in several neuropsychiatric disorders, such as autism, anxiety, depression, Alzheimer's disease and anorexia nervosa.
Collapse
Affiliation(s)
- Ana Larroya-García
- Department of Biochemistry and Molecular Biology-A, University of Murcia, Murcia, Spain
| | - Diana Navas-Carrillo
- Department of Surgery, Hospital de la Vega Lorenzo Guirao, University of Murcia, Murcia, Spain
| | - Esteban Orenes-Piñero
- Department of Biochemistry and Molecular Biology-A, University of Murcia, Murcia, Spain
| |
Collapse
|
44
|
Edmiston E, Jones KL, Vu T, Ashwood P, Van de Water J. Identification of the antigenic epitopes of maternal autoantibodies in autism spectrum disorders. Brain Behav Immun 2018; 69:399-407. [PMID: 29289663 PMCID: PMC5857423 DOI: 10.1016/j.bbi.2017.12.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 12/07/2017] [Accepted: 12/27/2017] [Indexed: 10/18/2022] Open
Abstract
Several groups have described the presence of fetal brain-reactive maternal autoantibodies in the plasma of some mothers whose children have autism spectrum disorder (ASD). We previously identified seven autoantigens targeted by these maternal autoantibodies, each of which is expressed at significant levels in the developing brain and has demonstrated roles in typical neurodevelopment. To further understand the binding repertoire of the maternal autoantibodies, as well as the presence of any meaningful differences with respect to the recognition and binding of these ASD-specific autoantibodies to each of these neuronal autoantigens, we utilized overlapping peptide microarrays incubated with maternal plasma samples obtained from the Childhood Autism Risk from Genetics and Environment (CHARGE) Study. In an effort to identify the most commonly recognized (immunodominant) epitope sequences targeted by maternal autoantibodies for each of the seven ASD-specific autoantigens, arrays were screened with plasma from mothers with children across diagnostic groups (ASD and typically developing (TD)) that were positive for at least one antigen by western blot (N = 67) or negative control mothers unreactive to any of the autoantigens (N = 18). Of the 63 peptides identified with the discovery microarrays, at least one immunodominant peptide was successfully identified for each of the seven antigenic proteins using subsequent selective screening microarrays. Furthermore, while limited by our relatively small sample size, there were peptides that were distinctly recognized by autoantibodies relative to diagnosis For example, reactivity was observed exclusively in mothers of children of ASD towards several peptides, including the LDH-B peptides DCIIIVVSNPVDILT (9.1% ASD vs. 0% TD; odds ratio (95% CI) = 6.644 (0.355-124.384)) and PVAEEEATVPNNKIT (5.5% ASD vs. 0% TD; odds ratio (95% CI) = 4.067 (0.203-81.403)).These results suggest that there are differences in the binding repertoire between the antigen positive ASD and TD maternal samples. Further, the autoantibodies in plasma from mothers of children with ASD bound to a more diverse set of peptides, and there were specific peptide binding combinations observed only in this group. Future studies are underway to determine the critical amino acids necessary for autoantibody binding, which will be essential in developing a potential therapeutic strategy for maternal autoantibody related (MAR) ASD.
Collapse
Affiliation(s)
- Elizabeth Edmiston
- Department of Internal Medicine, Division of Rheumatology, Allergy and Clinical Immunology, One Shields Avenue, University of California, Davis, CA 95616, USA; UC Davis MIND Institute, 2825 50th St, Sacramento, CA 95817, USA.
| | - Karen L Jones
- Department of Internal Medicine, Division of Rheumatology, Allergy and Clinical Immunology, One Shields Avenue, University of California, Davis, CA 95616, USA; UC Davis MIND Institute, 2825 50th St, Sacramento, CA 95817, USA.
| | - Tam Vu
- Department of Internal Medicine, Division of Rheumatology, Allergy and Clinical Immunology, One Shields Avenue, University of California, Davis, CA 95616, USA.
| | - Paul Ashwood
- Department of Internal Medicine, Division of Rheumatology, Allergy and Clinical Immunology, One Shields Avenue, University of California, Davis, CA 95616, USA; Department of Medical Microbiology and Immunology, One Shields Avenue, University of California, Davis, CA 95616, USA.
| | - Judy Van de Water
- Department of Internal Medicine, Division of Rheumatology, Allergy and Clinical Immunology, One Shields Avenue, University of California, Davis, CA 95616, USA; UC Davis MIND Institute, 2825 50th St, Sacramento, CA 95817, USA.
| |
Collapse
|
45
|
Gréa H, Scheid I, Gaman A, Rogemond V, Gillet S, Honnorat J, Bolognani F, Czech C, Bouquet C, Toledano E, Bouvard M, Delorme R, Groc L, Leboyer M. Clinical and autoimmune features of a patient with autism spectrum disorder seropositive for anti-NMDA-receptor autoantibody. DIALOGUES IN CLINICAL NEUROSCIENCE 2017. [PMID: 28566948 PMCID: PMC5442365 DOI: 10.31887/dcns.2017.19.1/mleboyer] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by dysfunctions in social interactions resulting from a complex interplay between immunogenetic and environmental risk factors. Autoimmunity has been proposed as a major etiological component of ASD. Whether specific autoantibodies directed against brain targets are involved in ASD remains an open question. Here, we identified within a cohort an ASD patient with multiple circulating autoantibodies, including the well-characterized one against glutamate NMDA receptor (NMDAR-Ab). The patient exhibited alexithymia and previously suffered from two major depressive episodes without psychotic symptoms. Using a single molecule-based imaging approach, we demonstrate that neither NMDAR-Ab type G immunoglobulin purified from the ASD patient serum, nor that from a seropositive healthy subject, disorganize membrane NMDAR complexes at synapses. These findings suggest that the autistic patient NMDAR-Abs do not play a direct role in the etiology of ASD and that other autoantibodies directed against neuronal targets should be investigated.
Collapse
Affiliation(s)
- Hélène Gréa
- University of Bordeaux, Interdisciplinary Institute for Neuroscience (HNS), Mixed Research Unit (UMR) 5297, Bordeaux, France; National Center for Scientific Research (CNRS), IINS UMR 5297, Bordeaux, France (Equal contribution)
| | - Isabelle Scheid
- University Paris Est Créteil, Psychiatry department, University Hospital Henri Mondor, Public Hospitals of Paris (AP-HP), University Hospital Department of Personalized Neurology and Psychiatry (DHU PePSY), France; Translational Psychiatry Laboratory, National Institute of Health and Medical Research (Inserm) U955, France; FondaMental Foundation, France
| | - Alexandru Gaman
- University Paris Est Créteil, Psychiatry department, University Hospital Henri Mondor, Public Hospitals of Paris (AP-HP), University Hospital Department of Personalized Neurology and Psychiatry (DHU PePSY), France; Translational Psychiatry Laboratory, National Institute of Health and Medical Research (Inserm) U955, France; FondaMental Foundation, France
| | - Véronique Rogemond
- Institut NeuroMyoGene Inserm U1217/CNRS UMR 5310, Lyon, France; Lyon Civil Hospitals, Neurological Hospital, Bron, France; University of Lyon - University Claude Bernard Lyon 1, Lyon, France
| | - Sandy Gillet
- Institut NeuroMyoGene Inserm U1217/CNRS UMR 5310, Lyon, France; Lyon Civil Hospitals, Neurological Hospital, Bron, France; University of Lyon - University Claude Bernard Lyon 1, Lyon, France
| | - Jérôme Honnorat
- Institut NeuroMyoGene Inserm U1217/CNRS UMR 5310, Lyon, France; Lyon Civil Hospitals, Neurological Hospital, Bron, France; University of Lyon - University Claude Bernard Lyon 1, Lyon, France
| | - Federico Bolognani
- Roche Institute, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Christian Czech
- Roche Institute, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Céline Bouquet
- Roche Institute, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | | | - Manuel Bouvard
- Aquitane Autism Resource Center, Charles Perrens Hospital, Bordeaux
| | | | - Laurent Groc
- University of Bordeaux, Interdisciplinary Institute for Neuroscience (IINS), Mixed Research Unit (UMR) 5297, Bordeaux, France; National Center for Scientific Research (CNRS), IINS UMR 5297, Bordeaux, France; FondaMental Foundation, France (Equal seniority)
| | - Marion Leboyer
- University Paris Est Créteil, Psychiatry department, University Hospital Henri Mondor, Public Hospitals of Paris (AP-HP), University Hospital Department of Personalized Neurology and Psychiatry (DHU PePSY), France; Translational Psychiatry Laboratory, National Institute of Health and Medical Research (Inserm) U955, France; FondaMental Foundation, France
| |
Collapse
|
46
|
Affiliation(s)
- Harvey S Singer
- Professor of Neurology and Pediatric, Johns Hopkins University School of Medicine, Rubenstein Child Health Building, Baltimore, Maryland.
| |
Collapse
|
47
|
Bilbo SD, Block CL, Bolton JL, Hanamsagar R, Tran PK. Beyond infection - Maternal immune activation by environmental factors, microglial development, and relevance for autism spectrum disorders. Exp Neurol 2017; 299:241-251. [PMID: 28698032 DOI: 10.1016/j.expneurol.2017.07.002] [Citation(s) in RCA: 207] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 06/26/2017] [Accepted: 07/05/2017] [Indexed: 12/17/2022]
Abstract
Immune molecules such as cytokines and chemokines and the cells that produce them within the brain, notably microglia, are critical for normal brain development. This recognition has in recent years led to the working hypothesis that inflammatory events during pregnancy, e.g. in response to infection, may disrupt the normal expression of immune molecules during critical stages of neural development and thereby contribute to the risk for neurodevelopmental disorders such as autism spectrum disorder (ASD). This hypothesis has in large part been shepherded by the work of Dr. Paul Patterson and colleagues, which has elegantly demonstrated that a single viral infection or injection of a viral mimetic to pregnant mice significantly and persistently impacts offspring immune and nervous system function, changes that underlie ASD-like behavioral dysfunction including social and communication deficits. Subsequent studies by many labs - in humans and in non-human animal models - have supported the hypothesis that ongoing disrupted immune molecule expression and/or neuroinflammation contributes to at least a significant subset of ASD. The heterogeneous clinical and biological phenotypes observed in ASD strongly suggest that in genetically susceptible individuals, environmental risk factors combine or synergize to create a tipping or threshold point for dysfunction. Importantly, animal studies showing a link between maternal immune activation (MIA) and ASD-like outcomes in offspring involve different species and diverse environmental factors associated with ASD in humans, beyond infection, including toxin exposures, maternal stress, and maternal obesity, all of which impact inflammatory or immune pathways. The goal of this review is to highlight the broader implications of Dr. Patterson's work for the field of autism, with a focus on the impact that MIA by diverse environmental factors has on fetal brain development, immune system development, and the pathophysiology of ASD.
Collapse
Affiliation(s)
- Staci D Bilbo
- Pediatrics and Neuroscience, Harvard Medical School, Lurie Center for Autism, Massachusetts General Hospital for Children, Boston, MA 02126, United States.
| | - Carina L Block
- Psychology and Neuroscience, Duke University, Durham, NC 27708, United States
| | - Jessica L Bolton
- Pediatrics and Anatomy/Neurobiology, University of California-Irvine, Irvine, CA 92697, United States
| | - Richa Hanamsagar
- Pediatrics and Neuroscience, Harvard Medical School, Lurie Center for Autism, Massachusetts General Hospital for Children, Boston, MA 02126, United States
| | - Phuong K Tran
- Pediatrics and Neuroscience, Harvard Medical School, Lurie Center for Autism, Massachusetts General Hospital for Children, Boston, MA 02126, United States
| |
Collapse
|
48
|
Vinet É, Bernatsky S. Outcomes in Children Born to Women with Rheumatic Diseases. Rheum Dis Clin North Am 2017; 43:263-273. [DOI: 10.1016/j.rdc.2016.12.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
49
|
Lyall K, Croen L, Daniels J, Fallin MD, Ladd-Acosta C, Lee BK, Park BY, Snyder NW, Schendel D, Volk H, Windham GC, Newschaffer C. The Changing Epidemiology of Autism Spectrum Disorders. Annu Rev Public Health 2017; 38:81-102. [PMID: 28068486 PMCID: PMC6566093 DOI: 10.1146/annurev-publhealth-031816-044318] [Citation(s) in RCA: 589] [Impact Index Per Article: 73.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental condition with lifelong impacts. Genetic and environmental factors contribute to ASD etiology, which remains incompletely understood. Research on ASD epidemiology has made significant advances in the past decade. Current prevalence is estimated to be at least 1.5% in developed countries, with recent increases primarily among those without comorbid intellectual disability. Genetic studies have identified a number of rare de novo mutations and gained footing in the areas of polygenic risk, epigenetics, and gene-by-environment interaction. Epidemiologic investigations focused on nongenetic factors have established advanced parental age and preterm birth as ASD risk factors, indicated that prenatal exposure to air pollution and short interpregnancy interval are potential risk factors, and suggested the need for further exploration of certain prenatal nutrients, metabolic conditions, and exposure to endocrine-disrupting chemicals. We discuss future challenges and goals for ASD epidemiology as well as public health implications.
Collapse
Affiliation(s)
- Kristen Lyall
- A.J. Drexel Autism Institute, Philadelphia, Pennsylvania 19104;
| | - Lisa Croen
- Kaiser Permanente Division of Research, Oakland, California 94612
| | - Julie Daniels
- Department of Epidemiology, University of North Carolina Gillings School of Public Health, Chapel Hill, North Carolina 27599
| | - M Daniele Fallin
- Wendy Klag Center for Autism and Developmental Disabilities, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland 21205
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland 21205
| | - Christine Ladd-Acosta
- Wendy Klag Center for Autism and Developmental Disabilities, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland 21205
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland 21205
| | - Brian K Lee
- Department of Epidemiology and Biostatistics, Drexel University School of Public Health, Philadelphia, Pennsylvania 19104
- Department of Medical Epidemiology and Biostatistics and Department of Public Health Sciences, Karolinska Institute, SE 171-77 Stockholm, Sweden
| | - Bo Y Park
- Wendy Klag Center for Autism and Developmental Disabilities, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland 21205
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland 21205
| | | | - Diana Schendel
- Department of Economics and Business, National Centre for Register-Based Research, Aarhus University, DK-8210 Aarhus, Denmark
- Department of Public Health, Section for Epidemiology, Aarhus University, DK-8000 Aarhus, Denmark
- Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
| | - Heather Volk
- Wendy Klag Center for Autism and Developmental Disabilities, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland 21205
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland 21205
| | - Gayle C Windham
- California Department of Public Health, Division of Environmental and Occupational Disease Control, Richmond, California 94805
| | | |
Collapse
|
50
|
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: 100] [Impact Index Per Article: 12.5] [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
|