101
|
Do Autism Spectrum and Autoimmune Disorders Share Predisposition Gene Signature Due to mTOR Signaling Pathway Controlling Expression? Int J Mol Sci 2021; 22:ijms22105248. [PMID: 34065644 PMCID: PMC8156237 DOI: 10.3390/ijms22105248] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/08/2021] [Accepted: 05/14/2021] [Indexed: 12/22/2022] Open
Abstract
Autism spectrum disorder (ASD) is characterized by uncommon genetic heterogeneity and a high heritability concurrently. Most autoimmune disorders (AID), similarly to ASD, are characterized by impressive genetic heterogeneity and heritability. We conducted gene-set analyses and revealed that 584 out of 992 genes (59%) included in a new release of the SFARI Gene database and 439 out of 871 AID-associated genes (50%) could be attributed to one of four groups: 1. FMRP (fragile X mental retardation protein) target genes, 2. mTOR signaling network genes, 3. mTOR-modulated genes, and 4. vitamin D3-sensitive genes. With the exception of FMRP targets, which are obviously associated with the direct involvement of local translation disturbance in the pathological mechanisms of ASD, the remaining categories are represented among AID genes in a very similar percentage as among ASD predisposition genes. Thus, mTOR signaling pathway genes make up 4% of ASD and 3% of AID genes, mTOR-modulated genes-31% of both ASD and AID genes, and vitamin D-sensitive genes-20% of ASD and 23% of AID genes. The network analysis revealed 3124 interactions between 528 out of 729 AID genes for the 0.7 cutoff, so the great majority (up to 67%) of AID genes are related to the mTOR signaling pathway directly or indirectly. Our present research and available published data allow us to hypothesize that both a certain part of ASD and AID comprise a connected set of disorders sharing a common aberrant pathway (mTOR signaling) rather than a vast set of different disorders. Furthermore, an immune subtype of the autism spectrum might be a specific type of autoimmune disorder with an early manifestation of a unique set of predominantly behavioral symptoms.
Collapse
|
102
|
Liang Y, Yu H, Ke X, Eyles D, Sun R, Wang Z, Huang S, Lin L, McGrath JJ, Lu J, Guo X, Yao P. Vitamin D deficiency worsens maternal diabetes induced neurodevelopmental disorder by potentiating hyperglycemia-mediated epigenetic changes. Ann N Y Acad Sci 2021; 1491:74-88. [PMID: 33305416 DOI: 10.1111/nyas.14535] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 10/15/2020] [Accepted: 10/27/2020] [Indexed: 12/22/2022]
Abstract
Many studies have shown that vitamin D (VD) deficiency may be a risk factor for neurodevelopmental disorders, such as autism spectrum disorders (ASDs) and schizophrenia, although causative mechanisms remain unknown. In this study, we investigated the potential role and effect of VD on maternal diabetes induced autism-related phenotypes. The in vitro study found that enhancing genomic VD signaling by overexpressing the VD receptor (VDR) in human neural progenitor cells ACS-5003 protects against hyperglycemia-induced oxidative stress and inflammation by activating Nrf2 and its target genes, including SOD2 and HMOX1, and accordingly, VDR gene knockdown worsens the problem. In the two in vivo models we explored, maternal diabetes was used to establish an animal model of relevance to ASD, and mice lacking 25-hydroxyvitamin D 1-alpha-hydroxylase (the rate-limiting enzyme in the synthesis of 1,25(OH)2D3) were used to develop a model of VD deficiency (VDD). We show that although prenatal VDD itself does not produce ASD-relevant phenotypes, it significantly potentiates maternal diabetes induced epigenetic modifications and autism-related phenotypes. Postnatal manipulation of VD has no effect on maternal diabetes induced autism-related phenotypes. We conclude that VDD potentiates maternal diabetes induced autism-related phenotypes in offspring by epigenetic mechanisms. This study adds to other preclinical studies linking prenatal VDD with a neurodevelopmental disorder.
Collapse
Affiliation(s)
- Yujie Liang
- Department of Child Psychiatry, Kangning Hospital of Shenzhen, Shenzhen, P.R. China
| | - Hong Yu
- Department of Pediatrics, Foshan University, Foshan Maternity and Child Health Care Hospital, Foshan, P.R. China
| | - Xiaoyin Ke
- Department of Child Psychiatry, Kangning Hospital of Shenzhen, Shenzhen, P.R. China
| | - Darryl Eyles
- Queensland Centre for Mental Health Research, the Park Centre for Mental Health, Brisbane, Queensland, Australia
- Queensland Brain Institute, University of Queensland, St Lucia, Queensland, Australia
| | - Ruoyu Sun
- Department of Pediatrics, Foshan University, Foshan Maternity and Child Health Care Hospital, Foshan, P.R. China
| | - Zichen Wang
- Department of Child Psychiatry, Kangning Hospital of Shenzhen, Shenzhen, P.R. China
| | - Saijun Huang
- Department of Pediatrics, Foshan University, Foshan Maternity and Child Health Care Hospital, Foshan, P.R. China
| | - Ling Lin
- Department of Child Psychiatry, Kangning Hospital of Shenzhen, Shenzhen, P.R. China
| | - John J McGrath
- Queensland Centre for Mental Health Research, the Park Centre for Mental Health, Brisbane, Queensland, Australia
- Queensland Brain Institute, University of Queensland, St Lucia, Queensland, Australia
- National Centre for Register-based Research, Department of Economics and Business Economics, Aarhus University, Aarhus, Denmark
| | - Jianping Lu
- Department of Child Psychiatry, Kangning Hospital of Shenzhen, Shenzhen, P.R. China
| | - Xiaoling Guo
- Department of Pediatrics, Foshan University, Foshan Maternity and Child Health Care Hospital, Foshan, P.R. China
| | - Paul Yao
- Department of Child Psychiatry, Kangning Hospital of Shenzhen, Shenzhen, P.R. China
- Department of Pediatrics, Foshan University, Foshan Maternity and Child Health Care Hospital, Foshan, P.R. China
| |
Collapse
|
103
|
Jangjoo M, Goodman SJ, Choufani S, Trost B, Scherer SW, Kelley E, Ayub M, Nicolson R, Georgiades S, Crosbie J, Schachar R, Anagnostou E, Grunebaum E, Weksberg R. An Epigenetically Distinct Subset of Children With Autism Spectrum Disorder Resulting From Differences in Blood Cell Composition. Front Neurol 2021; 12:612817. [PMID: 33935932 PMCID: PMC8085304 DOI: 10.3389/fneur.2021.612817] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 03/15/2021] [Indexed: 12/23/2022] Open
Abstract
Background: Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder that often involves impaired cognition, communication difficulties and restrictive, repetitive behaviors. ASD is extremely heterogeneous both clinically and etiologically, which represents one of the greatest challenges in studying the molecular underpinnings of ASD. While hundreds of ASD-associated genes have been identified that confer varying degrees of risk, no single gene variant accounts for >1% of ASD cases. Notably, a large number of ASD-risk genes function as epigenetic regulators, indicating potential epigenetic dysregulation in ASD. As such, we compared genome-wide DNA methylation (DNAm) in the blood of children with ASD (n = 265) to samples from age- and sex-matched, neurotypical controls (n = 122) using the Illumina Infinium HumanMethylation450 arrays. Results: While DNAm patterns did not distinctly separate ASD cases from controls, our analysis identified an epigenetically unique subset of ASD cases (n = 32); these individuals exhibited significant differential methylation from both controls than the remaining ASD cases. The CpG sites at which this subset was differentially methylated mapped to known ASD risk genes that encode proteins of the nervous and immune systems. Moreover, the observed DNAm differences were attributable to altered blood cell composition, i.e., lower granulocyte proportion and granulocyte-to-lymphocyte ratio in the ASD subset, as compared to the remaining ASD cases and controls. This ASD subset did not differ from the rest of the ASD cases in the frequency or type of high-risk genomic variants. Conclusion: Within our ASD cohort, we identified a subset of individuals that exhibit differential methylation from both controls and the remaining ASD group tightly associated with shifts in immune cell type proportions. This is an important feature that should be assessed in all epigenetic studies of blood cells in ASD. This finding also builds on past reports of changes in the immune systems of children with ASD, supporting the potential role of altered immunological mechanisms in the complex pathophysiology of ASD. The discovery of significant molecular and immunological features in subgroups of individuals with ASD may allow clinicians to better stratify patients, facilitating personalized interventions and improved outcomes.
Collapse
Affiliation(s)
- Maryam Jangjoo
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Sarah J. Goodman
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Sanaa Choufani
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Brett Trost
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON, Canada
| | - Stephen W. Scherer
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- McLaughlin Centre, University of Toronto, Toronto, ON, Canada
| | - Elizabeth Kelley
- Department of Psychiatry, Queen's University, Kingston, ON, Canada
| | - Muhammad Ayub
- Department of Psychiatry, Queen's University, Kingston, ON, Canada
| | - Rob Nicolson
- Department of Psychiatry, University of Western Ontario, London, ON, Canada
| | - Stelios Georgiades
- Department of Psychiatry and Behavioural Neurosciences, Offord Centre for Child Studies, McMaster University, Hamilton, ON, Canada
| | - Jennifer Crosbie
- Neurosciences and Mental Health Program, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Russell Schachar
- Neurosciences and Mental Health Program, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Institute of Medical Science, School of Graduate Studies, University of Toronto, Toronto, ON, Canada
| | - Evdokia Anagnostou
- Holland Bloorview Kids Rehabilitation Hospital, Toronto, ON, Canada
- Department of Pediatrics, University of Toronto, Toronto, ON, Canada
| | - Eyal Grunebaum
- Institute of Medical Science, School of Graduate Studies, University of Toronto, Toronto, ON, Canada
- Division of Immunology and Allergy, The Hospital for Sick Children, Toronto, ON, Canada
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Rosanna Weksberg
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Institute of Medical Science, School of Graduate Studies, University of Toronto, Toronto, ON, Canada
- Department of Pediatrics, University of Toronto, Toronto, ON, Canada
- Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, Toronto, ON, Canada
| |
Collapse
|
104
|
Abstract
The term "Autism spectrum disorder" (ASD), in vogue at present, has evolved after continual substantial developments taking place over more than a century. ASD is a heterogeneous, multi-factorial, developmental disability in which an unusual pattern of development takes place during infant and toddler years. As per DSM-5, Autism spectrum disorder is a combined phrase for a family of complex developmental disabilities inclusive of "Autistic Disorder, Pervasive Developmental Disorder not Otherwise Specified (PDD-NOS), and Asperger's Disorder". "ASD is characterized not only by persistent impairments in reciprocal social communication and social interactions, but is also manifested by restricted, repetitive patterns of behavior, interests, or activities". The classical clinical signs that exist in two major domains, viz. the 'social domain' and the 'behavioral domain' for the precise diagnosis of ASD have been tabulated and major differences between DSM-5 and DSM-4 are depicted with the help of a figure in this basic review article. A sharp rise in the incidence of ASD cases has been observed worldwide owing to various risk factors such as genetic predisposition coupled with adverse environmental conditions, gynecological interventions, etc. Two official manuals viz. the "Diagnostic and Statistical Manual of Mental Disorders" (DSM) (published by the American Psychiatric Association), and the "International Classification of Diseases" (ICD) (published by the World Health Organization) is being regularly updated to facilitate diagnosis of ASD. ICD-11 guidelines being prospectively implemented with effect from January 2022 have attracted global attention.
Collapse
|
105
|
A pro-inflammatory phenotype is associated with behavioural traits in children with Prader-Willi syndrome. Eur Child Adolesc Psychiatry 2021; 30:899-908. [PMID: 32495042 PMCID: PMC8140962 DOI: 10.1007/s00787-020-01568-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Accepted: 05/27/2020] [Indexed: 12/16/2022]
Abstract
Several lines of evidence indicate that immune-inflammatory alterations are widely observed in various mental disorders. Genetic syndromes with high risk of psychiatric disorders may constitute a model for studies investigating this phenomenon. One of such genetically determined neurodevelopmental disorders is the Prader-Willi syndrome (PWS). Therefore, we aimed to profile a broad panel of immune-inflammatory markers in patients with PWS, taking into account co-morbid psychopathology. Participants were 20 children with PWS, and 20 healthy children matched for age, sex and body mass index. Behavioural symptoms and co-occurring psychopathological symptoms were assessed using the Child Behaviour Checklist (CBCL). We found significantly elevated levels of interleukin (IL)-1β and IL-13 in patients with PWS. There were significant positive correlations between the levels of IL-1β and scores of the following externalizing and internalizing CBCL domains: withdrawn/depressed, social problems, thought problems, attention problems, delinquent and aggressive behaviour in PWS children. Moreover, higher levels of IL-13 were associated with more severe psychopathology in terms of social and attention problems as well as delinquent and aggressive behaviour. Our findings imply that subclinical inflammation, observed as elevated IL-1β and IL-13 levels, appears only in PWS patients and is correlated to several psychopathological symptoms.
Collapse
|
106
|
Sarovic D. A Unifying Theory for Autism: The Pathogenetic Triad as a Theoretical Framework. Front Psychiatry 2021; 12:767075. [PMID: 34867553 PMCID: PMC8637925 DOI: 10.3389/fpsyt.2021.767075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/27/2021] [Indexed: 12/27/2022] Open
Abstract
This paper presents a unifying theory for autism by applying the framework of a pathogenetic triad to the scientific literature. It proposes a deconstruction of autism into three contributing features (an autistic personality dimension, cognitive compensation, and neuropathological risk factors), and delineates how they interact to cause a maladaptive behavioral phenotype that may require a clinical diagnosis. The autistic personality represents a common core condition, which induces a set of behavioral issues when pronounced. These issues are compensated for by cognitive mechanisms, allowing the individual to remain adaptive and functional. Risk factors, both exogenous and endogenous ones, show pathophysiological convergence through their negative effects on neurodevelopment. This secondarily affects cognitive compensation, which disinhibits a maladaptive behavioral phenotype. The triad is operationalized and methods for quantification are presented. With respect to the breadth of findings in the literature that it can incorporate, it is the most comprehensive model yet for autism. Its main implications are that (1) it presents the broader autism phenotype as a non-pathological core personality domain, which is shared across the population and uncoupled from associated features such as low cognitive ability and immune dysfunction, (2) it proposes that common genetic variants underly the personality domain, and that rare variants act as risk factors through negative effects on neurodevelopment, (3) it outlines a common pathophysiological mechanism, through inhibition of neurodevelopment and cognitive dysfunction, by which a wide range of endogenous and exogenous risk factors lead to autism, and (4) it suggests that contributing risk factors, and findings of immune and autonomic dysfunction are clinically ascertained rather than part of the core autism construct.
Collapse
Affiliation(s)
- Darko Sarovic
- Gillberg Neuropsychiatry Centre, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Radiology, Sahlgrenska University Hospital, Gothenburg, Sweden.,MedTech West, Gothenburg, Sweden
| |
Collapse
|
107
|
Behl S, Mehta S, Pandey MK. Abnormal Levels of Metal Micronutrients and Autism Spectrum Disorder: A Perspective Review. Front Mol Neurosci 2020; 13:586209. [PMID: 33362464 PMCID: PMC7759187 DOI: 10.3389/fnmol.2020.586209] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 10/20/2020] [Indexed: 12/16/2022] Open
Abstract
The aim of the present review is to summarize the prevalence of abnormal levels of various metal micronutrients including copper (Cu), iron (Fe), magnesium (Mg), zinc (Zn), and selenium (Se) in Autism Spectrum Disorder (ASD) using hair, nail and serum samples. A correlation of selected abnormal metal ions with known neurodevelopmental processes using Gene Ontology (GO) term was also conducted. Data included in this review are derived from ASD clinical studies performed globally. Metal ion disparity data is also analyzed and discussed based on gender (Male/Female) to establish any gender dependent correlation. Finally, a rational perspective and possible path to better understand the role of metal micronutrients in ASD is suggested.
Collapse
Affiliation(s)
- Supriya Behl
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, United States
| | - Sunil Mehta
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, United States
| | - Mukesh K Pandey
- Department of Radiology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
108
|
Sotgiu S, Manca S, Gagliano A, Minutolo A, Melis MC, Pisuttu G, Scoppola C, Bolognesi E, Clerici M, Guerini FR, Carta A. Immune regulation of neurodevelopment at the mother-foetus interface: the case of autism. Clin Transl Immunology 2020; 9:e1211. [PMID: 33209302 PMCID: PMC7662086 DOI: 10.1002/cti2.1211] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/20/2020] [Accepted: 10/20/2020] [Indexed: 12/16/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder defined by deficits in social communication and stereotypical behaviours. ASD’s aetiology remains mostly unclear, because of a complex interaction between genetic and environmental factors. Recently, a strong consensus has developed around ASD’s immune‐mediated pathophysiology, which is the subject of this review. For many years, neuroimmunological studies tried to understand ASD as a prototypical antibody‐ or cell‐mediated disease. Other findings indicated the importance of autoimmune mechanisms such as familial and individual autoimmunity, adaptive immune abnormalities and the influence of infections during gestation. However, recent studies have challenged the idea that autism may be a classical autoimmune disease. Modern neurodevelopmental immunology shows the double‐edged nature of many immune effectors, which can be either beneficial or detrimental depending on tissue homeostasis, stressors, neurodevelopmental stage, inherited and de novo gene mutations and other variables. Nowadays, mother–child interactions in the prenatal environment appear to be crucial for the occurrence of ASD. Studies of animal maternal–foetal immune interaction are being fruitfully carried out using different combinations of type and timing of infection, of maternal immune response and foetal vulnerability and of resilience factors to hostile events. The derailed neuroimmune crosstalk through the placenta initiates and maintains a chronic foetal neuroglial activation, eventually causing the alteration of neurogenesis, migration, synapse formation and pruning. The importance of pregnancy can also allow early immune interventions, which can significantly reduce the increasing risk of ASD and its heavy social burden.
Collapse
Affiliation(s)
- Stefano Sotgiu
- Unit of Child Neuropsychiatry Department of Medical Surgical and Experimental Sciences University of Sassari Sassari Italy
| | - Salvatorica Manca
- Unità Operativa di Neuropsichiatria Infanzia e Adolescenza (UONPIA) ASSL Sassari Sassari Italy
| | - Antonella Gagliano
- Child & Adolescent Neuropsychiatry Unit Department of Biomedical Sciences University of Cagliari Cagliari Italy
| | - Alessandra Minutolo
- Child & Adolescent Neuropsychiatry Unit Department of Biomedical Sciences University of Cagliari Cagliari Italy
| | - Maria Clotilde Melis
- Unit of Child Neuropsychiatry Department of Medical Surgical and Experimental Sciences University of Sassari Sassari Italy
| | - Giulia Pisuttu
- Unit of Child Neuropsychiatry Department of Medical Surgical and Experimental Sciences University of Sassari Sassari Italy
| | - Chiara Scoppola
- Unit of Child Neuropsychiatry Department of Medical Surgical and Experimental Sciences University of Sassari Sassari Italy
| | | | - Mario Clerici
- IRCCS Fondazione Don Carlo Gnocchi - ONLUS Milan Italy.,Department of Pathophysiology and Transplantation University of Milano Milan Italy
| | | | - Alessandra Carta
- Unit of Child Neuropsychiatry Department of Medical Surgical and Experimental Sciences University of Sassari Sassari Italy
| |
Collapse
|
109
|
Bjørklund G, Pivina L, Dadar M, Meguid NA, Semenova Y, Anwar M, Chirumbolo S. Gastrointestinal alterations in autism spectrum disorder: What do we know? Neurosci Biobehav Rev 2020; 118:111-120. [DOI: 10.1016/j.neubiorev.2020.06.033] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/21/2020] [Accepted: 06/28/2020] [Indexed: 02/07/2023]
|
110
|
de Leão ERLP, de Souza DNC, de Moura LVB, da Silveira Júnior AM, Dos Santos ALG, Diniz DG, Diniz CWP, Sosthenes MCK. Lateral septum microglial changes and behavioral abnormalities of mice exposed to valproic acid during the prenatal period. J Chem Neuroanat 2020; 111:101875. [PMID: 33127448 DOI: 10.1016/j.jchemneu.2020.101875] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/08/2020] [Accepted: 10/20/2020] [Indexed: 12/14/2022]
Abstract
Most animal model studies of autism spectrum disorder (ASD) have been performed in males, which may be a reflex of the 3-times higher prevalence in boys than in girls. For this reason, little is known about the mechanisms underlying disease progression in females, and nothing is known about potential associations between microglial changes in the lateral septum (LS) and adult female cognition. Prenatal exposure to valproic acid (VPA) in mice has been widely used as an experimental model of autism-like behaviors associated with cellular changes. However, no study has reported the influence of VPA exposure in utero and its consequences on limbic system-dependent tasks or the microglial response in the LS in adult female mice. We compared the exploratory activity and risk assessment in novel environments of BALB/c control mice to mice exposed in utero to VPA and estimated the total number of microglia in the LS using an optical fractionator. On day 12.5 of pregnancy, females received diluted VPA or saline by gavage. After weaning, VPA exposed or control pups were separately housed in standard laboratory cages. At 5 months of age, all mice underwent behavioral testing and their brain sections were immunolabelled using IBA-1 antibody. In the open field test, VPA group showed a greater distance traveled, which was accompanied by less immobility, less time spent on the periphery and a greater number, crossed lines. Similar findings were found in the elevated plus maze test, where VPA mice traveled greater distances, immobility was significantly higher than that of control and VPA group spent less time on the closed arms of apparatus. Stereological analysis demonstrated higher microglial total number and density in the LS of VPA mice, as the cell count was greater, but the volume was similar. Therefore, we suggest that an increase in microglia in the LS may be part of the cellular changes associated with behavioral dysfunction in the VPA model of ASD.
Collapse
Affiliation(s)
- Ellen Rose Leandro Ponce de Leão
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário "João de Barros Barreto", Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Pará, Brazil
| | - Dilza Nazaré Colares de Souza
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário "João de Barros Barreto", Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Pará, Brazil
| | - Larissa Victória Barra de Moura
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário "João de Barros Barreto", Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Pará, Brazil
| | - Antonio Morais da Silveira Júnior
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário "João de Barros Barreto", Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Pará, Brazil
| | - Alinne Lorrany Gomes Dos Santos
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário "João de Barros Barreto", Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Pará, Brazil
| | - Daniel Guerreiro Diniz
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário "João de Barros Barreto", Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Pará, Brazil; Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Belém, Pará, Brazil
| | - Cristovam Wanderley Picanço Diniz
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário "João de Barros Barreto", Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Pará, Brazil
| | - Marcia Consentino Kronka Sosthenes
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário "João de Barros Barreto", Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Pará, Brazil.
| |
Collapse
|
111
|
Ciernia AV, Link VM, Careaga M, LaSalle JM, Ashwood P. Genetic variants drive altered epigenetic regulation of endotoxin response in BTBR macrophages. Brain Behav Immun 2020; 89:20-31. [PMID: 32454135 PMCID: PMC7572655 DOI: 10.1016/j.bbi.2020.05.058] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 05/06/2020] [Accepted: 05/21/2020] [Indexed: 01/09/2023] Open
Abstract
The BTBR T+Itpr3tf/J (BTBR) mouse has been used as a complex genetic model of Autism Spectrum Disorders (ASD). While the specific mechanisms underlying BTBR behavioral phenotypes are poorly understood, prior studies have implicated profound differences in innate immune system control of pro-inflammatory cytokines. Innate immune activation and elevated pro-inflammatory cytokines are also detected in blood of children with ASD. In this study, we examined how underlying BTBR genetic variants correspond to strain-specific changes in chromatin accessibility, resulting in a pro-inflammatory response specifically in BTBR bone marrow derived macrophages (BMDM). In response to repeated lipopolysaccharide (LPS) treatments, C57BL/6J (C57) BMDM exhibited intact endotoxin tolerance. In contrast, BTBR BMDM exhibited hyper-responsive expression of genes that were normally tolerized in C57. This failure in formation of endotoxin tolerance in BTBR was mirrored at the level of chromatin accessibility. Using ATAC-seq, we specifically identified promoter and enhancer regions with strain-specific differential chromatin accessibility both at baseline and in response to LPS. Regions with strain-specific differences in chromatin accessibility were significantly enriched for BTBR genetic variants, such that an average of 22% of the differential chromatin regions had at least one variant. Together, these results demonstrate that BTBR genetic variants contribute to altered chromatin responsiveness to endotoxin challenge resulting in hyper-responsive innate immunity in BTBR. These findings provide evidence for an interaction between complex genetic variants and differential epigenetic regulation of innate immune responses.
Collapse
Affiliation(s)
- Annie Vogel Ciernia
- Department Biochemistry and Molecular Biology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver V6T2A1, Canada.
| | - Verena M Link
- Metaorganism Immunity Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Milo Careaga
- Department of Medical Microbiology and Immunology, MIND Institute, Genome Center, University of California, Davis, CA 95616, USA
| | - Janine M LaSalle
- Department of Medical Microbiology and Immunology, MIND Institute, Genome Center, University of California, Davis, CA 95616, USA
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, MIND Institute, Genome Center, University of California, Davis, CA 95616, USA
| |
Collapse
|
112
|
Gagliano A, Galati C, Ingrassia M, Ciuffo M, Alquino MA, Tanca MG, Carucci S, Zuddas A, Grossi E. Pediatric Acute-Onset Neuropsychiatric Syndrome: A Data Mining Approach to a Very Specific Constellation of Clinical Variables. J Child Adolesc Psychopharmacol 2020; 30:495-511. [PMID: 32460516 DOI: 10.1089/cap.2019.0165] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Objectives: Pediatric acute onset neuropsychiatric syndrome (PANS) is a clinically heterogeneous disorder presenting with: unusually abrupt onset of obsessive compulsive disorder (OCD) or severe eating restrictions, with at least two concomitant cognitive, behavioral, or affective symptoms such as anxiety, obsessive-compulsive behavior, and irritability/depression. This study describes the clinical and laboratory variables of 39 children (13 female and 26 male) with a mean age at recruitment of 8.6 years (standard deviation 3.1). Methods: Using a mathematical approach based on Artificial Neural Networks, the putative associations between PANS working criteria, as defined at the NIH in July 2010 (Swedo et al. 2012), were explored by the Auto Contractive Map (Auto-CM) system, a mapping method able to compute the multidimensional association of strength of each variable with all other variables in predefined dataset. Results: The PANS symptoms were strictly linked to one another on the semantic connectivity map, shaping a central "diamond" encompassing anxiety, irritability/oppositional defiant disorder symptoms, obsessive-compulsive symptoms, behavioral regression, sensory motor abnormalities, school performance deterioration, sleep disturbances, and emotional lability/depression. The semantic connectivity map also showed the aggregation between PANS symptoms and laboratory and clinical variables. In particular, the emotional lability/depression resulted as a highly connected hub linked to autoimmune disease in pregnancy, allergic and atopic disorders, and low Natural Killer percentage. Also anxiety symptoms were shown to be strongly related with recurrent infectious disease remarking the possible role of infections as a risk factor for PANS. Conclusion: Our data mining approach shows a very specific constellation of symptoms having strong links to laboratory and clinical variables consistent with PANS feature.
Collapse
Affiliation(s)
- Antonella Gagliano
- Child & Adolescent Neuropsychiatry Unit, Department of Biomedical Sciences, University of Cagliari, & "G. Brotzu" Hospital Trust, Cagliari, Italy
- Funding: The authors received no specific funding
| | - Cecilia Galati
- Division of Child Neurology and Psychiatry, Department of Paediatrics, University of Messina, Messina, Italy
- Funding: The authors received no specific funding
| | - Massimo Ingrassia
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
- Funding: The authors received no specific funding
| | - Massimo Ciuffo
- Department of Cognitive Psychological Pedagogical Sciences and Cultural Studies, University of Messina, Messina, Italy
- Funding: The authors received no specific funding
| | - Maria Ausilia Alquino
- Division of Child Neurology and Psychiatry, Department of Paediatrics, University of Messina, Messina, Italy
- Funding: The authors received no specific funding
| | - Marcello G Tanca
- Child & Adolescent Neuropsychiatry Unit, Department of Biomedical Sciences, University of Cagliari, & "G. Brotzu" Hospital Trust, Cagliari, Italy
- Funding: The authors received no specific funding
| | - Sara Carucci
- Child & Adolescent Neuropsychiatry Unit, Department of Biomedical Sciences, University of Cagliari, & "G. Brotzu" Hospital Trust, Cagliari, Italy
- Funding: The authors received no specific funding
| | - Alessandro Zuddas
- Child & Adolescent Neuropsychiatry Unit, Department of Biomedical Sciences, University of Cagliari, & "G. Brotzu" Hospital Trust, Cagliari, Italy
- Funding: The authors received no specific funding
| | - Enzo Grossi
- Autism Research Unit, Villa Santa Maria Foundation, Tavernerio, Italy
- Funding: The authors received no specific funding
| |
Collapse
|
113
|
Nadeem A, Ahmad SF, Al-Harbi NO, Al-Ayadhi LY, Attia SM, Alasmari AF, As Sobeai HM, Bakheet SA. Ubiquitous plasticizer, Di-(2-ethylhexyl) phthalate enhances existing inflammatory profile in monocytes of children with autism. Toxicology 2020; 446:152597. [PMID: 32991955 DOI: 10.1016/j.tox.2020.152597] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 09/07/2020] [Accepted: 09/17/2020] [Indexed: 12/29/2022]
Abstract
Genetic as well as environmental factors are believed to play a significant role in the pathogenesis and progression of autism spectrum disorder (ASD). Phthalates are ubiquitous environmental contaminants as they are used plasticizers in several household/industrial products such as vinyl flooring, plastic toys, and cosmetic products. One of the plasticizers that is quite prevalent in these products is di-2-ethylhexyl phthalate (DEHP) which can cause human exposure via dermal/inhalation/ingestion routes. DEHP and its metabolites are associated with behavioral dysregulations and reported to be increased in systemic circulation of ASD children. DEHP is reported to cause upregulation of several inflammatory cytokines in different cells/tissues, however its role in inflammatory signaling of ASD monocytes has not been investigated earlier. Therefore, this study evaluated the effects of DEHP (at 5 μM final concentration for 24 h) on inflammatory profile (NFkB, STAT3, IL-6, TNF-α, IL-1β) in monocytes of ASD subjects and typically developing control (TDC) children. Our data show that DEHP upregulates NFkB/STAT3 expression which is associated with increased inflammatory profile in monocytes of ASD and TDC subjects, however its effect is much greater in magnitude in the former group. This was confirmed by utilization of NFkB inhibitor, PDTC and STAT3 inhibitor, Stattic which caused reduction in inflammatory cytokines from DEHP-treated monocytes in ASD group. In short, DEHP causes further elevation in inflammatory signaling in ASD monocytes which could be due to existing inflammation in this group. These data suggest that use of plasticizers such as DEHP should be minimized in order to avoid their potential effects on immune dysfunction associated with ASD.
Collapse
Affiliation(s)
- Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.
| | - Sheikh F Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Naif O Al-Harbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Laila Y Al-Ayadhi
- Autism Research and Treatment Center, AL-Amodi Autism Research Chair, Department of Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdullah F Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Homood M As Sobeai
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Saleh A Bakheet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
114
|
Petruzzelli MG, Marzulli L, Margari F, De Giacomo A, Gabellone A, Giannico OV, Margari L. Vitamin D Deficiency in Autism Spectrum Disorder: A Cross-Sectional Study. DISEASE MARKERS 2020; 2020:9292560. [PMID: 33014190 PMCID: PMC7520686 DOI: 10.1155/2020/9292560] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 05/06/2020] [Accepted: 09/10/2020] [Indexed: 12/13/2022]
Abstract
Vitamin D plays a role in central nervous system (CNS) development. Recent literature focused on vitamin D status in children and adolescents with autism spectrum disorder (ASD), but with inconsistent results. Our case-control study is aimed at evaluating serum 25-hydroxyl-vitamin D (25(OH)D) concentration in children with ASD (ASD group, n = 54) compared to children affected by other neurological and psychiatric disorders (non-ASD group, n = 36). All patients were admitted at the Complex Operative Unit of Child Neuropsychiatry, Polyclinic of Bari, Italy. 25(OH)D was quantified by chemiluminescence immunoassay and level defined as: deficiency (<20 ng/mL); insufficiency (20-30); normality (30-100); toxicity (>100). Statistical analysis was performed using SPSS20 (significance < 0.05). The ASD group showed 25(OH)D a mean level significantly lower than control (p = 0.014). Multivariable logistic regression analysis showed an association between ASD and vitamin D deficiency (p = 0.006). The nature of such association is unclear. Vitamin D deficiency may probably act as a risk factor for the development of ASD. Further studies are needed to unravel the role of vitamin D in ASD etiology and investigate its therapeutic potential.
Collapse
Affiliation(s)
- Maria G. Petruzzelli
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari “Aldo Moro”, Bari, Italy
| | - Lucia Marzulli
- Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, Bari, Italy
| | - Francesco Margari
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari “Aldo Moro”, Bari, Italy
| | - Andrea De Giacomo
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari “Aldo Moro”, Bari, Italy
| | - Alessandra Gabellone
- Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, Bari, Italy
| | - Orazio V. Giannico
- Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, Bari, Italy
| | - Lucia Margari
- Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, Bari, Italy
| |
Collapse
|
115
|
Analyzing the Potential Biological Determinants of Autism Spectrum Disorder: From Neuroinflammation to the Kynurenine Pathway. Brain Sci 2020; 10:brainsci10090631. [PMID: 32932826 PMCID: PMC7563403 DOI: 10.3390/brainsci10090631] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/31/2020] [Accepted: 09/10/2020] [Indexed: 12/22/2022] Open
Abstract
Autism Spectrum Disorder (ASD) etiopathogenesis is still unclear and no effective preventive and treatment measures have been identified. Research has focused on the potential role of neuroinflammation and the Kynurenine pathway; here we review the nature of these interactions. Pre-natal or neonatal infections would induce microglial activation, with secondary consequences on behavior, cognition and neurotransmitter networks. Peripherally, higher levels of pro-inflammatory cytokines and anti-brain antibodies have been identified. Increased frequency of autoimmune diseases, allergies, and recurring infections have been demonstrated both in autistic patients and in their relatives. Genetic studies have also identified some important polymorphisms in chromosome loci related to the human leukocyte antigen (HLA) system. The persistence of immune-inflammatory deregulation would lead to mitochondrial dysfunction and oxidative stress, creating a self-sustaining cytotoxic loop. Chronic inflammation activates the Kynurenine pathway with an increase in neurotoxic metabolites and excitotoxicity, causing long-term changes in the glutamatergic system, trophic support and synaptic function. Furthermore, overactivation of the Kynurenine branch induces depletion of melatonin and serotonin, worsening ASD symptoms. Thus, in genetically predisposed subjects, aberrant neurodevelopment may derive from a complex interplay between inflammatory processes, mitochondrial dysfunction, oxidative stress and Kynurenine pathway overexpression. To validate this hypothesis a new translational research approach is necessary.
Collapse
|
116
|
Matelski L, Keil Stietz KP, Sethi S, Taylor SL, Van de Water J, Lein PJ. The influence of sex, genotype, and dose on serum and hippocampal cytokine levels in juvenile mice developmentally exposed to a human-relevant mixture of polychlorinated biphenyls. Curr Res Toxicol 2020; 1:85-103. [PMID: 34296199 PMCID: PMC8294704 DOI: 10.1016/j.crtox.2020.09.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Polychlorinated biphenyls (PCBs) are pervasive environmental contaminants implicated as risk factors for neurodevelopmental disorders (NDDs). Immune dysregulation is another NDD risk factor, and developmental PCB exposures are associated with early life immune dysregulation. Studies of the immunomodulatory effects of PCBs have focused on the higher-chlorinated congeners found in legacy commercial mixtures. Comparatively little is known about the immune effects of contemporary, lower-chlorinated PCBs. This is a critical data gap given recent reports that lower-chlorinated congeners comprise >70% of the total PCB burden in serum of pregnant women enrolled in the MARBLES study who are at increased risk for having a child with an NDD. To examine the influence of PCBs, sex, and genotype on cytokine levels, mice were exposed throughout gestation and lactation to a PCB mixture in the maternal diet, which was based on the 12 most abundant PCBs in sera from MARBLES subjects. Using multiplex array, cytokines were quantified in the serum and hippocampus of weanling mice expressing either a human gain-of-function mutation in ryanodine receptor 1 (T4826I mice), a human CGG premutation repeat expansion in the fragile X mental retardation gene 1 (CGG mice), or both mutations (DM mice). Congenic wildtype (WT) mice were used as controls. There were dose-dependent effects of PCB exposure on cytokine concentrations in the serum but not hippocampus. Differential effects of genotype were observed in the serum and hippocampus. Hippocampal cytokines were consistently elevated in T4826I mice and also in WT animals for some cytokines compared to CGG and DM mice, while serum cytokines were usually elevated in the mutant genotypes compared to the WT group. Males had elevated levels of 19 cytokines in the serum and 4 in the hippocampus compared to females, but there were also interactions between sex and genotype for 7 hippocampal cytokines. Only the chemokine CCL5 in the serum showed an interaction between PCB dose, genotype, and sex. Collectively, these findings indicate differential influences of PCB exposure and genotype on cytokine levels in serum and hippocampal tissue of weanling mice. These results suggest that developmental PCB exposure has chronic effects on baseline serum, but not hippocampal, cytokine levels in juvenile mice.
Collapse
Affiliation(s)
- Lauren Matelski
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, Davis, CA 95616, USA
| | - Kimberly P. Keil Stietz
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, Davis, CA 95616, USA
| | - Sunjay Sethi
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, Davis, CA 95616, USA
| | - Sandra L. Taylor
- Division of Biostatistics, Department of Public Health Sciences, University of California, Davis, School of Medicine, Davis, CA 95616, USA
| | - Judy Van de Water
- MIND Institute, University of California, Davis, School of Medicine, Sacramento, CA 95817, USA,Division of Rheumatology, Allergy, and Clinical Immunology, Department of Internal Medicine, University of California, Davis, School of Medicine, Davis, CA 95616, USA
| | - Pamela J. Lein
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, Davis, CA 95616, USA,MIND Institute, University of California, Davis, School of Medicine, Sacramento, CA 95817, USA,Corresponding author at: Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, 1089 Veterinary Medicine Drive, Davis, CA 95616, USA.
| |
Collapse
|
117
|
Patel S, Dale RC, Rose D, Heath B, Nordahl CW, Rogers S, Guastella AJ, Ashwood P. Maternal immune conditions are increased in males with autism spectrum disorders and are associated with behavioural and emotional but not cognitive co-morbidity. Transl Psychiatry 2020; 10:286. [PMID: 32796821 PMCID: PMC7429839 DOI: 10.1038/s41398-020-00976-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/21/2020] [Accepted: 07/24/2020] [Indexed: 12/16/2022] Open
Abstract
Epidemiological and animal research shows that maternal immune activation increases the risk of autism spectrum disorders (ASD) in offspring. Emerging evidence suggests that maternal immune conditions may play a role in the phenotypic expression of neurodevelopmental difficulties in children with ASD and this may be moderated by offspring sex. This study aimed to investigate whether maternal immune conditions were associated with increased severity of adverse neurodevelopmental outcomes in children with ASD. Maternal immune conditions were examined as predictors of ASD severity, behavioural and emotional well-being, and cognitive functioning in a cohort of 363 children with ASD (n = 363; 252 males, 111 females; median age 3.07 [interquartile range 2.64-3.36 years]). We also explored whether these outcomes varied between male and female children. Results showed that maternal asthma was the most common immune condition reported in mothers of children with ASD. A history of maternal immune conditions (p = 0.009) was more common in male children with ASD, compared to female children. Maternal immune conditions were associated with increased behavioural and emotional problems in male and female children. By contrast, maternal immune conditions were not associated with decreased cognitive function. The findings demonstrate that MIA may influence the expression of symptoms in children with ASD and outcomes may vary between males and females.
Collapse
Affiliation(s)
- Shrujna Patel
- grid.1013.30000 0004 1936 834XAutism Clinic for Translational Research, Brain and Mind Centre, Children’s Hospital Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW Australia
| | - Russell C. Dale
- grid.1013.30000 0004 1936 834XKids Neuroscience Centre, The Children’s Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, Westmead, NSW Australia
| | - Destanie Rose
- grid.27860.3b0000 0004 1936 9684Department of Medical Microbiology and Immunology and MIND Institute, UC Davis, Davis, CA USA
| | - Brianna Heath
- grid.27860.3b0000 0004 1936 9684Department of Psychiatry and MIND Institute, UC Davis, Davis, CA USA
| | - Christine W. Nordahl
- grid.27860.3b0000 0004 1936 9684Department of Psychiatry and MIND Institute, UC Davis, Davis, CA USA
| | - Sally Rogers
- grid.27860.3b0000 0004 1936 9684Department of Psychiatry and MIND Institute, UC Davis, Davis, CA USA
| | - Adam J. Guastella
- grid.1013.30000 0004 1936 834XAutism Clinic for Translational Research, Brain and Mind Centre, Children’s Hospital Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW Australia
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology and MIND Institute, UC Davis, Davis, CA, USA.
| |
Collapse
|
118
|
Daaboul J, Tamouza R, Leboyer M. [Immunopsychiatry and SARS-CoV-2 pandemic: Links and possible consequences]. Encephale 2020; 47:151-156. [PMID: 32928535 PMCID: PMC7373027 DOI: 10.1016/j.encep.2020.07.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 07/20/2020] [Indexed: 12/28/2022]
Abstract
OBJECTIVE The SARS-CoV-2 (or COVID-19) pandemic has been propagating since December 2019, inducing a drastic increase in the prevalence of anxious and depressive disorders in the general population. Psychological trauma can partly explain these disorders. However, since psychiatric disorders also have an immuno-inflammatory component, the direct effects of the virus on the host's immune system, with a marked inflammatory response, but also the secondary inflammation to these psychosocial stressors, may cause the apparition or the worsening of psychiatric disorders. We describe here the probable immunopsychiatric consequences of the SARS-CoV-2 pandemic, to delineate possible screening actions and care that could be planned. METHOD Data from previous pandemics, and existing data on the psychopathological consequences of the SARS-CoV-2 pandemic, allowed us to review the possible immunopsychiatric consequences of the SARS-CoV-2 pandemic, on the gestational environment, with the risk of consecutive neurodevelopmental disorders for the fetus on one hand, on the children and adults directly infected being at increased risks of psychiatric disorders on the other hand. RESULTS As in previous pandemics, the activation of the immune system due to psychological stress and/or to infection during pregnancy, might lead to an increased risk of neurodevelopmental disorders for the fetus (schizophrenia and autism spectrum disorders). Furthermore, in individuals exposed to psychological trauma and/or infected by the virus, the risk of psychiatric disorders, especially mood disorders, is probably increased. CONCLUSION In this context, preventive measures and specialized care are necessary. Thus, it is important to propose a close follow-up to the individuals who have been infected by the virus, in order to set up the earliest care possible. Likewise, in pregnant women, screening of mood disorders during the pregnancy or the postpartum period must be facilitated. The follow-up of the babies born during the pandemic must be strengthened to screen and care for possible neurodevelopmental disorders.
Collapse
Affiliation(s)
- J Daaboul
- Université de Lille, CHU de Lille, Lille, France; DMU IMPACT, département médico-universitaire de psychiatrie et d'addictologie du groupe hospitalier universitaire Henri-Mondor, AP-HP, Créteil, France
| | - R Tamouza
- DMU IMPACT, département médico-universitaire de psychiatrie et d'addictologie du groupe hospitalier universitaire Henri-Mondor, AP-HP, Créteil, France; Fondation FondaMental, Créteil, France; Université Paris Est Créteil, UPEC, Inserm, U955, équipe 15 neuro-psychiatrie translationnelle, Institut Mondor de Recherche Biomédicale, IMRB, Créteil, France
| | - M Leboyer
- DMU IMPACT, département médico-universitaire de psychiatrie et d'addictologie du groupe hospitalier universitaire Henri-Mondor, AP-HP, Créteil, France; Fondation FondaMental, Créteil, France; Université Paris Est Créteil, UPEC, Inserm, U955, équipe 15 neuro-psychiatrie translationnelle, Institut Mondor de Recherche Biomédicale, IMRB, Créteil, France.
| |
Collapse
|
119
|
Vargason T, Grivas G, Hollowood-Jones KL, Hahn J. Towards a Multivariate Biomarker-Based Diagnosis of Autism Spectrum Disorder: Review and Discussion of Recent Advancements. Semin Pediatr Neurol 2020; 34:100803. [PMID: 32446437 PMCID: PMC7248126 DOI: 10.1016/j.spen.2020.100803] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
An ever-evolving understanding of autism spectrum disorder (ASD) pathophysiology necessitates that diagnostic standards also evolve from being observation-based to include quantifiable clinical measurements. The multisystem nature of ASD motivates the use of multivariate methods of statistical analysis over common univariate approaches for discovering clinical biomarkers relevant to this goal. In addition to characterization of important behavioral patterns for improving current diagnostic instruments, multivariate analyses to date have allowed for thorough investigation of neuroimaging-based, genetic, and metabolic abnormalities in individuals with ASD. This review highlights current research using multivariate statistical analyses to quantify the value of these behavioral and physiological markers for ASD diagnosis. A detailed discussion of a blood-based diagnostic test for ASD using specific metabolite concentrations is also provided. The advancement of ASD biomarker research promises to provide earlier and more accurate diagnoses of the disorder.
Collapse
Affiliation(s)
- Troy Vargason
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY
| | - Genevieve Grivas
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY
| | - Kathryn L Hollowood-Jones
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY
| | - Juergen Hahn
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY; Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, NY.
| |
Collapse
|
120
|
Ashitha SNM, Ramachandra NB. Integrated Functional Analysis Implicates Syndromic and Rare Copy Number Variation Genes as Prominent Molecular Players in Pathogenesis of Autism Spectrum Disorders. Neuroscience 2020; 438:25-40. [DOI: 10.1016/j.neuroscience.2020.04.051] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 04/28/2020] [Accepted: 04/29/2020] [Indexed: 01/05/2023]
|
121
|
Anderson G, Betancort Medina SR. Autism Spectrum Disorders: Role of Pre- and Post-Natal GammaDelta (γδ) T Cells and Immune Regulation. Curr Pharm Des 2020; 25:4321-4330. [PMID: 31682211 DOI: 10.2174/1381612825666191102170125] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 10/31/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND It is widely accepted that alterations in immune functioning are an important aspect of the pathoetiology and pathophysiology of autism spectrum disorders (ASD). A relatively under-explored aspect of these alterations is the role of gammaDelta (γδ) T cells, prenatally and in the postnatal gut, which seem important hubs in driving the course of ASD. METHODS The present article describes the role of γδ T cells in ASD, including their interactions with other immune cells shown to be altered in this spectrum of conditions, including natural killer cells and mast cells. RESULTS Other risk factors in ASD, such as decreased vitamins A & D, as well as toxin-associated activation of the aryl hydrocarbon receptor, may also be intimately linked to γδ T cells, and alterations in the regulation of these cells. A growing body of data has highlighted an important role for alterations in mitochondria functioning in the regulation of immune cells, including natural killer cells and mast cells. This is an area that requires investigation in γδ T cells and their putative subtypes. CONCLUSION It is also proposed that maternal stress may act through alterations in the maternal microbiome, leading to changes in how the balance of short-chain fatty acids, such as butyrate, which may act to regulate the placenta and foetal development. Following an overview of previous research on immune, especially γδ T cells, effects in ASD, the future research implications are discussed in detail.
Collapse
Affiliation(s)
- George Anderson
- CRC Scotland & London, Eccleston Square, London, United Kingdom
| | | |
Collapse
|
122
|
Pacheva I, Ivanov I. Targeted Biomedical Treatment for Autism Spectrum Disorders. Curr Pharm Des 2020; 25:4430-4453. [PMID: 31801452 DOI: 10.2174/1381612825666191205091312] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 12/02/2019] [Indexed: 12/29/2022]
Abstract
BACKGROUND A diagnosis of autism spectrum disorders (ASD) represents presentations with impairment in communication and behaviour that vary considerably in their clinical manifestations and etiology as well as in their likely pathophysiology. A growing body of data indicates that the deleterious effect of oxidative stress, mitochondrial dysfunction, immune dysregulation and neuroinflammation, as well as their interconnections are important aspects of the pathophysiology of ASD. Glutathione deficiency decreases the mitochondrial protection against oxidants and tumor necrosis factor (TNF)-α; immune dysregulation and inflammation inhibit mitochondrial function through TNF-α; autoantibodies against the folate receptors underpin cerebral folate deficiency, resulting in disturbed methylation, and mitochondrial dysfunction. Such pathophysiological processes can arise from environmental and epigenetic factors as well as their combined interactions, such as environmental toxicant exposures in individuals with (epi)genetically impaired detoxification. The emerging evidence on biochemical alterations in ASD is forming the basis for treatments aimed to target its biological underpinnings, which is of some importance, given the uncertain and slow effects of the various educational interventions most commonly used. METHODS Literature-based review of the biomedical treatment options for ASD that are derived from established pathophysiological processes. RESULTS Most proposed biomedical treatments show significant clinical utility only in ASD subgroups, with specified pre-treatment biomarkers that are ameliorated by the specified treatment. For example, folinic acid supplementation has positive effects in ASD patients with identified folate receptor autoantibodies, whilst the clinical utility of methylcobalamine is apparent in ASD patients with impaired methylation capacity. Mitochondrial modulating cofactors should be considered when mitochondrial dysfunction is evident, although further research is required to identify the most appropriate single or combined treatment. Multivitamins/multiminerals formulas, as well as biotin, seem appropriate following the identification of metabolic abnormalities, with doses tapered to individual requirements. A promising area, requiring further investigations, is the utilization of antipurinergic therapies, such as low dose suramin. CONCLUSION The assessment and identification of relevant physiological alterations and targeted intervention are more likely to produce positive treatment outcomes. As such, current evidence indicates the utility of an approach based on personalized and evidence-based medicine, rather than treatment targeted to all that may not always be beneficial (primum non nocere).
Collapse
Affiliation(s)
- Iliyana Pacheva
- Department of Pediatrics and Medical Genetics, Medical University - Plovdiv, Plovdiv 4002, Bulgaria
| | - Ivan Ivanov
- Department of Pediatrics and Medical Genetics, Medical University - Plovdiv, Plovdiv 4002, Bulgaria
| |
Collapse
|
123
|
Runge K, Tebartz van Elst L, Maier S, Nickel K, Denzel D, Matysik M, Kuzior H, Robinson T, Blank T, Dersch R, Domschke K, Endres D. Cerebrospinal Fluid Findings of 36 Adult Patients with Autism Spectrum Disorder. Brain Sci 2020; 10:E355. [PMID: 32521749 PMCID: PMC7349103 DOI: 10.3390/brainsci10060355] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 06/01/2020] [Accepted: 06/01/2020] [Indexed: 12/27/2022] Open
Abstract
Autism spectrum disorder (ASD) is a common neurodevelopmental disorder characterized by difficulties with social interaction, repetitive behavior, and additional features, such as special interests. Its precise etiology is unclear. Recently, immunological mechanisms, such as maternal autoantibodies/infections, have increasingly been the subject of discussion. Cerebrospinal fluid (CSF) investigations play a decisive role in the detection of immunological processes in the brain. This study therefore retrospectively analyzed the CSF findings of adult patients with ASD. CSF basic measures (white blood cell count, total protein, albumin quotient, immunoglobulin G (IgG) index, and oligoclonal bands) and various antineuronal antibody findings of 36 adult patients with ASD, who had received lumbar puncture, were compared with an earlier described mentally healthy control group of 39 patients with idiopathic intracranial hypertension. CSF protein concentrations and albumin quotients of patients with ASD were significantly higher as compared to controls (age corrected: p = 0.003 and p = 0.004, respectively); 17% of the patients with ASD showed increased albumin quotients. After correction for age and gender, the group effect for total protein remained significant (p = 0.041) and showed a tendency for albumin quotient (p = 0.079). In the CSF of two ASD patients, an intrathecal synthesis of anti-glutamate decarboxylase 65 (GAD65) antibodies was found. In total, more of the ASD patients (44%) presented abnormal findings in CSF basic diagnostics compared to controls (18%; p = 0.013). A subgroup of the patients with adult ASD showed indication of a blood-brain barrier dysfunction, and two patients displayed an intrathecal synthesis of anti-GAD65 antibodies; thus, the role of these antibodies in patients with ASD should be further investigated. The results of the study are limited by its retrospective and open design. The group differences in blood-brain barrier markers could be influenced by a different gender distribution between ASD patients and controls.
Collapse
Affiliation(s)
- Kimon Runge
- Section for Experimental Neuropsychiatry, Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany; (K.R.); (S.M.); (K.N.); (D.D.); (M.M.); (H.K.); (D.E.)
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany;
| | - Ludger Tebartz van Elst
- Section for Experimental Neuropsychiatry, Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany; (K.R.); (S.M.); (K.N.); (D.D.); (M.M.); (H.K.); (D.E.)
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany;
| | - Simon Maier
- Section for Experimental Neuropsychiatry, Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany; (K.R.); (S.M.); (K.N.); (D.D.); (M.M.); (H.K.); (D.E.)
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany;
| | - Kathrin Nickel
- Section for Experimental Neuropsychiatry, Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany; (K.R.); (S.M.); (K.N.); (D.D.); (M.M.); (H.K.); (D.E.)
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany;
| | - Dominik Denzel
- Section for Experimental Neuropsychiatry, Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany; (K.R.); (S.M.); (K.N.); (D.D.); (M.M.); (H.K.); (D.E.)
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany;
| | - Miriam Matysik
- Section for Experimental Neuropsychiatry, Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany; (K.R.); (S.M.); (K.N.); (D.D.); (M.M.); (H.K.); (D.E.)
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany;
| | - Hanna Kuzior
- Section for Experimental Neuropsychiatry, Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany; (K.R.); (S.M.); (K.N.); (D.D.); (M.M.); (H.K.); (D.E.)
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany;
| | - Tilman Robinson
- Department of Neurology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany; (T.R.); (R.D.)
| | - Thomas Blank
- Institute of Neuropathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany;
| | - Rick Dersch
- Department of Neurology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany; (T.R.); (R.D.)
| | - Katharina Domschke
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany;
- Center for Basics in Neuromodulation, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Dominique Endres
- Section for Experimental Neuropsychiatry, Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany; (K.R.); (S.M.); (K.N.); (D.D.); (M.M.); (H.K.); (D.E.)
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany;
| |
Collapse
|
124
|
Price J. Cell therapy approaches to autism: a review of clinical trial data. Mol Autism 2020; 11:37. [PMID: 32448347 PMCID: PMC7245880 DOI: 10.1186/s13229-020-00348-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 05/11/2020] [Indexed: 12/31/2022] Open
Abstract
A number of clinical trials of cell therapies for autism spectrum disorder have been conducted, and some have published their outcomes. This review considers the data that have emerged from this small set of published trials, evaluates their success, and proposes further steps that could be taken if this field of endeavour is to be pursued further. A number of reservations arise from this tranche of studies, specifically the absence of identified therapeutic targets, and deficiencies in the therapeutic approach that is being employed. If this therapeutic direction is to be pursued further, then additional pre-clinical studies are recommended that might lead to improvements in patient stratification, biomarkers, the defined mode of action, and the preparation and identification of the therapeutic cells themselves.
Collapse
Affiliation(s)
- Jack Price
- Institute for Psychiatry, Psychology, & Neuroscience, King's College London, De Crespigny Park, Denmark Hill, London, SE5 8AF, UK.
| |
Collapse
|
125
|
De Simone R, Butera A, Armida M, Pezzola A, Boirivant M, Potenza RL, Ricceri L. Beneficial Effects of Fingolimod on Social Interaction, CNS and Peripheral Immune Response in the BTBR Mouse Model of Autism. Neuroscience 2020; 435:22-32. [PMID: 32229233 DOI: 10.1016/j.neuroscience.2020.03.041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/01/2020] [Accepted: 03/24/2020] [Indexed: 12/13/2022]
Abstract
Autism Spectrum Disorders (ASD) are neurodevelopmental disorders characterized by social communication deficits and repetitive/stereotyped behaviours. We evaluated the effects of a chronic treatment with the immunomodulator drug Fingolimod (FTY720 - a non-selective Sphingosine 1-Phosphate Receptor ligand) in an ASD model, the BTBR T+tf/J (BTBR) mouse strain. In adult BTBR males, chronic FTY720 treatment (4 weeks) increased social and vocal response during a male-female interaction and hippocampal expression of BDNF and Neuregulin 1, two trophic factors reduced in BTBR when compared to control C57 mice. FTY720 also re-established the expression of IL-1β and MnSOD in the hippocampus, whereas it did not modify IL-6 mRNA content. In addition to its central effect, FTY720 modulated the activation state of peripheral macrophages in the BTBR model, both in basal conditions and after stimulation with an immune challenge. Furthermore, IL-6 mRNA colonic content of BTBR mice, reduced when compared with C57 mice, was normalized by chronic treatment with FTY720. Our study, while indicating FTY720 as a tool to attenuate relevant alterations of the BTBR neurobehavioural phenotype, emphasizes the importance of gut mucosal immune evaluation as an additional target that deserve to be investigated in preclinical studies of anti-inflammatory therapeutic approaches in ASD.
Collapse
Affiliation(s)
- Roberta De Simone
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Alessia Butera
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Monica Armida
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Antonella Pezzola
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Monica Boirivant
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Rosa Luisa Potenza
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy.
| | - Laura Ricceri
- Centre for Behavioural Science and Mental Health, Istituto Superiore di Sanità, Rome, Italy.
| |
Collapse
|
126
|
Park DI. Genomics, transcriptomics, proteomics and big data analysis in the discovery of new diagnostic markers and targets for therapy development. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 173:61-90. [PMID: 32711818 DOI: 10.1016/bs.pmbts.2020.04.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Highly complex endophenotypes and underlying molecular mechanisms have prevented effective diagnosis and treatment of autism spectrum disorder. Despite extensive studies to identify relevant biosignatures, no biomarker and therapeutic targets are available in the current clinical practice. While our current knowledge is still largely incomplete, -omics technology and machine learning-based big data analysis have provided novel insights on the etiology of autism spectrum disorders, elucidating systemic impairments that can be translated into biomarker and therapy target candidates. However, more integrated and sophisticated approaches are vital to realize molecular stratification and individualized treatment strategy. Ultimately, systemic approaches based on -omics and big data analysis will significantly contribute to more effective biomarker and therapy development for autism spectrum disorder.
Collapse
Affiliation(s)
- Dong Ik Park
- Danish Research Institute of Translational Neuroscience (DANDRITE)-Nordic EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, Aarhus, Denmark; The Danish National Research Foundation Center, PROMEMO, Department of Biomedicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
127
|
Nadeem A, Ahmad SF, Al-Harbi NO, Alasmari AF, Al-Ayadhi LY, Alasmari F, Ibrahim KE, Attia SM, Bakheet SA. Upregulation of enzymatic antioxidants in CD4 + T cells of autistic children. Biochimie 2020; 171-172:205-212. [PMID: 32173487 DOI: 10.1016/j.biochi.2020.03.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 03/10/2020] [Indexed: 01/06/2023]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder which begins in early childhood and presents itself with characteristic symptoms such as repetitive behavioral patterns and problems in speech/social interactions. Adaptive immune system is thought to be involved in the etiology of ASD. T cells orchestrate amplification of inflammation through release of inflammatory mediators; however, antioxidant defenses have not been evaluated in CD4+ T cells of ASD subjects. In this study we evaluated intracellular enzymatic antioxidant potential through measurement of major antioxidant enzymes (SOD, GPx, and GR) in ASD subjects and typically developing control (TDC) children and further assessed its role in modulation of inflammation. Our data reveal that there is an increase in antioxidant potential (SOD, GPx, GR) in CD4+ T cells of ASD subjects as compared to TDC children at both protein and activity level. Further, this antioxidant increase was associated with upregulated IL-17A levels in CD4+ T cells. This was corroborated by oxidant treatment in vitro. Pretreatment with oxidant, H2O2 led to attenuation of IL-17A levels along with increased oxidative stress in stimulated CD4+ T cells from ASD subjects. These data reveal that antioxidant play an essential role in modulation of inflammatory potential in CD4+ T cells of ASD subjects.
Collapse
Affiliation(s)
- Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.
| | - Sheikh F Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Naif O Al-Harbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdullah F Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Laila Y Al-Ayadhi
- Autism Research and Treatment Center, AL-Amodi Autism Research Chair, Department of Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Fawaz Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Khalid E Ibrahim
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Saleh A Bakheet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
128
|
Bellavite P. Causality assessment of adverse events following immunization: the problem of multifactorial pathology. F1000Res 2020; 9:170. [PMID: 32269767 PMCID: PMC7111503 DOI: 10.12688/f1000research.22600.1] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/04/2020] [Indexed: 07/22/2023] Open
Abstract
The analysis of Adverse Events Following Immunization (AEFI) is important in a balanced epidemiological evaluation of vaccines and in the issues related to national vaccine injury compensation programs. If manufacturing defects or vaccine storage and delivering errors are excluded, the majority of adverse reactions to vaccines occur as excessive or biased inflammatory and immune responses. These unwanted phenomena, occasionally severe, are associated with many different endogenous and exogenous factors, which often interact in complex ways. The confirmation or denial of the causal link between an AEFI and vaccination is determined pursuant to WHO guidelines, which propose a four-step analysis and algorithmic diagramming. The evaluation process from the onset considers all possible "other causes" that can explain the AEFI and thus exclude the role of the vaccine. Subsequently, even if there was biological plausibility and temporal compatibility for a causal association between the vaccine and the AEFI, the guidelines ask to look for any possible evidence that the vaccine could not have caused that event. Such an algorithmic method presents some concerns that are discussed here, in the light of the multifactorial nature of the inflammatory and immune pathologies induced by vaccines, including emerging knowledge of genetic susceptibility to adverse effects. It is proposed that the causality assessment could exclude a consistent association of the adverse event with the vaccine only when the presumed "other cause" is independent of an interaction with the vaccine. Furthermore, the scientific literature should be viewed not as an exclusion criterion but as a comprehensive analysis of all the evidence for or against the role of the vaccine in causing an adverse reaction. These issues are discussed in relation to the laws that, in some countries, regulate the mandatory vaccinations and the compensation for those who have suffered serious adverse effects.
Collapse
Affiliation(s)
- Paolo Bellavite
- Department of Medicine, Section of General Pathology, University of Verona Medical School, Verona, 37134, Italy
| |
Collapse
|
129
|
Abstract
The analysis of Adverse Events Following Immunization (AEFI) is important in a balanced epidemiological evaluation of vaccines and in the issues related to vaccine injury compensation programs. The majority of adverse reactions to vaccines occur as excessive or biased inflammatory and immune responses. These unwanted phenomena, occasionally severe, are associated with many different endogenous and exogenous factors, which often interact in complex ways. The confirmation or denial of the causal link between an AEFI and vaccination is determined pursuant to WHO guidelines, which propose a four-step analysis and algorithmic diagramming. The evaluation process from the onset considers all possible "other causes" that might explain the AEFI and thus exclude the role of the vaccine. Subsequently, even if there was biological plausibility and temporal compatibility for a causal association between the vaccine and the AEFI, the guidelines ask to look for any possible evidence that the vaccine could not have caused that event. Such an algorithmic method presents several concerns that are discussed here, in the light of the multifactorial nature of the inflammatory and immune pathologies induced by vaccines, including emerging knowledge of genetic susceptibility to adverse effects. It is proposed that the causality assessment could exclude a consistent association of the adverse event with the vaccine only when the presumed "other cause" is independent of an interaction with the vaccine. Furthermore, the scientific literature should be viewed not as an exclusion criterion but as a comprehensive analysis of all the evidence for or against the role of the vaccine in causing an adverse reaction. Given these inadequacies in the evaluation of multifactorial diseases, the WHO guidelines need to be reevaluated and revised. These issues are discussed in relation to the laws that, in some countries, regulate the mandatory vaccinations and the compensation for those who have suffered serious adverse effects.
Collapse
Affiliation(s)
- Paolo Bellavite
- Department of Medicine, Section of General Pathology, University of Verona Medical School, Verona, 37134, Italy
| |
Collapse
|
130
|
Nadeem A, Ahmad SF, Attia SM, Al-Ayadhi LY, Al-Harbi NO, Bakheet SA. Dysregulation in IL-6 receptors is associated with upregulated IL-17A related signaling in CD4+ T cells of children with autism. Prog Neuropsychopharmacol Biol Psychiatry 2020; 97:109783. [PMID: 31655158 DOI: 10.1016/j.pnpbp.2019.109783] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 09/13/2019] [Accepted: 10/06/2019] [Indexed: 11/17/2022]
Abstract
Autism spectrum disorder (ASD) is a heterogeneous syndrome characterized by dysregulations in speech and social interactions as well as repetitive and stereotypical behavioral patterns in which immune system plays a significant role. IL-6, an essential cytokine for polarization of Th0 cells into Th17 cells has been demonstrated to be crucial in the etiology of ASD in past studies both in humans and mice. Th17 cells are also believed to be central players in the pathogenesis of ASD through release of IL-17A. However, there is still insufficient data regarding identification of Th17 cells with respect to IL-6 signaling in ASD subjects. Therefore, this study explored IL-6 receptors (IL-6R/sIL-6R) and Th17 (p-STAT3/IL-17A/IL-23R) related markers comprehensively in the blood of typically-developing control (TDC, n = 35) and ASD children (n = 45). Our data show that there is enhanced sIL-6R levels in plasma and CD4+ T cells of ASD subjects as compared to TDC group. Increased sIL-6R signaling is associated with upregulated Th17 development in ASD subjects. Further, severe ASD subjects have higher inflammation in terms of IL-6/IL-17A related signaling as compared to moderate ASD patients. Furthermore, treatment of CD4 + T cells in vitro with IL-6 leads to much greater upregulation of p-STAT3, and IL-17A in ASD subjects than similarly treated CD4+ T cells in TDC group. Antagonism of IL-6 signaling by SC144 in vitro led to blockade of IL-6 mediated effects on CD4+ T cells. These data display unequivocally that IL-6 signaling components are dysregulated which play a crucial in enhancement of Th17 development in ASD subjects.
Collapse
Affiliation(s)
- Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.
| | - Sheikh F Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Laila Y Al-Ayadhi
- Autism Research and Treatment Center, AL-Amodi Autism Research Chair, Department of Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Naif O Al-Harbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Saleh A Bakheet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
131
|
Differential regulation of Nrf2 is linked to elevated inflammation and nitrative stress in monocytes of children with autism. Psychoneuroendocrinology 2020; 113:104554. [PMID: 31884317 DOI: 10.1016/j.psyneuen.2019.104554] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 12/10/2019] [Accepted: 12/11/2019] [Indexed: 12/17/2022]
Abstract
Autism spectrum disorder (ASD) is a very complex neurodevelopmental disorder characterized by deficits in social and communication skills. Innate immune cells like monocytes are believed to play a cardinal role in neuroimmune inflammation and nitrative stress. On the other hand, Nrf2, a basic leucine zipper transcription factor plays a significant role in protecting the immune cells against inflammation and oxidants. However, its role in monocytes of ASD children and typically developing control (TDC) children has not been elucidated in relation with inflammation and nitrative stress. Therefore, this study was undertaken to evaluate Nrf2 expression/activity along with parameters of inflammation (NFkB, IL-6, IL-1β) and nitrative stress (iNOS, nitrotyrosine) in monocytes of ASD/TDC children. Further, sulforaphane (SFN) was utilized as an Nrf2 activator to assess its effect on above said inflammatory and nitrative stress parameters. Our study shows that monocytes of ASD subjects have decreased Nrf2 expression/activity along with increased inflammation and nitrative stress. Further, monocytes from ASD have deficiency in induction of Nrf2 activity upon stimulation with LPS. However, activation of Nrf2 in vitro by SFN reverses LPS-induced effects on inflammation in monocytes by reduction in NFkB signaling. Further, treatment with SFN also reverses LPS-induced effects on nitrative stress (iNOS, nitrotyrosine) in monocytes of ASD subjects. This study propounds the idea that SFN protects against nitrative stress and inflammation by downregulating oxidative stress and inflammation through blockade of NFkB signaling in autistic children. This may be the reason behind reported ameliorative effects of SFN in ASD subjects.
Collapse
|
132
|
Sardaar S, Qi B, Dionne-Laporte A, Rouleau GA, Rabbany R, Trakadis YJ. Machine learning analysis of exome trios to contrast the genomic architecture of autism and schizophrenia. BMC Psychiatry 2020; 20:92. [PMID: 32111185 PMCID: PMC7049199 DOI: 10.1186/s12888-020-02503-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 02/17/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Machine learning (ML) algorithms and methods offer great tools to analyze large complex genomic datasets. Our goal was to compare the genomic architecture of schizophrenia (SCZ) and autism spectrum disorder (ASD) using ML. METHODS In this paper, we used regularized gradient boosted machines to analyze whole-exome sequencing (WES) data from individuals SCZ and ASD in order to identify important distinguishing genetic features. We further demonstrated a method of gene clustering to highlight which subsets of genes identified by the ML algorithm are mutated concurrently in affected individuals and are central to each disease (i.e., ASD vs. SCZ "hub" genes). RESULTS In summary, after correcting for population structure, we found that SCZ and ASD cases could be successfully separated based on genetic information, with 86-88% accuracy on the testing dataset. Through bioinformatic analysis, we explored if combinations of genes concurrently mutated in patients with the same condition ("hub" genes) belong to specific pathways. Several themes were found to be associated with ASD, including calcium ion transmembrane transport, immune system/inflammation, synapse organization, and retinoid metabolic process. Moreover, ion transmembrane transport, neurotransmitter transport, and microtubule/cytoskeleton processes were highlighted for SCZ. CONCLUSIONS Our manuscript introduces a novel comparative approach for studying the genetic architecture of genetically related diseases with complex inheritance and highlights genetic similarities and differences between ASD and SCZ.
Collapse
Affiliation(s)
- Sameer Sardaar
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Bill Qi
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Alexandre Dionne-Laporte
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Guy A Rouleau
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Reihaneh Rabbany
- School of Computer Science, McGill University, Montreal, QC, Canada
- Montreal Institute for Learning Algorithms, Université de Montréal, Montreal, QC, Canada
| | - Yannis J Trakadis
- Department of Human Genetics, McGill University, Montreal, QC, Canada.
- Department of Medical Genetics, McGill University Health Center Room A04.3140, Montreal Children's Hospital,1001 Boul. Décarie, H4A 3J1, Montreal, Quebec, Canada.
| |
Collapse
|
133
|
Bodnar TS, Raineki C, Wertelecki W, Yevtushok L, Plotka L, Granovska I, Zymak-Zakutnya N, Pashtepa A, Wells A, Honerkamp-Smith G, Coles CD, Kable JA, Chambers CD, Weinberg J. Immune network dysregulation associated with child neurodevelopmental delay: modulatory role of prenatal alcohol exposure. J Neuroinflammation 2020; 17:39. [PMID: 31992316 PMCID: PMC6988366 DOI: 10.1186/s12974-020-1717-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Accepted: 01/14/2020] [Indexed: 12/19/2022] Open
Abstract
Background Evidence suggests that cytokine imbalances may be at the root of deficits that occur in numerous neurodevelopmental disorders, including schizophrenia and autism spectrum disorder. Notably, while clinical studies have demonstrated maternal cytokine imbalances with alcohol consumption during pregnancy—and data from animal models have identified immune disturbances in alcohol-exposed offspring—to date, immune alterations in alcohol-exposed children have not been explored. Thus, here we hypothesized that perturbations in the immune environment as a result of prenatal alcohol exposure will program the developing immune system, and result in immune dysfunction into childhood. Due to the important role of cytokines in brain development/function, we further hypothesized that child immune profiles might be associated with their neurodevelopmental status. Methods As part of a longitudinal study in Ukraine, children of mothers reporting low/no alcohol consumption or moderate-to-heavy alcohol consumption during pregnancy were enrolled in the study and received neurodevelopmental assessments. Group stratification was based on maternal alcohol consumption and child neurodevelopmental status resulting in the following groups: A/TD, alcohol-consuming mother, typically developing child; A/ND, alcohol-consuming mother, neurodevelopmental delay in the child; C/TD, control mother (low/no alcohol consumption), typically development child; and C/ND, control mother, neurodevelopmental delay in the child. Forty cytokines/chemokines were measured in plasma and data were analyzed using regression and constrained principle component analysis. Results Analyses revealed differential cytokine network activity associated with both prenatal alcohol exposure and neurodevelopmental status. Specifically, alcohol-exposed children showed activation of a cytokine network including eotaxin-3, eotaxin, and bFGF, irrespective of neurodevelopmental status. However, another cytokine network was differentially activated based on neurodevelopmental outcome: A/TD showed activation of MIP-1β, MDC, and MCP-4, and inhibition of CRP and PlGF, with opposing pattern of activation/inhibition detected in the A/ND group. By contrast, in the absence of alcohol-exposure, activation of a network including IL-2, TNF-β, IL-10, and IL-15 was associated with neurodevelopmental delay. Conclusions Taken together, this comprehensive assessment of immune markers allowed for the identification of unique immune milieus that are associated with alcohol exposure as well as both alcohol-related and alcohol-independent neurodevelopmental delay. These findings are a critical step towards establishing unique immune biomarkers for alcohol-related and alcohol-independent neurodevelopmental delay.
Collapse
Affiliation(s)
- Tamara S Bodnar
- Department of Cellular and Physiological Sciences, University of British Columbia, 3307 - 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada.
| | - Charlis Raineki
- Department of Cellular and Physiological Sciences, University of British Columbia, 3307 - 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | | | - Lyubov Yevtushok
- OMNI-Net for Children International Charitable Fund, Rivne Oblast Medical Diagnostic Center, Rivne, Ukraine
| | - Larisa Plotka
- OMNI-Net for Children International Charitable Fund, Rivne Oblast Medical Diagnostic Center, Rivne, Ukraine
| | - Irina Granovska
- OMNI-Net for Children International Charitable Fund, Rivne Oblast Medical Diagnostic Center, Rivne, Ukraine
| | - Natalya Zymak-Zakutnya
- OMNI-Net for Children International Charitable Fund, Khmelnytsky Perinatal Center, Khmelnytsky, Ukraine
| | - Alla Pashtepa
- OMNI-Net for Children International Charitable Fund, Khmelnytsky Perinatal Center, Khmelnytsky, Ukraine
| | - Alan Wells
- Department of Pediatrics, University of California San Diego, La Jolla, USA
| | | | - Claire D Coles
- Department of Psychiatry and Behavioral Sciences; Department of Pediatrics, Emory University School of Medicine, Atlanta, USA
| | - Julie A Kable
- Department of Psychiatry and Behavioral Sciences; Department of Pediatrics, Emory University School of Medicine, Atlanta, USA
| | - Christina D Chambers
- Department of Pediatrics, University of California San Diego, La Jolla, USA.,Department of Family Medicine and Public Health, University of California San Diego, La Jolla, CA, USA
| | - Joanne Weinberg
- Department of Cellular and Physiological Sciences, University of British Columbia, 3307 - 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | | |
Collapse
|
134
|
Alzghoul L. Role of Vitamin D in Autism Spectrum Disorder. Curr Pharm Des 2020; 25:4357-4367. [DOI: 10.2174/1381612825666191122092215] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 11/15/2019] [Indexed: 12/19/2022]
Abstract
:
Autism spectrum disorder (ASD) is a pervasive developmental disorder with heterogeneous etiology.
Vitamin D can function as a fat-soluble vitamin as well as a hormone, and can exert its effect through both genomic
and non-genomic mechanisms. In the last decades, several studies have examined the relationship between
vitamin D levels and ASD. These studies demonstrated that low vitamin D status in early development has been
hypothesized as an environmental risk factor for ASD. Both in vivo and in vitro studies have demonstrated that
vitamin D deficiency in early life can alter brain development, dysregulates neurotransmitter balance in the brain,
decreases body and brain antioxidant ability, and alters the immune system in ways that resemble pathological
features commonly seen in ASD. In this review, we focused on the association between vitamin D and ASD. In
addition, the above-mentioned mechanisms of action that link vitamin D deficiency with ASD were also discussed.
Finally, clinical trials of vitamin D supplementation treatment of ASD have also been discussed.
Collapse
Affiliation(s)
- Loai Alzghoul
- Department of Physiology and Biochemistry, School of Medicine, The University of Jordan, Amman, Jordan
| |
Collapse
|
135
|
Yadama AP, Kelly RS, Lee-Sarwar K, Mirzakhani H, Chu SH, Kachroo P, Litonjua AA, Lasky-Su J, Weiss ST. Allergic disease and low ASQ communication score in children. Brain Behav Immun 2020; 83:293-297. [PMID: 31606476 PMCID: PMC6906237 DOI: 10.1016/j.bbi.2019.10.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/20/2019] [Accepted: 10/05/2019] [Indexed: 01/28/2023] Open
Abstract
Autism Spectrum Disorders (ASD) are complex and multifactorial. Previous investigations have revealed associations between allergic disease and ASD, which are characterized by impaired communication skills. In this study we observed an association between allergic disease and communication skills development as assessed by the Ages and Stages Questionnaire (ASQ) communication score, as a proxy for ASD, among children who participated in the Vitamin D Antenatal Asthma Reduction Trial (VDAART). In particular, we observed significant associations between both a diagnosis of eczema at age 3 years (OR = 1.87; confidence interval [CI]: 0.97-3.47; p = 0.054) and a diagnosis of food allergy at age 6 years (OR = 3.61; 95% CI: 1.18-9.85; p = 0.015) with ASQ communication score. Plasma metabolomics analyses suggest that dysregulated tryptophan metabolism may contribute to the pathogenesis of these co-morbidities.
Collapse
Affiliation(s)
- Aishwarya P. Yadama
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston MA 02115
| | - Rachel S. Kelly
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston MA 02115
| | - Kathleen Lee-Sarwar
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston MA 02115,Division of Rheumatology, Immunology and Allergy, Brigham and Women’s Hospital, Harvard Medical School, Boston MA 02115
| | - Hooman Mirzakhani
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston MA 02115
| | - Su H. Chu
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston MA 02115
| | - Priyadarshini Kachroo
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston MA 02115
| | - Augusto A. Litonjua
- Division of Pediatric Pulmonary Medicine, University of Rochester Medical Center, Rochester, NY 14642
| | - Jessica Lasky-Su
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston MA 02115
| | - Scott T. Weiss
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston MA 02115
| |
Collapse
|
136
|
Quan L, Yi J, Zhao Y, Zhang F, Shi XT, Feng Z, Miller HL. Plasma trimethylamine N-oxide, a gut microbe-generated phosphatidylcholine metabolite, is associated with autism spectrum disorders. Neurotoxicology 2020; 76:93-98. [PMID: 31704102 PMCID: PMC7385710 DOI: 10.1016/j.neuro.2019.10.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 10/15/2019] [Accepted: 10/28/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVE The compositions of the gut microbiota and its metabolites were altered in individuals with Autism Spectrum Disorder (ASD). The aim of this study was to assess whether plasma levels of gut-derived metabolite trimethylamine N-oxide (TMAO) were associated with ASD and the degree of symptom severity. METHODS From September 2017 to January 2019, a total of three hundred and twenty-eight Chinese children (164 with ASD and 164 their age-sex matched control subjects) aged 3-8 years were included. TMAO levels in plasma were determined using high-performance liquid chromatography tandem mass spectrometry (LC/MS/MS). Logistic regression analysis was used to examine the TMAO-ASD association. RESULTS In the study, the median age of the ASD group was 5 years (interquartile range [IQR], 4-6 years) and 129 (78.7%) were boys. The median plasma levels of TMAO in children with ASD and typically-developing (TD) children at admission were 4.2 (IQR, 3.0-5.6) μmol/l and 3.0 (2.0-4.4) μmol/l, respectively (P < 0.001). For each 1 μmol/l increase of plasma TMAO, the unadjusted and adjusted risk of ASD would be increased by 54% (with the odds ratios [OR] of 1.54; 95% confidence intervals [CI]: 1.32-1.78; P < 0.001) and 27% (1.27 [1.10-1.45], P < 0.001), respectively. Symptom severity was classified as mild-to-moderate (CARS < 37) for 66 children with ASD (40.2%). In these children, the plasma levels of TMAO were lower than in the 98 children with ASD (59.8%) whose symptoms were classified as severe (CARS > 36) (3.5[2.5-4.9] μmol/l vs. 4.5(3.7-6.0) μmol/l; P < 0.001). For each 1 μmol/l increase of plasma TMAO, the unadjusted and adjusted risk of severe autism would be increased by 61% (with the OR of 1.61 [95% CI 1.28-2.01], P < 0.001) and 31% (1.31 [1.08-1.49], P < 0.001), respectively. CONCLUSIONS Elevated plasma levels of TMAO were associated with ASD and symptom severity.
Collapse
Affiliation(s)
- Lijuan Quan
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China.
| | - Jinping Yi
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yue Zhao
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Feng Zhang
- Department of Pediatric Neurology and Rehabilitation, Maternal and Child Health Hospital of Ganzhou, Ganzhou, China
| | - Xiao-Tong Shi
- Autism Treatment Center, Rehabilitation Medical College of Capital Medical University, Beijing, China
| | - Zhen Feng
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Haylie L Miller
- Department of Physical Therapy, University of North Texas Health Science Center, Fort Worth, USA.
| |
Collapse
|
137
|
Larijani B, Foroughi Heravani N, Alavi-Moghadam S, Goodarzi P, Rezaei-Tavirani M, Payab M, Gholami M, Razi F, Arjmand B. Cell Therapy Targets for Autism Spectrum Disorders: Hopes, Challenges and Future Directions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1341:107-124. [PMID: 32072476 DOI: 10.1007/5584_2020_491] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Autism spectrum disorders as a group of pediatric neurodevelopmental diseases is a crucial part of the worldwide disabilities which have influence in communication skills, social interactions, and ability to understand the concepts. The precise pathophysiology of autism spectrum disorders due to the abundance of involved mechanisms is unknown. Some of these involved mechanisms are related to genetic factors, chronic neuro inflammation, mitochondrial dysfunction, oxidative stress, immune dysregulation, hormonal imbalance, and environmental factors. Current main treatments for autisms are behavioral, nutritional and medical therapies, however there is not definitive treatment approach. Therein, more novel therapies are still required to improve the symptoms. Several preclinical and clinical evidence were shown that stem cell therapy is a potential treatment option for autism spectrum disorders individuals. Considering the significant factors which can affect the outcome of stem cell therapeutic effects including stem cell types, route and dosage of administration, and mechanism of activity along with selecting best animal models can be very important in performing clinical trials.
Collapse
Affiliation(s)
- Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Najmeh Foroughi Heravani
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Alavi-Moghadam
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Parisa Goodarzi
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Moloud Payab
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Gholami
- Department of Toxicology & Pharmacology, Faculty of Pharmacy; Toxicology and Poisoning Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Farideh Razi
- Diabetes Research Center, Endocrinology and Metabolism Clinical Siences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
138
|
Endres D, Rauer S, Pschibul A, Süß P, Venhoff N, Runge K, Feige B, Denzel D, Nickel K, Schweizer T, Maier S, Egger K, Domschke K, Meyer PT, Prüss H, Tebartz van Elst L. Novel Antineuronal Autoantibodies With Somatodendritic Staining Pattern in a Patient With Autoimmune Psychosis. Front Psychiatry 2020; 11:627. [PMID: 32848899 PMCID: PMC7424063 DOI: 10.3389/fpsyt.2020.00627] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 06/15/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Autoimmune encephalitis, such as anti-NMDA-receptor encephalitis, typically presenting with subacute onset of neuropsychiatric symptoms, can be detected by antineuronal autoantibodies or inflammatory changes in the cerebrospinal fluid (CSF), as well as pathological alterations in electroencephalography (EEG), magnetic resonance imaging (MRI), or [18F]fluorodeoxyglucose positron emission tomography (FDG PET). For patients with predominant psychotic symptoms, the term autoimmune psychosis was proposed. Here, the authors present the case of a patient with probable autoimmune psychosis associated with unknown antineuronal antibodies. CASE PRESENTATION A 18-year-old male patient with preexisting autism spectrum disorder developed a severe catatonic syndrome over 2.5 years. The MRI showed normal findings, the EEG depicted intermittent slowing, and the independent component analyses showed additional sharp spikes. However, FDG PET, the basic laboratory analysis and testing of the serum/CSF for well-characterized antineuronal autoantibodies were unsuspicious. The serum and CSF "tissue-based assay" using indirect immunofluorescence on unfixed murine brain tissue revealed antineuronal autoantibodies against an unknown epitope in granule cells in the cerebellum and to neurites of hippocampal interneurons with a somatodendritic staining pattern. The immunosuppressive treatment with high-dose glucocorticoids, plasma exchange, and rituximab led to partial improvement. CONCLUSION The patient probably suffered from autoantibody-associated autoimmune psychosis. The special features of the case were that the patient (1) presented with mostly inconspicuous basic diagnostics, except for the altered EEG in combination with the detection of CSF autoantibodies directed against a currently unknown epitope, (2) experienced an isolated and long-lasting psychotic course, and (3) had pre-existing autism spectrum disorder. The detection of a probable autoimmune pathophysiology in such cases seems important, as it offers new and more causal immunosuppressive treatment alternatives.
Collapse
Affiliation(s)
- Dominique Endres
- Section for Experimental Neuropsychiatry, Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sebastian Rauer
- Department of Neurology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Alexander Pschibul
- Department of Neuropediatrics and Muscle Disorders, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Patrick Süß
- Department of Molecular Neurology, University Hospital Erlangen, Erlangen, Germany
| | - Nils Venhoff
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Kimon Runge
- Section for Experimental Neuropsychiatry, Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Bernd Feige
- Section for Experimental Neuropsychiatry, Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dominik Denzel
- Section for Experimental Neuropsychiatry, Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Kathrin Nickel
- Section for Experimental Neuropsychiatry, Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tina Schweizer
- Section for Experimental Neuropsychiatry, Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Simon Maier
- Section for Experimental Neuropsychiatry, Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Karl Egger
- Department of Neuroradiology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Katharina Domschke
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Basics in Neuromodulation, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Philipp T Meyer
- Department of Nuclear Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Harald Prüss
- Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany
| | - Ludger Tebartz van Elst
- Section for Experimental Neuropsychiatry, Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
139
|
Hodges SL, Nolan SO, Tomac LA, Muhammad IDA, Binder MS, Taube JH, Lugo JN. Lipopolysaccharide-induced inflammation leads to acute elevations in pro-inflammatory cytokine expression in a mouse model of Fragile X syndrome. Physiol Behav 2019; 215:112776. [PMID: 31838149 DOI: 10.1016/j.physbeh.2019.112776] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/11/2019] [Accepted: 12/11/2019] [Indexed: 02/07/2023]
Abstract
Fragile X syndrome (FXS) is a neurodevelopmental disorder caused by a single genetic mutation in the Fmr1 gene, serving as the largest genetic cause of intellectual disability. Trinucleotide expansion mutations in Fmr1 result in silencing and hypermethylation of the gene, preventing synthesis of the RNA binding protein Fragile X mental retardation protein which functions as a translational repressor. Abnormal immune responses have been demonstrated to play a role in FXS pathophysiology, however, whether these alterations impact how those with FXS respond to an immune insult behaviorally is not entirely known. In the current study, we examine how Fmr1 knockout (KO) and wild type (WT) mice respond to the innate immune stimulus lipopolysaccharide (LPS), both on a molecular and behavioral level, to determine if Fmr1 mutations impact the normal physiological response to an immune insult. In response to LPS, Fmr1 KO mice had elevated hippocampal IL-1β and IL-6 mRNA levels 4 h post-treatment compared to WT mice, with no differences detected in any cytokines at baseline or between genotypes 24 h post-LPS administration. Fmr1 KO mice also had upregulated hippocampal BDNF gene expression 4 h post-treatment compared to WT mice, which was not dependent on LPS administration. There were no differences in hippocampal protein expression between genotypes in microglia (Iba1) or astrocyte (GFAP) reactivity. Further, both genotypes displayed the typical sickness response following LPS stimulation, demonstrated by a significant reduction in food burrowed by LPS-treated mice in a burrowing task. Additional investigation is critical to determine if the transient increases in cytokine expression could lead to long-term changes in downstream molecular signaling in FXS.
Collapse
Affiliation(s)
- Samantha L Hodges
- Institute of Biomedical Studies, Baylor University, Waco, TX 76798, USA
| | - Suzanne O Nolan
- Department of Psychology and Neuroscience, Baylor University, One Bear Place # 97334, Waco, TX 76798, USA
| | - Lindsay A Tomac
- Department of Psychology and Neuroscience, Baylor University, One Bear Place # 97334, Waco, TX 76798, USA
| | - Ilyasah D A Muhammad
- Department of Psychology and Neuroscience, Baylor University, One Bear Place # 97334, Waco, TX 76798, USA
| | - Matthew S Binder
- Department of Psychology and Neuroscience, Baylor University, One Bear Place # 97334, Waco, TX 76798, USA
| | - Joseph H Taube
- Institute of Biomedical Studies, Baylor University, Waco, TX 76798, USA; Department of Biology, Baylor University, Waco, TX 76798, USA
| | - Joaquin N Lugo
- Institute of Biomedical Studies, Baylor University, Waco, TX 76798, USA; Department of Psychology and Neuroscience, Baylor University, One Bear Place # 97334, Waco, TX 76798, USA; Department of Biology, Baylor University, Waco, TX 76798, USA.
| |
Collapse
|
140
|
Shin C, Kim YK. Autoimmunity in microbiome-mediated diseases and novel therapeutic approaches. Curr Opin Pharmacol 2019; 49:34-42. [DOI: 10.1016/j.coph.2019.04.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 04/25/2019] [Indexed: 12/30/2022]
|
141
|
Salloum-Asfar S, Satheesh NJ, Abdulla SA. Circulating miRNAs, Small but Promising Biomarkers for Autism Spectrum Disorder. Front Mol Neurosci 2019; 12:253. [PMID: 31680857 PMCID: PMC6808050 DOI: 10.3389/fnmol.2019.00253] [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: 06/17/2019] [Accepted: 09/30/2019] [Indexed: 12/14/2022] Open
Abstract
Autism spectrum disorder (ASD) refers to a heterogeneous group of complex neurodevelopmental disorders characterized by social skill and communication deficits, along with stereotyped repetitive behavior. miRNAs, small non-coding RNAs that have been recognized as critical regulators of gene expression, play a key role in the neurodevelopmental transcriptional networks of the human brain. Previous investigations have proven that circulating miRNAs open up new possibilities for the emerging roles of diagnostic and prognostic biomarkers in human disorders and diseases. Biomarker development has been progressively becoming more recognized as a cornerstone in medical diagnosis, paving the way to drug discoveries and limiting the progression of various diseases. Due to the complexity of ASD, considerable endeavors have either unsuccessfully identified biomarkers for the disorder or have not yet been established. Cell-free circulating miRNAs in biofluids are extraordinarily stable and considered to represent the next-generation of clinical, non-invasive, biomarkers for many pathologies including neurological and neurodevelopmental disorders. Here, we conducted a review of all peer-reviewed articles addressing the circulating profiles of miRNAs, mostly performed in serum and saliva samples in individuals with ASD.
Collapse
Affiliation(s)
- Salam Salloum-Asfar
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Noothan J Satheesh
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Sara A Abdulla
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| |
Collapse
|
142
|
Tamouza R, Fernell E, Eriksson MA, Anderlid BM, Manier C, Mariaselvam CM, Boukouaci W, Leboyer M, Gillberg C. HLA Polymorphism in Regressive and Non-Regressive Autism: A Preliminary Study. Autism Res 2019; 13:182-186. [PMID: 31593375 DOI: 10.1002/aur.2217] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 08/27/2019] [Accepted: 09/12/2019] [Indexed: 12/16/2022]
Abstract
Autism spectrum disorders (ASD) comprises heterogeneous neurodevelopmental conditions with symptom onset usually during infancy. However, about 10%-30% of affected cases experience a loss of language and social skills around 18-30 months, so-called regressive autism. In this subset with regression, immune dysfunctions including inflammation and autoimmunity have been proposed to be at risk factors. Given the implication of the human histocompatibility antigens (HLA) system in various aspects of immune responses, including autoimmunity, and in ASD, we investigate here the distribution of the HLA Class I and Class II haplotypes in 131 children with ASD meeting DSM-IV TR criteria, with and without regression. We found that 62 of the 98 non-regressive ASD patients carry the HLA-DPA1*01-DPB1*04 sub-haplotype as compared to 14 of the 33 patients with regression (63% vs. 43% respectively, Pc = 0.02), suggesting that this HLA haplotype may exert a protective effect against regression. Similarly, the HLA-DPA1*01-DPB1*04 has also been found to be more represented in healthy controls as compared to patients affected with common nonpsychiatric autoimmune disorders. Overall our findings suggest a possible involvement of HLA polymorphism in the context of regressive ASD. Autism Res 2020, 13: 182-186. © 2019 The Authors. Autism Research published by International Society for Autism Research published by Wiley Periodicals, Inc. LAY SUMMARY: Immune dysfunctions including inflammatory and autoimmune processes have been reported in autism, particularly in regressive forms. In this study, we analyzed the distribution of HLA haplotypes among children with autism spectrum disorder (ASD), with and without regression from Sweden and observed that HLA-DPA1*01-DPB1*04 sub-haplotype was less represented in patients with regressive autism as compared with those without regression. Such possible protective effect, also observed in other common autoimmune disorders, may constitute a link between HLA-mediated immune processes and regressive ASD.
Collapse
Affiliation(s)
- Ryad Tamouza
- Translational Psychiatry Laboratory, Mondor Institute of Biomedical Research, INSERM U955, Créteil, France.,DHU PePSY, AP-HP, Pôle de Psychiatrie, Hôpitaux Universitaires Henri Mondor, Université Paris-Est-Créteil, Créteil, France.,Fondation FondaMental, Créteil, France
| | - Elisabeth Fernell
- Gillberg Neuropsychiatry Centre, Institute of Neuroscience and Physiology, University of Gothenburg, Sahlgrenska Academy, Gothenburg, Sweden.,Sahlgren University Hospital, Gothenburg, Sweden
| | - Mats Anders Eriksson
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Britt-Marie Anderlid
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| | - Céline Manier
- Translational Psychiatry Laboratory, Mondor Institute of Biomedical Research, INSERM U955, Créteil, France
| | - Christina Mary Mariaselvam
- Translational Psychiatry Laboratory, Mondor Institute of Biomedical Research, INSERM U955, Créteil, France
| | - Wahid Boukouaci
- Translational Psychiatry Laboratory, Mondor Institute of Biomedical Research, INSERM U955, Créteil, France
| | - Marion Leboyer
- Translational Psychiatry Laboratory, Mondor Institute of Biomedical Research, INSERM U955, Créteil, France.,DHU PePSY, AP-HP, Pôle de Psychiatrie, Hôpitaux Universitaires Henri Mondor, Université Paris-Est-Créteil, Créteil, France.,Fondation FondaMental, Créteil, France
| | - Christopher Gillberg
- Gillberg Neuropsychiatry Centre, Institute of Neuroscience and Physiology, University of Gothenburg, Sahlgrenska Academy, Gothenburg, Sweden.,Sahlgren University Hospital, Gothenburg, Sweden
| |
Collapse
|
143
|
Glinton KE, Elsea SH. Untargeted Metabolomics for Autism Spectrum Disorders: Current Status and Future Directions. Front Psychiatry 2019; 10:647. [PMID: 31551836 PMCID: PMC6746843 DOI: 10.3389/fpsyt.2019.00647] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 08/12/2019] [Indexed: 12/20/2022] Open
Abstract
Autism spectrum disorders (ASDs) are a group of neurodevelopment disorders characterized by childhood onset deficits in social communication and interaction. Although the exact etiology of most cases of ASDs is unknown, a portion has been proposed to be associated with various metabolic abnormalities including mitochondrial dysfunction, disorders of cholesterol metabolism, and folate abnormalities. Targeted biochemical testing like plasma amino acid and acylcarnitine profiles have demonstrated limited utility in helping to diagnose and manage such patients. Untargeted metabolomics has emerged, however, as a promising tool in screening for underlying biochemical abnormalities and managing treatment and as a means of investigating possible novel biomarkers for the disorder. Here, we review the principles and methodology behind untargeted metabolomics, recent pilot studies utilizing this technology, and areas in which it may be integrated into the care of children with this disorder in the future.
Collapse
Affiliation(s)
- Kevin E. Glinton
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
| | - Sarah H. Elsea
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
144
|
Luoto S, Krams I, Rantala MJ. Response to Commentaries: Life History Evolution, Causal Mechanisms, and Female Sexual Orientation. ARCHIVES OF SEXUAL BEHAVIOR 2019; 48:1335-1347. [PMID: 31119422 DOI: 10.1007/s10508-019-1439-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 03/16/2019] [Indexed: 06/09/2023]
Affiliation(s)
- Severi Luoto
- English, Drama and Writing Studies, University of Auckland, Arts 1, Bldg. 206, Room 616, 14A Symonds St., Auckland, 1010, New Zealand.
- School of Psychology, University of Auckland, Auckland, New Zealand.
| | - Indrikis Krams
- Department of Zoology and Animal Ecology, University of Latvia, Rīga, Latvia
- Institute of Ecology and Earth Sciences, University of Tartu, Tartu, Estonia
| | | |
Collapse
|
145
|
Frye RE. Behaviorally associated changes in neuroconnectivity following autologous umbilical cord blood infusion in young children with autism spectrum disorder. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:S123. [PMID: 31576330 PMCID: PMC6685876 DOI: 10.21037/atm.2019.05.55] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 05/21/2019] [Indexed: 01/05/2024]
Affiliation(s)
- Richard E. Frye
- Division of Neurodevelopmental Disorders, Department of Neurology, Barrow Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ, USA
- Department of Child Health, University of Arizona College of Medicine, Phoenix, AZ, USA
| |
Collapse
|
146
|
Rose D, Ashwood P. Rapid Communication: Plasma Interleukin-35 in Children with Autism. Brain Sci 2019; 9:E152. [PMID: 31252635 PMCID: PMC6680732 DOI: 10.3390/brainsci9070152] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 06/21/2019] [Accepted: 06/24/2019] [Indexed: 12/29/2022] Open
Abstract
In autism spectrum disorders (ASD) many individuals have co-morbid immune dysregulation that can lead to inflammation in the brain and periphery. The novel cytokine interleukin (IL)-35 has described anti-inflammatory properties; however, the plasma levels of IL-35 in children with ASD have never been investigated. The plasma levels of IL-35 were measured by an enzyme-linked immunosorbent assay in 30 children with ASD and 39 typically developing (TD) controls. In the current study, we found that plasma IL-35 levels were significantly decreased in children with ASD compared with TD children. Furthermore, lower IL-35 levels were associated with worse behaviors as assessed using the aberrant behavior checklist. These findings are in line with other observations of decreased regulatory cytokines such as transforming growth factor beta and IL-10 in ASD, and associations with severity of behaviors. In conclusion, regulating the expression of IL-35 may provide a new possible target for the treatment of immune issues in ASD to address an imbalance between pro- and anti-inflammatory signals that alter the behavioral phenotype.
Collapse
Affiliation(s)
- Destanie Rose
- Department of Medical Microbiology and Immunology, and The Medical Investigation of Neurodevelopmental Disorders Institute, University of California, Davis, CA 95817, USA
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, and The Medical Investigation of Neurodevelopmental Disorders Institute, University of California, Davis, CA 95817, USA.
| |
Collapse
|
147
|
Vargason T, Frye RE, McGuinness DL, Hahn J. Clustering of co-occurring conditions in autism spectrum disorder during early childhood: A retrospective analysis of medical claims data. Autism Res 2019; 12:1272-1285. [PMID: 31149786 DOI: 10.1002/aur.2128] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 03/20/2019] [Accepted: 05/05/2019] [Indexed: 12/18/2022]
Abstract
Individuals with autism spectrum disorder (ASD) are frequently affected by co-occurring medical conditions (COCs), which vary in severity, age of onset, and pathophysiological characteristics. The presence of COCs contributes to significant heterogeneity in the clinical presentation of ASD between individuals and a better understanding of COCs may offer greater insight into the etiology of ASD in specific subgroups while also providing guidance for diagnostic and treatment protocols. This study retrospectively analyzed medical claims data from a private United States health plan between years 2000 and 2015 to investigate patterns of COC diagnoses in a cohort of 3,278 children with ASD throughout their first 5 years of enrollment compared to 279,693 children from the general population without ASD diagnoses (POP cohort). Three subgroups of children with ASD were identified by k-means clustering using these COC patterns. The first cluster was characterized by generally high rates of COC diagnosis and comprised 23.7% (n = 776) of the cohort. Diagnoses of developmental delays were dominant in the second cluster containing 26.5% (n = 870) of the cohort. Children in the third cluster, making up 49.8% (n = 1,632) of the cohort, had the lowest rates of COC diagnosis, which were slightly higher than rates observed in the POP cohort. A secondary analysis using these data found that gastrointestinal and immune disorders showed similar longitudinal patterns of prevalence, as did seizure and sleep disorders. These findings may help to better inform the development of diagnostic workup and treatment protocols for COCs in children with ASD. Autism Res 2019, 12: 1272-1285. © 2019 International Society for Autism Research, Wiley Periodicals, Inc. LAY SUMMARY: Medical conditions that co-occur with autism spectrum disorder (ASD) vary significantly from person to person. This study analyzed patterns in diagnosis of co-occurring conditions from medical claims data and observed three subtypes of children with ASD. These results may aid with screening for co-occurring conditions in children with ASD and with understanding ASD subtypes.
Collapse
Affiliation(s)
- Troy Vargason
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York.,Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York.,OptumLabs Visiting Fellow, Cambridge, Massachusetts
| | - Richard E Frye
- Department of Child Health, University of Arizona College of Medicine, Phoenix, Arizona.,Phoenix Children's Hospital, Phoenix, Arizona
| | - Deborah L McGuinness
- Department of Computer Science, Rensselaer Polytechnic Institute, Troy, New York.,Department of Cognitive Science, Rensselaer Polytechnic Institute, Troy, New York
| | - Juergen Hahn
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York.,Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York.,Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, New York
| |
Collapse
|
148
|
Sauer AK, Bockmann J, Steinestel K, Boeckers TM, Grabrucker AM. Altered Intestinal Morphology and Microbiota Composition in the Autism Spectrum Disorders Associated SHANK3 Mouse Model. Int J Mol Sci 2019; 20:ijms20092134. [PMID: 31052177 PMCID: PMC6540607 DOI: 10.3390/ijms20092134] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 04/26/2019] [Accepted: 04/28/2019] [Indexed: 02/07/2023] Open
Abstract
Autism spectrum disorders (ASD) are a group of neurodevelopmental disorders characterized by deficits in social interaction and communication, and repetitive behaviors. In addition, co-morbidities such as gastro-intestinal problems have frequently been reported. Mutations and deletion of proteins of the SH3 and multiple ankyrin repeat domains (SHANK) gene-family were identified in patients with ASD, and Shank knock-out mouse models display autism-like phenotypes. SHANK3 proteins are not only expressed in the central nervous system (CNS). Here, we show expression in gastrointestinal (GI) epithelium and report a significantly different GI morphology in Shank3 knock-out (KO) mice. Further, we detected a significantly altered microbiota composition measured in feces of Shank3 KO mice that may contribute to inflammatory responses affecting brain development. In line with this, we found higher E. coli lipopolysaccharide levels in liver samples of Shank3 KO mice, and detected an increase in Interleukin-6 and activated astrocytes in Shank3 KO mice. We conclude that apart from its well-known role in the CNS, SHANK3 plays a specific role in the GI tract that may contribute to the ASD phenotype by extracerebral mechanisms.
Collapse
Affiliation(s)
- Ann Katrin Sauer
- Cellular Neurobiology and Neuro-Nanotechnology lab, Dept. of Biological Sciences, University of Limerick, V94PH61 Limerick, Ireland.
| | - Juergen Bockmann
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany.
| | - Konrad Steinestel
- Gerhard-Domagk-Institute of Pathology, Muenster University Medical Center, 48149 Münster, Germany.
| | - Tobias M Boeckers
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany.
| | - Andreas M Grabrucker
- Cellular Neurobiology and Neuro-Nanotechnology lab, Dept. of Biological Sciences, University of Limerick, V94PH61 Limerick, Ireland.
- Health Research Institute (HRI), University of Limerick, V94PH61 Limerick, Ireland.
- Bernal Institute, University of Limerick, V94PH61 Limerick, Ireland.
| |
Collapse
|
149
|
Kovács Z, D'Agostino DP, Diamond D, Kindy MS, Rogers C, Ari C. Therapeutic Potential of Exogenous Ketone Supplement Induced Ketosis in the Treatment of Psychiatric Disorders: Review of Current Literature. Front Psychiatry 2019; 10:363. [PMID: 31178772 PMCID: PMC6543248 DOI: 10.3389/fpsyt.2019.00363] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 05/10/2019] [Indexed: 12/16/2022] Open
Abstract
Globally, psychiatric disorders, such as anxiety disorder, bipolar disorder, schizophrenia, depression, autism spectrum disorder, and attention-deficit/hyperactivity disorder (ADHD) are becoming more prevalent. Although the exact pathological alterations are not yet clear, recent studies have demonstrated that widespread changes of very complex metabolic pathways may partially underlie the pathophysiology of many psychiatric diseases. Thus, more attention should be directed to metabolic-based therapeutic interventions in the treatment of psychiatric disorders. Emerging evidence from numerous studies suggests that administration of exogenous ketone supplements, such as ketone salts or ketone esters, generates rapid and sustained nutritional ketosis and metabolic changes, which may evoke potential therapeutic effects in cases of central nervous system (CNS) disorders, including psychiatric diseases. Therefore, the aim of this review is to summarize the current information on ketone supplementation as a potential therapeutic tool for psychiatric disorders. Ketone supplementation elevates blood levels of the ketone bodies: D-β-hydroxybutyrate (βHB), acetoacetate (AcAc), and acetone. These compounds, either directly or indirectly, beneficially affect the mitochondria, glycolysis, neurotransmitter levels, activity of free fatty acid receptor 3 (FFAR3), hydroxycarboxylic acid receptor 2 (HCAR2), and histone deacetylase, as well as functioning of NOD-like receptor pyrin domain 3 (NLRP3) inflammasome and mitochondrial uncoupling protein (UCP) expression. The result of downstream cellular and molecular changes is a reduction in the pathophysiology associated with various psychiatric disorders. We conclude that supplement-induced nutritional ketosis leads to metabolic changes and improvements, for example, in mitochondrial function and inflammatory processes, and suggest that development of specific adjunctive ketogenic protocols for psychiatric diseases should be actively pursued.
Collapse
Affiliation(s)
- Zsolt Kovács
- Savaria Department of Biology, ELTE Eötvös Loránd University, Savaria University Centre, Szombathely, Hungary
| | - Dominic P D'Agostino
- Department of Molecular Pharmacology and Physiology, Laboratory of Metabolic Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States.,Institute for Human and Machine Cognition, Ocala, FL, United States
| | - David Diamond
- Department of Molecular Pharmacology and Physiology, Laboratory of Metabolic Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States.,Department of Psychology, Hyperbaric Neuroscience Research Laboratory, University of South Florida, Tampa, FL, United States
| | - Mark S Kindy
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, United States.,James A. Haley VA Medical Center, Tampa, FL, United States.,Shriners Hospital for Children, Tampa, FL, United States
| | - Christopher Rogers
- Department of Molecular Pharmacology and Physiology, Laboratory of Metabolic Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Csilla Ari
- Department of Psychology, Hyperbaric Neuroscience Research Laboratory, University of South Florida, Tampa, FL, United States
| |
Collapse
|
150
|
Tye C, Runicles AK, Whitehouse AJO, Alvares GA. Characterizing the Interplay Between Autism Spectrum Disorder and Comorbid Medical Conditions: An Integrative Review. Front Psychiatry 2018; 9:751. [PMID: 30733689 PMCID: PMC6354568 DOI: 10.3389/fpsyt.2018.00751] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 12/19/2018] [Indexed: 12/18/2022] Open
Abstract
Co-occurring medical disorders and associated physiological abnormalities in individuals with autism spectrum disorder (ASD) may provide insight into causal pathways or underlying biological mechanisms. Here, we review medical conditions that have been repeatedly highlighted as sharing the strongest associations with ASD-epilepsy, sleep, as well as gastrointestinal and immune functioning. We describe within each condition their prevalence, associations with behavior, and evidence for successful treatment. We additionally discuss research aiming to uncover potential aetiological mechanisms. We then consider the potential interaction between each group of conditions and ASD and, based on the available evidence, propose a model that integrates these medical comorbidities in relation to potential shared aetiological mechanisms. Future research should aim to systematically examine the interactions between these physiological systems, rather than considering these in isolation, using robust and sensitive biomarkers across an individual's development. A consideration of the overlap between medical conditions and ASD may aid in defining biological subtypes within ASD and in the development of specific targeted interventions.
Collapse
Affiliation(s)
- Charlotte Tye
- Child & Adolescent Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Abigail K Runicles
- Child & Adolescent Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Andrew J O Whitehouse
- Telethon Kids Institute, University of Western Australia, Perth, WA, Australia.,Cooperative Research Centre for Living with Autism (Autism CRC), Brisbane, QLD, Australia
| | - Gail A Alvares
- Telethon Kids Institute, University of Western Australia, Perth, WA, Australia.,Cooperative Research Centre for Living with Autism (Autism CRC), Brisbane, QLD, Australia
| |
Collapse
|