101
|
Bjørklund G, Skalny AV, Rahman MM, Dadar M, Yassa HA, Aaseth J, Chirumbolo S, Skalnaya MG, Tinkov AA. Toxic metal(loid)-based pollutants and their possible role in autism spectrum disorder. ENVIRONMENTAL RESEARCH 2018; 166:234-250. [PMID: 29902778 DOI: 10.1016/j.envres.2018.05.020] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 05/18/2018] [Accepted: 05/18/2018] [Indexed: 06/08/2023]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by deficits in social interaction, verbal and non-verbal communication, and stereotypic behaviors. Many studies support a significant relationship between many different environmental factors in ASD etiology. These factors include increased daily exposure to various toxic metal-based environmental pollutants, which represent a cause for concern in public health. This article reviews the most relevant toxic metals, commonly found, environmental pollutants, i.e., lead (Pb), mercury (Hg), aluminum (Al), and the metalloid arsenic (As). Additionally, it discusses how pollutants can be a possible pathogenetic cause of ASD through various mechanisms including neuroinflammation in different regions of the brain, fundamentally occurring through elevation of the proinflammatory profile of cytokines and aberrant expression of nuclear factor kappa B (NF-κB). Due to the worldwide increase in toxic environmental pollution, studies on the role of pollutants in neurodevelopmental disorders, including direct effects on the developing brain and the subjects' genetic susceptibility and polymorphism, are of utmost importance to achieve the best therapeutic approach and preventive strategies.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine, Mo i Rana, Norway.
| | - Anatoly V Skalny
- Peoples' Friendship University of Russia (RUDN University), Moscow, Russia; Yaroslavl State University, Yaroslavl, Russia; All-Russian Research Institute of Medicinal and Aromatic Plants, Moscow, Russia
| | - Md Mostafizur Rahman
- Department of Environmental Sciences, Jahangirnagar University, Dhaka, Bangladesh; Graduate School of Environmental Science, Hokkaido University, Japan
| | - Maryam Dadar
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Heba A Yassa
- Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Jan Aaseth
- Faculty of Health and Social Sciences, Inland Norway University of Applied Sciences, Elverum, Norway; Department of Research, Innlandet Hospital Trust, Brumunddal, Norway
| | - Salvatore Chirumbolo
- Department of Neurological and Movement Sciences, University of Verona, Verona, Italy
| | | | - Alexey A Tinkov
- Peoples' Friendship University of Russia (RUDN University), Moscow, Russia; Yaroslavl State University, Yaroslavl, Russia
| |
Collapse
|
102
|
Ahmad SF, Ansari MA, Nadeem A, Bakheet SA, Alshammari MA, Attia SM. Protection by tyrosine kinase inhibitor, tyrphostin AG126, through the suppression of IL-17A, RORγt, and T-bet signaling, in the BTBR mouse model of autism. Brain Res Bull 2018; 142:328-337. [DOI: 10.1016/j.brainresbull.2018.08.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 08/16/2018] [Accepted: 08/27/2018] [Indexed: 12/17/2022]
|
103
|
Hammond TR, Robinton D, Stevens B. Microglia and the Brain: Complementary Partners in Development and Disease. Annu Rev Cell Dev Biol 2018; 34:523-544. [PMID: 30089221 DOI: 10.1146/annurev-cellbio-100616-060509] [Citation(s) in RCA: 209] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
An explosion of findings driven by powerful new technologies has expanded our understanding of microglia, the resident immune cells of the central nervous system (CNS). This wave of discoveries has fueled a growing interest in the roles that these cells play in the development of the CNS and in the neuropathology of a diverse array of disorders. In this review, we discuss the crucial roles that microglia play in shaping the brain-from their influence on neurons and glia within the developing CNS to their roles in synaptic maturation and brain wiring-as well as some of the obstacles to overcome when assessing their contributions to normal brain development. Furthermore, we examine how normal developmental functions of microglia are perturbed or remerge in neurodevelopmental and neurodegenerative disease.
Collapse
Affiliation(s)
- Timothy R Hammond
- FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA;
| | - Daisy Robinton
- FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA;
| | - Beth Stevens
- FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA; .,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
| |
Collapse
|
104
|
DeWitt JC, Patisaul HB. Endocrine disruptors and the developing immune system. CURRENT OPINION IN TOXICOLOGY 2018. [DOI: 10.1016/j.cotox.2017.12.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
105
|
Abstract
Autism Spectrum Disorder (ASD) refers to a group of neurodevelopmental disorders including autism, Asperger's syndrome (AS) and pervasive developmental disorder-not otherwise specified (PDD-NOS). The new diagnostic criteria of ASD focuses on two core domains: social communication impairment and restricted interests/repetitive behaviors. The prevalence of ASD has been steadily increasing over the past two decades, with current estimates reaching up to 1 in 36 children. Hereditary factors, parental history of psychiatric disorders, pre-term births, and fetal exposure to psychotropic drugs or insecticides have all been linked to higher risk of ASD. Several scales such as the Childhood Autism Rating Scale (CARS), The Autism Spectrum Disorder-Observation for Children (ASD-OC), The Developmental, Dimensional, and Diagnostic Interview (3di), are available to aid in better assessing the behaviors and symptoms associated with ASD. Nearly 75% of ASD patients suffer from comorbid psychiatric illnesses or conditions, which may include attention-deficit hyperactivity disorder (ADHD), anxiety, bipolar disorder, depression, Tourette syndrome, and others. Both pharmacological and non-pharmacological interventions are available for ASD. Pharmacological treatments include psychostimulants, atypical antipsychotics, antidepressants, and alpha-2 adrenergic receptor agonists. These medications provide partial symptomatic relief of core symptoms of ASD or manage the symptoms of comorbid conditions. Non-pharmacological interventions, which show promising evidence in improving social interaction and verbal communication of ASD patients, include music therapy, cognitive behavioral therapy and social behavioral therapy. Hormonal therapies with oxytocyin or vasopressin receptor antagonists have also shown some promise in improving core ASD symptoms. The use of vitamins, herbal remedies and nutritional supplements in conjunction with pharmacological and behavioral treatment appear to have some effect in symptomatic improvement in ASD, though additional studies are needed to confirm these benefits. Developing novel disease-modifying therapies may prove to be the ultimate intervention for sustained improvement of symptoms in ASD.
Collapse
Affiliation(s)
- Samata R Sharma
- Department of Psychiatry, Harvard Medical School, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Xenia Gonda
- Department of Psychiatry and Psychotherapy, Kutvolgyi Clinical Centre, Semmelweis University, Kutvolgyi ut 4, 1125 Budapest, Hungary
| | - Frank I Tarazi
- Department of Psychiatry and Neuroscience Program, , Harvard Medical School, McLean Hospital, Belmont, MA 02478, USA.
| |
Collapse
|
106
|
Nadeem A, Ahmad SF, El-Sherbeeny AM, Al-Harbi NO, Bakheet SA, Attia SM. Systemic inflammation in asocial BTBR T + tf/J mice predisposes them to increased psoriatic inflammation. Prog Neuropsychopharmacol Biol Psychiatry 2018; 83:8-17. [PMID: 29287831 DOI: 10.1016/j.pnpbp.2017.12.015] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 12/12/2017] [Accepted: 12/24/2017] [Indexed: 01/09/2023]
Abstract
Autistic Spectrum disorder (ASD) is a neurobehavioral disorder characterized by defects in communication skills leading to restricted sociability. ASD has immense dysregulation in immune responses which is thought to affect neuronal system and thus behavior. ASD patients and BTBR T+ tf/J (BTBR) autistic mice have increased systemic inflammation due to dysfunction in innate and adaptive immune responses. Recent studies suggest that ASD patients are associated with several co-morbid autoimmune disorders including psoriasis. However underlying mechanisms for this phenomenon have not been explored. In this study, we used imiquimod (IMQ)-induced psoriatic inflammation in social C57BL/6 (C57) mice and asocial BTBR mice to investigate whether systemic inflammation in BTBR is associated with increased susceptibility to psoriatic inflammation. Our data shows that BTBR mice have increased expression of TLR7/IL-6/IL-23 in systemic DCs but not in skin as compared to C57 mice at baseline. This leads to much greater psoriatic inflammation in BTBR mice upon IMQ application than C57 mice. Consequently, BTBR mice also have higher Th17 related immune responses in the skin and systemic compartment. Overall our study suggests that systemic innate (TLR7/IL-23/IL-6 in DCs) and adaptive (Th17 related signaling) immune responses are heightened in BTBR mice at baseline which predisposes them for greater psoriatic inflammation than C57 mice upon IMQ application. This could be one of the reasons for increased psoriatic inflammation in patients with ASD. Therapies that aim to decrease immune activation may not only benefit ASD-associated neurobehavioral abnormalities but also comorbid disorders such as psoriasis.
Collapse
Affiliation(s)
- Ahmed Nadeem
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.
| | - Sheikh F Ahmad
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.
| | - Ahmed M El-Sherbeeny
- Industrial Engineering Department, College of Engineering, King Saud University, Riyadh, Saudi Arabia
| | - Naif O Al-Harbi
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Saleh A Bakheet
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
107
|
Farah R, Haraty H, Salame Z, Fares Y, Ojcius DM, Said Sadier N. Salivary biomarkers for the diagnosis and monitoring of neurological diseases. Biomed J 2018; 41:63-87. [PMID: 29866603 PMCID: PMC6138769 DOI: 10.1016/j.bj.2018.03.004] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 03/13/2018] [Accepted: 03/29/2018] [Indexed: 12/17/2022] Open
Abstract
Current research efforts on neurological diseases are focused on identifying novel disease biomarkers to aid in diagnosis, provide accurate prognostic information and monitor disease progression. With advances in detection and quantification methods in genomics, proteomics and metabolomics, saliva has emerged as a good source of samples for detection of disease biomarkers. Obtaining a sample of saliva offers multiple advantages over the currently tested biological fluids as it is a non-invasive, painless and simple procedure that does not require expert training or harbour undesirable side effects for the patients. Here, we review the existing literature on salivary biomarkers and examine their validity in diagnosing and monitoring neurodegenerative and neuropsychiatric disorders such as autism and Alzheimer's, Parkinson's and Huntington's disease. Based on the available research, amyloid beta peptide, tau protein, lactoferrin, alpha-synuclein, DJ-1 protein, chromogranin A, huntingtin protein, DNA methylation disruptions, and micro-RNA profiles provide display a reliable degree of consistency and validity as disease biomarkers.
Collapse
Affiliation(s)
- Raymond Farah
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Hayat Haraty
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Ziad Salame
- Research Department, Faculty of Dental Medicine, Lebanese University, Beirut, Lebanon
| | - Youssef Fares
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - David M Ojcius
- Department of Biomedical Sciences, University of the Pacific, Arthur Dugoni School of Dentistry, San Francisco, CA, USA.
| | - Najwane Said Sadier
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon.
| |
Collapse
|
108
|
Chez M, Lepage C, Parise C, Dang‐Chu A, Hankins A, Carroll M. Safety and Observations from a Placebo-Controlled, Crossover Study to Assess Use of Autologous Umbilical Cord Blood Stem Cells to Improve Symptoms in Children with Autism. Stem Cells Transl Med 2018; 7:333-341. [PMID: 29405603 PMCID: PMC5866927 DOI: 10.1002/sctm.17-0042] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 11/30/2017] [Accepted: 12/21/2017] [Indexed: 12/30/2022] Open
Abstract
The aim of this exploratory study was to assess the safety and clinical effects of autologous umbilical cord blood (AUCB) infusion in children with idiopathic autism spectrum disorder (ASD). Twenty-nine children 2 to 6 years of age with a confirmed diagnosis of ASD participated in this randomized, blinded, placebo-controlled, crossover trial. Participants were randomized to receive AUCB or placebo, evaluated at baseline, 12, and 24 weeks, received the opposite infusion, then re-evaluated at the same time points. Evaluations included assessments of safety, Expressive One Word Picture Vocabulary Test, 4th edition, Receptive One Word Picture Vocabulary Test, 4th edition, Clinical Global Impression, Stanford-Binet Fluid Reasoning and Knowledge, and the Vineland Adaptive Behavior and Socialization Subscales. Generalized linear models were used to assess the effects of the response variables at the 12- and 24-week time periods under each condition (AUCB, placebo). There were no serious adverse events. There were trends toward improvement, particularly in socialization, but there were no statistically significant differences for any endpoints. The results of this study suggest that autologous umbilical cord infusions are safe for children with ASD. Tightly controlled trials are necessary to further progress the study of AUCB for autism. Stem Cells Translational Medicine 2018;7:333-341.
Collapse
Affiliation(s)
- Michael Chez
- Pediatric NeuroscienceSacramentoCaliforniaUSA
- Sutter Institute for Medical Research (SIMR)SacramentoCaliforniaUSA
| | - Christopher Lepage
- Pediatric NeuroscienceSacramentoCaliforniaUSA
- Sutter Institute for Medical Research (SIMR)SacramentoCaliforniaUSA
| | - Carol Parise
- Sutter Institute for Medical Research (SIMR)SacramentoCaliforniaUSA
| | - Ashley Dang‐Chu
- Sutter Institute for Medical Research (SIMR)SacramentoCaliforniaUSA
| | - Andrea Hankins
- Sutter Institute for Medical Research (SIMR)SacramentoCaliforniaUSA
| | - Michael Carroll
- Bone Marrow Transplant, Sutter Medical GroupSacramentoCaliforniaUSA
| |
Collapse
|
109
|
Doenyas C. Gut Microbiota, Inflammation, and Probiotics on Neural Development in Autism Spectrum Disorder. Neuroscience 2018; 374:271-286. [DOI: 10.1016/j.neuroscience.2018.01.060] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 01/26/2018] [Accepted: 01/30/2018] [Indexed: 01/01/2023]
|
110
|
High expression of Endogenous Retroviruses from intrauterine life to adulthood in two mouse models of Autism Spectrum Disorders. Sci Rep 2018; 8:629. [PMID: 29330412 PMCID: PMC5766538 DOI: 10.1038/s41598-017-19035-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 12/12/2017] [Indexed: 12/19/2022] Open
Abstract
Retroelements, such as Human Endogenous Retroviruses (HERVs), have been implicated in many complex diseases, including neurological and neuropsychiatric disorders. Previously, we demonstrated a distinctive expression profile of specific HERV families in peripheral blood mononuclear cells from Autistic Spectrum Disorders (ASD) patients, suggesting their involvement in ASD. Here we used two distinct ASD mouse models: inbred BTBR T+tf/J mice and CD-1 outbred mice prenatally exposed to valproic acid. Whole embryos, blood and brain samples from the offspring were collected at different ages and the expression of several ERV families (ETnI, ETnII-α, ETnII-β, ETnII-γ, MusD and IAP), proinflammatory cytokines (IL-1β, IL-6 and TNF-α) and Toll-like receptors (TLR3 and TLR4) was assessed. In the two distinct mouse models analysed, the transcriptional activity of the ERV families was significant higher in comparison with corresponding controls, in whole embryos, blood and brain samples. Also the expression levels of the proinflammatory cytokines and TLRs were significantly higher than controls. Current results are in agreement with our previous findings in ASD children, supporting the hypothesis that ERVs may serve as biomarkers of atypical brain development. Moreover, the changes in ERVs and proinflammatory cytokines expression could be related with the autistic-like traits acquisition in the two mouse models.
Collapse
|
111
|
Shaw CA. Aluminum as a CNS and Immune System Toxin Across the Life Span. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1091:53-83. [DOI: 10.1007/978-981-13-1370-7_4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
|
112
|
Solek CM, Farooqi N, Verly M, Lim TK, Ruthazer ES. Maternal immune activation in neurodevelopmental disorders. Dev Dyn 2017; 247:588-619. [PMID: 29226543 DOI: 10.1002/dvdy.24612] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 11/30/2017] [Accepted: 12/01/2017] [Indexed: 12/12/2022] Open
Abstract
Converging lines of evidence from basic science and clinical studies suggest a relationship between maternal immune activation (MIA) and neurodevelopmental disorders such as autism spectrum disorder (ASD) and schizophrenia. The mechanisms through which MIA increases the risk of neurodevelopmental disorders have become a subject of intensive research. This review aims to describe how dysregulation of microglial function and immune mechanisms may link MIA and neurodevelopmental pathologies. We also summarize the current evidence in animal models of MIA. Developmental Dynamics 247:588-619, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Cynthia M Solek
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Nasr Farooqi
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Myriam Verly
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Tony K Lim
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Edward S Ruthazer
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
113
|
Ryu J, Ha EH, Kim BN, Ha M, Kim Y, Park H, Hong YC, Kim KN. Associations of prenatal and early childhood mercury exposure with autistic behaviors at 5years of age: The Mothers and Children's Environmental Health (MOCEH) study. THE SCIENCE OF THE TOTAL ENVIRONMENT 2017; 605-606:251-257. [PMID: 28667852 DOI: 10.1016/j.scitotenv.2017.06.227] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 06/24/2017] [Accepted: 06/26/2017] [Indexed: 06/07/2023]
Abstract
BACKGROUND Although mercury is an established neurotoxin, only few longitudinal studies have investigated the association between prenatal and early childhood mercury exposure and autistic behaviors. METHODS We conducted a longitudinal cohort study using an ongoing prospective birth cohort initiated in 2006, wherein blood mercury levels were measured at early and late pregnancy; in cord blood; and at 2 and 3years of age. We analyzed 458 mother-child pairs. Autistic behaviors were assessed using the Social Responsiveness Scale (SRS) at 5years of age. Both continuous SRS T-scores and T-scores dichotomized by a score of ≥60 or <60 were used as outcomes. RESULTS The geometric mean of mercury concentrations in cord blood was 5.52μg/L. In adjusted models, a doubling of blood mercury levels at late pregnancy (β=1.84, 95% confidence interval [CI]: 0.39, 3.29), in cord blood (β=2.24, 95% CI: 0.22, 4.27), and at 2years (β=2.12, 95% CI: 0.54, 3.70) and 3years (β=2.80, 95% CI: 0.89, 4.72) of age was positively associated with the SRS T-scores. When the SRS T-scores were dichotomized, we observed positive associations with mercury levels at late pregnancy (relative risk [RR]=1.31, 95% CI: 1.08, 1.60) and in cord blood (RR=1.28, 95% CI: 1.01, 1.63). CONCLUSION We found that blood mercury levels at late pregnancy and early childhood were associated with more autistic behaviors in children at 5years of age. Further study on the long-term effects of mercury exposure is recommended.
Collapse
Affiliation(s)
- Jia Ryu
- Department of Occupational and Environmental Medicine, School of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Eun-Hee Ha
- Department of Occupational and Environmental Medicine, School of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Boong-Nyun Kim
- Division of Child & Adolescent Psychiatry, Department of Psychiatry and Institute of Human Behavioral Medicine, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Mina Ha
- Department of Preventive Medicine, College of Medicine, Dankook University, Cheonan, Republic of Korea
| | - Yangho Kim
- Department of Occupational and Environmental Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea
| | - Hyesook Park
- Department of Preventive Medicine, School of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Yun-Chul Hong
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kyoung-Nam Kim
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea; Institute of Public Health and Medical Service, Seoul National University Hospital, Seoul, Republic of Korea.
| |
Collapse
|
114
|
Kalkman HO, Feuerbach D. Microglia M2A Polarization as Potential Link between Food Allergy and Autism Spectrum Disorders. Pharmaceuticals (Basel) 2017; 10:ph10040095. [PMID: 29232822 PMCID: PMC5748650 DOI: 10.3390/ph10040095] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 12/05/2017] [Accepted: 12/05/2017] [Indexed: 12/18/2022] Open
Abstract
Atopic diseases are frequently co-morbid with autism spectrum disorders (ASD). Allergic responses are associated with an activation of mast cells, innate lymphoid cells, and Th2 cells. These cells produce type-2 cytokines (IL4 and IL13), which stimulate microglia and macrophages to adopt a phenotype referred to as ‘alternative activation’ or ‘M2A’. M2A-polarized macrophages and microglia play a physiological role in tissue repair by secreting growth factors such as brain-derived neurotrophic factor (BDNF) and insulin-like growth factor-1. In ASD there is evidence for increased type-2 cytokines, microglia activation, M2A polarization, and increased levels of growth factors. In neurons, these growth factors drive a signal transduction pathway that leads to activation of the enzyme mammalian Target of Rapamycin (mTOR), and thereby to the inhibition of autophagy. Activation of mTOR is an effect that is also common to several of the genetic forms of autism. In the central nervous system, redundant synapses are removed via an autophagic process. Activation of mTOR would diminish the pruning of redundant synapses, which in the context of ASD is likely to be undesired. Based on this line of reasoning, atopic diseases like food allergy, eczema or asthma would represent risk factors for autism spectrum disorders.
Collapse
|
115
|
Li K, Li L, Cui B, Gai Z, Li Q, Wang S, Yan J, Lin B, Tian L, Liu H, Liu X, Xi Z. Early Postnatal Exposure to Airborne Fine Particulate Matter Induces Autism-like Phenotypes in Male Rats. Toxicol Sci 2017; 162:189-199. [DOI: 10.1093/toxsci/kfx240] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Affiliation(s)
- Kang Li
- Department of Toxicology
- Department of Stress Medicine
| | | | - Bo Cui
- Department of Occupational Hygiene, Institute of Health and Environmental Medicine, Tianjin, China
| | - Zhihui Gai
- Department of Occupational Hygiene, Institute of Health and Environmental Medicine, Tianjin, China
| | | | | | | | | | | | | | | | | |
Collapse
|
116
|
Werneburg S, Feinberg PA, Johnson KM, Schafer DP. A microglia-cytokine axis to modulate synaptic connectivity and function. Curr Opin Neurobiol 2017; 47:138-145. [PMID: 29096242 DOI: 10.1016/j.conb.2017.10.002] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 09/25/2017] [Accepted: 10/07/2017] [Indexed: 02/05/2023]
Abstract
Microglia have recently been recognized as key regulators of synapse development, function, and plasticity. Critical to progressing the field is the identification of molecular underpinnings necessary for microglia to carry out these important functions within neural circuits. Here, we focus a review specifically on roles for microglial cytokine signaling within developing and mature neural circuits. We review exciting new studies demonstrating essential roles for microglial cytokine signaling in axon outgrowth, synaptogenesis and synapse maturation during development, as well as synaptic transmission and plasticity in adulthood. Together, these studies identify microglia and cytokines as critical modulators of neural circuits within the healthy brain, with implications for a broad range of neurological disorders with disruptions in synaptic structure and function.
Collapse
Affiliation(s)
- Sebastian Werneburg
- Department of Neurobiology, University of Massachusetts Medical School, 364 Plantation Street, Worcester MA 01605, USA
| | - Philip A Feinberg
- Department of Neurobiology, University of Massachusetts Medical School, 364 Plantation Street, Worcester MA 01605, USA
| | - Kasey M Johnson
- Department of Neurobiology, University of Massachusetts Medical School, 364 Plantation Street, Worcester MA 01605, USA
| | - Dorothy P Schafer
- Department of Neurobiology, University of Massachusetts Medical School, 364 Plantation Street, Worcester MA 01605, USA.
| |
Collapse
|
117
|
Microglia emerge as central players in brain disease. Nat Med 2017; 23:1018-1027. [PMID: 28886007 DOI: 10.1038/nm.4397] [Citation(s) in RCA: 1159] [Impact Index Per Article: 144.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 07/26/2017] [Indexed: 02/07/2023]
Abstract
There has been an explosion of new findings recently giving us insights into the involvement of microglia in central nervous system (CNS) disorders. A host of new molecular tools and mouse models of disease are increasingly implicating this enigmatic type of nervous system cell as a key player in conditions ranging from neurodevelopmental disorders such as autism to neurodegenerative disorders such as Alzheimer's disease and chronic pain. Contemporaneously, diverse roles are emerging for microglia in the healthy brain, from sculpting developing neuronal circuits to guiding learning-associated plasticity. Understanding the physiological functions of these cells is crucial to determining their roles in disease. Here we focus on recent developments in our rapidly expanding understanding of the function, as well as the dysfunction, of microglia in disorders of the CNS.
Collapse
|
118
|
Abstract
Early autism research focused on behavior and cognition. In recent decades, the pace of research has accelerated, and advances in imaging and genetics have allowed the accumulation of biological data. Nevertheless, a coherent picture of the syndrome at either phenotypic or biological level has not emerged. We see two fundamental obstacles to progress in basic understanding of autism. First, the two defining features (impairment in social interactions and communication, and restricted, repetitive behaviors and interests) are historically seen as integrally related. Others hold that these two major traits are fractionable and must be studied independently, casting doubt on autism as a coherent syndrome. Second, despite much recent research on brain structure and function, environmental factors, and genetics/genomics, findings on the biological level have not generally aligned well with those on the phenotypic level. In the first two sections, we explore these challenges, and in the third section, we review approaches that may facilitate progress, such as (1) including in studies all individuals defined by social impairment without regard to repetitive behaviors, (2) forming narrowly defined subtypes by thorough characterization on specific features, both diagnostic and non-diagnostic, (3) focusing on characteristics that may be relatively robust to environmental influence, (4) studying children as early as possible, minimizing environmental influence, and including longitudinal course as an important part of the phenotype, (5) subtyping by environmental risk factors, (6) distinguishing between what participants can do and what they typically do, and (7) aggregating large data sets across sites. (JINS, 2017, 23, 903-915).
Collapse
|
119
|
Tural Hesapcioglu S, Kasak M, Cıtak Kurt AN, Ceylan MF. High monocyte level and low lymphocyte to monocyte ratio in autism spectrum disorders. INTERNATIONAL JOURNAL OF DEVELOPMENTAL DISABILITIES 2017; 65:73-81. [PMID: 34141326 PMCID: PMC8115457 DOI: 10.1080/20473869.2017.1371369] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Objective: This study aims to investigate the level of peripheral blood mononuclear cells and their ratios which may point to the immunological mechanisms involved in the etiopathogenesis of ASD. Method: The complete blood count parameters of the 45 ASD cases were compared with those of healthy controls.Childhood Autism Rating Scale (CARS) was performed to measure the disease severity. Results: The monocytes of ASD group were significantly higher; and the lymphocyte-to-monocyte ratio (LMR) was lower than the controls'. LMR and neutrophil-to-lymphocyte ratio were found to be predictors of ASD. The decrease in LMR (B: -0.744; P=0.035; CI: -1.431 to -0.056) and the increase in age (B: 0.432; P=0.045; CI: 0.011-0.853) were related to high CARS scores in linear regression analyses. Conclusions: The results of this study support the role of altered immune cell counts and ratios in ASD. A high monocyte level and low LMR may have diagnostic values in autism.
Collapse
Affiliation(s)
- Selma Tural Hesapcioglu
- Faculty of Medicine, Child and Adolescent Psychiatry Department, Ankara Yildirim Beyazit University, Ankara, Turkey
| | - Meryem Kasak
- Faculty of Medicine, Child and Adolescent Psychiatry Department, Ankara Yildirim Beyazit University, Ankara, Turkey
| | - Aysegül Nese Cıtak Kurt
- Faculty of Medicine, Child Neurology Department, Ankara Yildirim Beyazit University, Ankara, Turkey
| | - Mehmet Fatih Ceylan
- Faculty of Medicine, Child and Adolescent Psychiatry Department, Ankara Yildirim Beyazit University, Ankara, Turkey
| |
Collapse
|
120
|
Li D, Tomljenovic L, Li Y, Shaw CA. RETRACTED: Subcutaneous injections of aluminum at vaccine adjuvant levels activate innate immune genes in mouse brain that are homologous with biomarkers of autism. J Inorg Biochem 2017; 177:39-54. [PMID: 28923356 DOI: 10.1016/j.jinorgbio.2017.08.035] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 08/30/2017] [Accepted: 08/31/2017] [Indexed: 01/11/2023]
Affiliation(s)
- Dan Li
- Dept. of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Lucija Tomljenovic
- Dept. of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Yongling Li
- Dept. of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Christopher A Shaw
- Dept. of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada; Program in Experimental Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
121
|
Bridging Autism Spectrum Disorders and Schizophrenia through inflammation and biomarkers - pre-clinical and clinical investigations. J Neuroinflammation 2017; 14:179. [PMID: 28870209 PMCID: PMC5584030 DOI: 10.1186/s12974-017-0938-y] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 08/08/2017] [Indexed: 12/15/2022] Open
Abstract
In recent years, evidence supporting a link between inflammation and neuropsychiatric disorders has been mounting. Autism spectrum disorders (ASD) and schizophrenia share some clinical similarities which we hypothesize might reflect the same biological basis, namely, in terms of inflammation. However, the diagnosis of ASD and schizophrenia relies solely on clinical symptoms, and to date, there is no clinically useful biomarker to diagnose or monitor the course of such illnesses. The focus of this review is the central role that inflammation plays in ASD and schizophrenia. It spans from pre-clinical animal models to clinical research and excludes in vitro studies. Four major areas are covered: (1) microglia, the inflammatory brain resident myeloid cells, (2) biomarkers, including circulating cytokines, oxidative stress markers, and microRNA players, known to influence cellular processes at brain and immune levels, (3) effect of anti-psychotics on biomarkers and other predictors of response, and (4) impact of gender on response to immune activation, biomarkers, and response to anti-psychotic treatments.
Collapse
|
122
|
Xu X, Wu D, Hou S, Zhu J, Li J, Tang J. Prenatal exposure to TAK242 affects the childhood autism in offspring in animal models of autism spectrum disorder. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2017; 20:1016-1020. [PMID: 29085596 PMCID: PMC5651454 DOI: 10.22038/ijbms.2017.9270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
OBJECTIVES To evaluate whether prenatal exposure to TAK242 affects childhood autism in the offspring in animal models of autism spectrum disorder (ASD). MATERIALS AND METHODS The pregnant rats were pseudo-randomly divided into three groups, the ASD model group, the TAK242 treatment group, and the control group. The ASD model was constructed by injecting IP with LPS. The blood samples from 1-month-old offspring were collected for cytokine evaluation and the social interaction test was used in the offspring of ASD rats. Rats were killed and the hippocampus, cerebral cortex, and cerebellum were used for the immunohistochemical study. RESULTS As compared to the control, the levels of IFN-γ, IL-1β, IL-2, and IL-6 were significantly increased (P<0.05), and the levels of IL-4, IL-10, and TGF-β were significantly decreased (P <0.05) in the offspring of ASD rats; whereas those cytokines were significantly reversed after prenatal exposure to TAK242 (P<0.05). The hesitation time and none-social interaction time were significantly increased as compared to the control (P<0.05); whereas they were both decreased after prenatal exposure to TAK242 (P<0.05). This was contrary to the social interaction time (P<0.05). The expression of GFAP and IBA1 in the cortex, hippocampus, and cerebellum were stronger in the LPS group as compared to control group, and this effect was reversed after prenatal exposure to TAK242. CONCLUSION Prenatal exposure to TAK242 affects serum cytokines levels and the social interaction time in rat offspring in animal models of ASD.
Collapse
Affiliation(s)
- Xiaoyan Xu
- The Children's Neurorehabilitation Center, the First Affiliated Hospital, Anhui Medical University, Hefei 230022, Anhui, China
| | - De Wu
- The Children's Neurorehabilitation Center, the First Affiliated Hospital, Anhui Medical University, Hefei 230022, Anhui, China
| | - Shu Hou
- Department of Pediatrics, the First Affiliated Hospital, Anhui Medical University, Hefei 230022, Anhui, China
| | - Jing Zhu
- The Children's Neurorehabilitation Center, the First Affiliated Hospital, Anhui Medical University, Hefei 230022, Anhui, China
| | - Jing Li
- The Children's Neurorehabilitation Center, the First Affiliated Hospital, Anhui Medical University, Hefei 230022, Anhui, China
| | - Jiulai Tang
- The Children's Neurorehabilitation Center, the First Affiliated Hospital, Anhui Medical University, Hefei 230022, Anhui, China
| |
Collapse
|
123
|
Bilbo SD, Block CL, Bolton JL, Hanamsagar R, Tran PK. Beyond infection - Maternal immune activation by environmental factors, microglial development, and relevance for autism spectrum disorders. Exp Neurol 2017; 299:241-251. [PMID: 28698032 DOI: 10.1016/j.expneurol.2017.07.002] [Citation(s) in RCA: 207] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 06/26/2017] [Accepted: 07/05/2017] [Indexed: 12/17/2022]
Abstract
Immune molecules such as cytokines and chemokines and the cells that produce them within the brain, notably microglia, are critical for normal brain development. This recognition has in recent years led to the working hypothesis that inflammatory events during pregnancy, e.g. in response to infection, may disrupt the normal expression of immune molecules during critical stages of neural development and thereby contribute to the risk for neurodevelopmental disorders such as autism spectrum disorder (ASD). This hypothesis has in large part been shepherded by the work of Dr. Paul Patterson and colleagues, which has elegantly demonstrated that a single viral infection or injection of a viral mimetic to pregnant mice significantly and persistently impacts offspring immune and nervous system function, changes that underlie ASD-like behavioral dysfunction including social and communication deficits. Subsequent studies by many labs - in humans and in non-human animal models - have supported the hypothesis that ongoing disrupted immune molecule expression and/or neuroinflammation contributes to at least a significant subset of ASD. The heterogeneous clinical and biological phenotypes observed in ASD strongly suggest that in genetically susceptible individuals, environmental risk factors combine or synergize to create a tipping or threshold point for dysfunction. Importantly, animal studies showing a link between maternal immune activation (MIA) and ASD-like outcomes in offspring involve different species and diverse environmental factors associated with ASD in humans, beyond infection, including toxin exposures, maternal stress, and maternal obesity, all of which impact inflammatory or immune pathways. The goal of this review is to highlight the broader implications of Dr. Patterson's work for the field of autism, with a focus on the impact that MIA by diverse environmental factors has on fetal brain development, immune system development, and the pathophysiology of ASD.
Collapse
Affiliation(s)
- Staci D Bilbo
- Pediatrics and Neuroscience, Harvard Medical School, Lurie Center for Autism, Massachusetts General Hospital for Children, Boston, MA 02126, United States.
| | - Carina L Block
- Psychology and Neuroscience, Duke University, Durham, NC 27708, United States
| | - Jessica L Bolton
- Pediatrics and Anatomy/Neurobiology, University of California-Irvine, Irvine, CA 92697, United States
| | - Richa Hanamsagar
- Pediatrics and Neuroscience, Harvard Medical School, Lurie Center for Autism, Massachusetts General Hospital for Children, Boston, MA 02126, United States
| | - Phuong K Tran
- Pediatrics and Neuroscience, Harvard Medical School, Lurie Center for Autism, Massachusetts General Hospital for Children, Boston, MA 02126, United States
| |
Collapse
|
124
|
Yenkoyan K, Grigoryan A, Fereshetyan K, Yepremyan D. Advances in understanding the pathophysiology of autism spectrum disorders. Behav Brain Res 2017; 331:92-101. [DOI: 10.1016/j.bbr.2017.04.038] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 04/16/2017] [Accepted: 04/18/2017] [Indexed: 10/19/2022]
|
125
|
Comprehensive investigation of temporal and autism-associated cell type composition-dependent and independent gene expression changes in human brains. Sci Rep 2017. [PMID: 28646201 PMCID: PMC5482876 DOI: 10.1038/s41598-017-04356-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The functions of human brains highly depend on the precise temporal regulation of gene expression, and the temporal brain transcriptome profile across lifespan has been observed. The substantial transcriptome alteration in neural disorders like autism has also been observed and is thought to be important for the pathology. While the cell type composition is known to be variable in brains, it remains unclear how it contributes to the temporal and pathological transcriptome changes in brains. Here, we applied a transcriptome deconvolution procedure to an age series RNA-seq dataset of healthy and autism samples, to quantify the contribution of cell type composition in shaping the temporal and autism pathological transcriptome in human brains. We estimated that composition change was the primary factor of both types of transcriptome changes. On the other hand, genes with substantial composition-independent expression changes were also observed in both cases. Those temporal and autism pathological composition-independent changes, many of which are related to synaptic functions, indicate the important intracellular regulatory changes in human brains in both processes.
Collapse
|
126
|
Qasem H, Al-Ayadhi L, Al Dera H, El-Ansary A. Increase of cytosolic phospholipase A2 as hydrolytic enzyme of phospholipids and autism cognitive, social and sensory dysfunction severity. Lipids Health Dis 2017; 16:117. [PMID: 28724385 PMCID: PMC5516334 DOI: 10.1186/s12944-016-0391-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 12/13/2016] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Autism is neurodevelopmental disorder that is characterized by developmental, behavioral, social and sensory abnormalities. Researchers have focused in last years in immunological alteration and inflammation as a hot subject in autism field. This work aims to study the alteration in phospholipids (PE, PS, and PC) together with the change in cPLA2 concentration as the main phospholipid hydrolytic enzyme in autistic patients compared to control. It was also extended to find a correlation between these biomarkers and severity of autism measured as childhood autism rating scale (CARS), Social responsiveness scale (SRS), and Short sensory profile (SSP). METHODS Phospholipids (PE, PS, PC) and cPLA2 as biochemical parameters were determined in the plasma of 48 Saudi autistic male patients, categorized as mild-moderate and severe as indicated by their Childhood Autism Rating Scale (CARS), social responsiveness scale (SRS) and short sensory profile (SSP) and compared to 40 age- and gender-matched control samples. RESULTS The reported data demonstrate significantly lower levels of PE, PS, and PC together with a significant increase in cPLA2. While association between severity of autism and impaired phospholipid concentration was completely lacked, an association between cPLA2 and impaired sensory processing was observed. CONCLUSIONS The impaired phospholipid level and remarkable increased in cPLA2 concentration asserted their roles in the etiology of autism. Receiver operating characteristic analysis together with predictiveness diagrams proved that the measured parameters could be used as predictive biomarkers of clinical symptoms and provide significant guidance for future therapeutic strategy to re-establish physiological homeostasis.
Collapse
Affiliation(s)
- Hanan Qasem
- Biochemistry Department, Science College, King Saud University, P.O. Box 22452, 11495 Riyadh, Saudi Arabia
| | - Laila Al-Ayadhi
- Autism Research and Treatment Center, Riyadh, Saudi Arabia
- Shaik AL-Amodi Autism Research Chair, King Saud University, Riyadh, Saudi Arabia
- Department of Physiology, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Hussain Al Dera
- Basic medical science dept. College of Medicine, King Saud bin Abdul Aziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center (Kaimrc), Riyadh, Saudi Arabia
| | - Afaf El-Ansary
- Biochemistry Department, Science College, King Saud University, P.O. Box 22452, 11495 Riyadh, Saudi Arabia
- Autism Research and Treatment Center, Riyadh, Saudi Arabia
- Shaik AL-Amodi Autism Research Chair, King Saud University, Riyadh, Saudi Arabia
- Central Laboratory, Center for Female Scientific and Medical Colleges at King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
127
|
Rodriguez Cetina Biefer H, Vasudevan A, Elkhal A. Aspects of Tryptophan and Nicotinamide Adenine Dinucleotide in Immunity: A New Twist in an Old Tale. Int J Tryptophan Res 2017; 10:1178646917713491. [PMID: 28659716 PMCID: PMC5476425 DOI: 10.1177/1178646917713491] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 04/07/2017] [Indexed: 12/26/2022] Open
Abstract
Increasing evidence underscores the interesting ability of tryptophan to regulate immune responses. However, the exact mechanisms of tryptophan's immune regulation remain to be determined. Tryptophan catabolism via the kynurenine pathway is known to play an important role in tryptophan's involvement in immune responses. Interestingly, quinolinic acid, which is a neurotoxic catabolite of the kynurenine pathway, is the major pathway for the de novo synthesis of nicotinamide adenine dinucleotide (NAD+). Recent studies have shown that NAD+, a natural coenzyme found in all living cells, regulates immune responses and creates homeostasis via a novel signaling pathway. More importantly, the immunoregulatory properties of NAD+ are strongly related to the overexpression of tryptophan hydroxylase 1 (Tph1). This review provides recent knowledge of tryptophan and NAD+ and their specific and intriguing roles in the immune system. Furthermore, it focuses on the mechanisms by which tryptophan regulates NAD+ synthesis as well as innate and adaptive immune responses.
Collapse
Affiliation(s)
| | - Anju Vasudevan
- Angiogenesis and Brain Development Laboratory, Division of Basic Neuroscience, McLean Hospital and Harvard Medical School, Belmont, MA, USA
| | - Abdallah Elkhal
- Division of Transplant Surgery and Transplantation Surgery Research Laboratory, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
128
|
Maternal Immune Activation Causes Behavioral Impairments and Altered Cerebellar Cytokine and Synaptic Protein Expression. Neuropsychopharmacology 2017; 42:1435-1446. [PMID: 28102228 PMCID: PMC5436129 DOI: 10.1038/npp.2017.7] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 12/21/2016] [Accepted: 01/02/2017] [Indexed: 01/10/2023]
Abstract
Emerging epidemiology studies indicate that maternal immune activation (MIA) resulting from inflammatory stimuli such as viral or bacterial infections during pregnancy serves as a risk factor for multiple neurodevelopmental disorders including autism spectrum disorders and schizophrenia. Although alterations in the cortex and hippocampus of MIA offspring have been described, less evidence exists on the impact on the cerebellum. Here, we report altered expression of cytokines and chemokines in the cerebellum of MIA offspring, including increase in the neuroinflammatory cytokine TNFα and its receptor TNFR1. We also report reduced expression of the synaptic organizing proteins cerebellin-1 and GluRδ2. These synaptic protein alterations are associated with a deficit in the ability of cerebellar neurons to form synapses and an increased number of dendritic spines that are not in contact with a presynaptic terminal. These impairments are likely contributing to the behavioral deficits in the MIA exposed offspring.
Collapse
|
129
|
Bolton JL, Marinero S, Hassanzadeh T, Natesan D, Le D, Belliveau C, Mason SN, Auten RL, Bilbo SD. Gestational Exposure to Air Pollution Alters Cortical Volume, Microglial Morphology, and Microglia-Neuron Interactions in a Sex-Specific Manner. Front Synaptic Neurosci 2017; 9:10. [PMID: 28620294 PMCID: PMC5449437 DOI: 10.3389/fnsyn.2017.00010] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 05/09/2017] [Indexed: 12/22/2022] Open
Abstract
Microglia are the resident immune cells of the brain, important for normal neural development in addition to host defense in response to inflammatory stimuli. Air pollution is one of the most pervasive and harmful environmental toxicants in the modern world, and several large scale epidemiological studies have recently linked prenatal air pollution exposure with an increased risk of neurodevelopmental disorders such as autism spectrum disorder (ASD). Diesel exhaust particles (DEP) are a primary toxic component of air pollution, and markedly activate microglia in vitro and in vivo in adult rodents. We have demonstrated that prenatal exposure to DEP in mice, i.e., to the pregnant dams throughout gestation, results in a persistent vulnerability to behavioral deficits in adult offspring, especially in males, which is intriguing given the greater incidence of ASD in males to females (∼4:1). Moreover, there is a striking upregulation of toll-like receptor (TLR) 4 gene expression within the brains of the same mice, and this expression is primarily in microglia. Here we explored the impact of gestational exposure to DEP or vehicle on microglial morphology in the developing brains of male and female mice. DEP exposure increased inflammatory cytokine protein and altered the morphology of microglia, consistent with activation or a delay in maturation, only within the embryonic brains of male mice; and these effects were dependent on TLR4. DEP exposure also increased cortical volume at embryonic day (E)18, which switched to decreased volume by post-natal day (P)30 in males, suggesting an impact on the developing neural stem cell niche. Consistent with this hypothesis, we found increased microglial-neuronal interactions in male offspring that received DEP compared to all other groups. Taken together, these data suggest a mechanism by which prenatal exposure to environmental toxins may affect microglial development and long-term function, and thereby contribute to the risk of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Jessica L Bolton
- Department of Psychology and Neuroscience, Duke University, DurhamNC, United States
| | - Steven Marinero
- Department of Neurobiology, Duke University Medical Center, DurhamNC, United States
| | - Tania Hassanzadeh
- Department of Psychology and Neuroscience, Duke University, DurhamNC, United States
| | - Divya Natesan
- Department of Psychology and Neuroscience, Duke University, DurhamNC, United States
| | - Dominic Le
- Department of Psychology and Neuroscience, Duke University, DurhamNC, United States
| | - Christine Belliveau
- Department of Psychology and Neuroscience, Duke University, DurhamNC, United States
| | - S N Mason
- Department of Pediatrics, Division of Neonatal Medicine, Duke University Medical Center, DurhamNC, United States
| | - Richard L Auten
- Department of Pediatrics, Division of Neonatal Medicine, Duke University Medical Center, DurhamNC, United States
| | - Staci D Bilbo
- Department of Psychology and Neuroscience, Duke University, DurhamNC, United States.,Department of Neurobiology, Duke University Medical Center, DurhamNC, United States.,Department of Pediatrics and Program in Neuroscience, Lurie Center for Autism, Harvard Medical School, Massachusetts General Hospital for Children, BostonMA, United States
| |
Collapse
|
130
|
Careaga M, Rogers S, Hansen RL, Amaral DG, Van de Water J, Ashwood P. Immune Endophenotypes in Children With Autism Spectrum Disorder. Biol Psychiatry 2017; 81:434-441. [PMID: 26493496 PMCID: PMC4788581 DOI: 10.1016/j.biopsych.2015.08.036] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 08/11/2015] [Accepted: 08/25/2015] [Indexed: 11/17/2022]
Abstract
BACKGROUND Autism spectrum disorder (ASD) is characterized by social communication deficits and restricted, repetitive patterns of behavior. Varied immunological findings have been reported in children with ASD. To address the question of heterogeneity in immune responses, we sought to examine the diversity of immune profiles within a representative cohort of boys with ASD. METHODS Peripheral blood mononuclear cells from male children with ASD (n = 50) and from typically developing age-matched male control subjects (n = 16) were stimulated with either lipopolysaccharide or phytohemagglutinin. Cytokine production was assessed after stimulation. The ASD study population was clustered into subgroups based on immune responses and assessed for behavioral outcomes. RESULTS Children with ASD who had a proinflammatory profile based on lipopolysaccharide stimulation were more developmentally impaired as assessed by the Mullen Scales of Early Learning. They also had greater impairments in social affect as measured by the Autism Diagnostic Observation Schedule. These children also displayed more frequent sleep disturbances and episodes of aggression. Similarly, children with ASD and a more activated T cell cytokine profile after phytohemagglutinin stimulation were more developmentally impaired as measured by the Mullen Scales of Early Learning. CONCLUSIONS Children with ASD may be phenotypically characterized based upon their immune profile. Those showing either an innate proinflammatory response or increased T cell activation/skewing display a more impaired behavioral profile than children with noninflamed or non-T cell activated immune profiles. These data suggest that there may be several possible immune subphenotypes within the ASD population that correlate with more severe behavioral impairments.
Collapse
Affiliation(s)
- Milo Careaga
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, California; MIND Institute, University of California, Davis, Davis, California
| | - Sally Rogers
- MIND Institute, University of California, Davis, Davis, California; Departments of Psychiatry, University of California, Davis, Davis, California
| | - Robin L Hansen
- MIND Institute, University of California, Davis, Davis, California; Pediatrics, University of California, Davis, Davis, California
| | - David G Amaral
- MIND Institute, University of California, Davis, Davis, California; Departments of Psychiatry, University of California, Davis, Davis, California
| | - Judy Van de Water
- MIND Institute, University of California, Davis, Davis, California; Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, Davis, California
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, California; MIND Institute, University of California, Davis, Davis, California.
| |
Collapse
|
131
|
AL-Ayadhi L, Halepoto DM. Camel Milk as a Potential Nutritional Therapy in Autism. NUTRIENTS IN DAIRY AND THEIR IMPLICATIONS ON HEALTH AND DISEASE 2017:389-405. [DOI: 10.1016/b978-0-12-809762-5.00030-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
132
|
Yeh E, Weiss LA. If genetic variation could talk: What genomic data may teach us about the importance of gene expression regulation in the genetics of autism. Mol Cell Probes 2016; 30:346-356. [PMID: 27751841 DOI: 10.1016/j.mcp.2016.10.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 10/09/2016] [Accepted: 10/13/2016] [Indexed: 11/25/2022]
Abstract
Autism spectrum disorder (ASD) has been long known to have substantial genetic etiology. Much research has attempted to identify specific genes contributing to ASD risk with the goal of tying gene function to a molecular pathological explanation for ASD. A unifying molecular pathology would potentially increase understanding of what is going wrong during development, and could lead to diagnostic biomarkers or targeted preventative or therapeutic directions. We review past and current genetic mapping approaches and discuss major results, leading to the hypothesis that global dysregulation of gene or protein expression may be implicated in ASD rather than disturbance of brain-specific functions. If substantiated, this hypothesis might indicate the need for novel experimental and analytical approaches in order to understand this neurodevelopmental disorder, develop biomarkers, or consider treatment approaches.
Collapse
Affiliation(s)
- Erika Yeh
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Lauren A Weiss
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA, 94143, USA; Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, 94143, USA.
| |
Collapse
|
133
|
Fiorentino M, Sapone A, Senger S, Camhi SS, Kadzielski SM, Buie TM, Kelly DL, Cascella N, Fasano A. Blood-brain barrier and intestinal epithelial barrier alterations in autism spectrum disorders. Mol Autism 2016; 7:49. [PMID: 27957319 PMCID: PMC5129651 DOI: 10.1186/s13229-016-0110-z] [Citation(s) in RCA: 305] [Impact Index Per Article: 33.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 11/12/2016] [Indexed: 12/11/2022] Open
Abstract
Background Autism spectrum disorders (ASD) are complex conditions whose pathogenesis may be attributed to gene–environment interactions. There are no definitive mechanisms explaining how environmental triggers can lead to ASD although the involvement of inflammation and immunity has been suggested. Inappropriate antigen trafficking through an impaired intestinal barrier, followed by passage of these antigens or immune-activated complexes through a permissive blood–brain barrier (BBB), can be part of the chain of events leading to these disorders. Our goal was to investigate whether an altered BBB and gut permeability is part of the pathophysiology of ASD. Methods Postmortem cerebral cortex and cerebellum tissues from ASD, schizophrenia (SCZ), and healthy subjects (HC) and duodenal biopsies from ASD and HC were analyzed for gene and protein expression profiles. Tight junctions and other key molecules associated with the neurovascular unit integrity and function and neuroinflammation were investigated. Results Claudin (CLDN)-5 and -12 were increased in the ASD cortex and cerebellum. CLDN-3, tricellulin, and MMP-9 were higher in the ASD cortex. IL-8, tPA, and IBA-1 were downregulated in SCZ cortex; IL-1b was increased in the SCZ cerebellum. Differences between SCZ and ASD were observed for most of the genes analyzed in both brain areas. CLDN-5 protein was increased in ASD cortex and cerebellum, while CLDN-12 appeared reduced in both ASD and SCZ cortexes. In the intestine, 75% of the ASD samples analyzed had reduced expression of barrier-forming TJ components (CLDN-1, OCLN, TRIC), whereas 66% had increased pore-forming CLDNs (CLDN-2, -10, -15) compared to controls. Conclusions In the ASD brain, there is an altered expression of genes associated with BBB integrity coupled with increased neuroinflammation and possibly impaired gut barrier integrity. While these findings seem to be specific for ASD, the possibility of more distinct SCZ subgroups should be explored with additional studies.
Collapse
Affiliation(s)
- Maria Fiorentino
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Boston, MA USA ; Department of Pediatrics, Harvard Medical School, Boston, MA USA
| | - Anna Sapone
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Boston, MA USA ; Department of Medicine, Celiac Center, Division of Gastroenterology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA USA
| | - Stefania Senger
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Boston, MA USA ; Department of Pediatrics, Harvard Medical School, Boston, MA USA
| | - Stephanie S Camhi
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Boston, MA USA ; Center for Celiac Research and Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital for Children, Boston, MA USA
| | | | - Timothy M Buie
- Department of Pediatrics, Harvard Medical School, Boston, MA USA
| | - Deanna L Kelly
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD USA
| | - Nicola Cascella
- Neuropsychiatry Program, Sheppard Pratt Health System, Baltimore, MD USA
| | - Alessio Fasano
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Boston, MA USA ; Center for Celiac Research and Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital for Children, Boston, MA USA ; Department of Pediatrics, Harvard Medical School, Boston, MA USA
| |
Collapse
|
134
|
Carter CJ, Blizard RA. Autism genes are selectively targeted by environmental pollutants including pesticides, heavy metals, bisphenol A, phthalates and many others in food, cosmetics or household products. Neurochem Int 2016; 101:S0197-0186(16)30197-8. [PMID: 27984170 DOI: 10.1016/j.neuint.2016.10.011] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 10/18/2016] [Accepted: 10/26/2016] [Indexed: 11/21/2022]
Abstract
The increasing incidence of autism suggests a major environmental influence. Epidemiology has implicated many candidates and genetics many susceptibility genes. Gene/environment interactions in autism were analysed using 206 autism susceptibility genes (ASG's) from the Autworks database to interrogate ∼1 million chemical/gene interactions in the comparative toxicogenomics database. Any bias towards ASG's was statistically determined for each chemical. Many suspect compounds identified in epidemiology, including tetrachlorodibenzodioxin, pesticides, particulate matter, benzo(a)pyrene, heavy metals, valproate, acetaminophen, SSRI's, cocaine, bisphenol A, phthalates, polyhalogenated biphenyls, flame retardants, diesel constituents, terbutaline and oxytocin, inter alia showed a significant degree of bias towards ASG's, as did relevant endogenous agents (retinoids, sex steroids, thyroxine, melatonin, folate, dopamine, serotonin). Numerous other suspected endocrine disruptors (over 100) selectively targeted ASG's including paraquat, atrazine and other pesticides not yet studied in autism and many compounds used in food, cosmetics or household products, including tretinoin, soy phytoestrogens, aspartame, titanium dioxide and sodium fluoride. Autism polymorphisms influence the sensitivity to some of these chemicals and these same genes play an important role in barrier function and control of respiratory cilia sweeping particulate matter from the airways. Pesticides, heavy metals and pollutants also disrupt barrier and/or ciliary function, which is regulated by sex steroids and by bitter/sweet taste receptors. Further epidemiological studies and neurodevelopmental and behavioural research is warranted to determine the relevance of large number of suspect candidates whose addition to the environment, household, food and cosmetics might be fuelling the autism epidemic in a gene-dependent manner.
Collapse
Affiliation(s)
- C J Carter
- PolygenicPathways, Flat 2, 40 Baldslow Road, Hastings, East Sussex, TN34 2EY, UK.
| | - R A Blizard
- Molecular Psychiatry Laboratory, Mental Health Sciences Unit, University College, London, UK
| |
Collapse
|
135
|
Uchino S, Waga C. Novel Therapeutic Approach for Autism Spectrum Disorder: Focus on SHANK3. Curr Neuropharmacol 2016; 13:786-92. [PMID: 26511836 PMCID: PMC4759317 DOI: 10.2174/1570159x13666151029105547] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 03/16/2015] [Accepted: 03/15/2015] [Indexed: 01/12/2023] Open
Abstract
SHANK3 is a synaptic scaffolding protein and plays an important role in neuronal
development. SHANK3 interacts with various synaptic molecules, including post-synaptic density-95
(PSD-95), homer and GluR1 AMPA receptor. SHANK3 gene is a causable gene of the Phelan-
McDermid syndrome (also known as the 22q13.3 deletion syndrome), whose manifestation is global
developmental delay and autistic behavior, especially shows severe speech and language deficit.
Additionally since cumulative gene analysis in autistic subjects identified several mutations in
SHANK3 gene, including deletion and duplication in a particular region, abnormality of SHANK3
gene is thought the be related with the neuropathology of autism spectrum disorder (ASD). We here review the recent
findings in regard to the roles of SHANK3 in higher brain functions, molecular-biologic studies of the complex
expression of Shank3 transcripts and production of SHANK3 isoforms, and behavioral studies of Shank3-mutant mice,
including our recent findings, and discuss a novel therapeutic approach for ASD.
Collapse
Affiliation(s)
- Shigeo Uchino
- Department of Neurobiology, Faculty of Biosciences, School of Science and Engineering, Teikyo University, 1-1 Toyosatodai, Utsunomiya, Tochigi 320-8551, Japan.
| | | |
Collapse
|
136
|
Kern JK, Geier DA, Sykes LK, Haley BE, Geier MR. The relationship between mercury and autism: A comprehensive review and discussion. J Trace Elem Med Biol 2016; 37:8-24. [PMID: 27473827 DOI: 10.1016/j.jtemb.2016.06.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 05/17/2016] [Accepted: 06/01/2016] [Indexed: 12/28/2022]
Abstract
The brain pathology in autism spectrum disorders (ASD) indicates marked and ongoing inflammatory reactivity with concomitant neuronal damage. These findings are suggestive of neuronal insult as a result of external factors, rather than some type of developmental mishap. Various xenobiotics have been suggested as possible causes of this pathology. In a recent review, the top ten environmental compounds suspected of causing autism and learning disabilities were listed and they included: lead, methyl-mercury, polychorinated biphenyls, organophosphate pesticides, organochlorine pesticides, endocrine disruptors, automotive exhaust, polycyclic aromatic hydrocarbons, polybrominated diphenyl ethers, and perfluorinated compounds. This current review, however, will focus specifically on mercury exposure and ASD by conducting a comprehensive literature search of original studies in humans that examine the potential relationship between mercury and ASD, categorizing, summarizing, and discussing the published research that addresses this topic. This review found 91 studies that examine the potential relationship between mercury and ASD from 1999 to February 2016. Of these studies, the vast majority (74%) suggest that mercury is a risk factor for ASD, revealing both direct and indirect effects. The preponderance of the evidence indicates that mercury exposure is causal and/or contributory in ASD.
Collapse
Affiliation(s)
- Janet K Kern
- Institute of Chronic Illnesses, Inc., 14 Redgate Court, Silver Spring, MD, 20905 USA; Council for Nutritional and Environmental Medicine, Mo i Rana, Norway; CoMeD, Inc., 14 Redgate Court, Silver Spring, MD, 20905 USA.
| | - David A Geier
- Institute of Chronic Illnesses, Inc., 14 Redgate Court, Silver Spring, MD, 20905 USA; CoMeD, Inc., 14 Redgate Court, Silver Spring, MD, 20905 USA
| | - Lisa K Sykes
- CoMeD, Inc., 14 Redgate Court, Silver Spring, MD, 20905 USA
| | - Boyd E Haley
- University of Kentucky, 410 Administration Drive, Lexington, KY, 40506 USA
| | - Mark R Geier
- Institute of Chronic Illnesses, Inc., 14 Redgate Court, Silver Spring, MD, 20905 USA; CoMeD, Inc., 14 Redgate Court, Silver Spring, MD, 20905 USA
| |
Collapse
|
137
|
Li YM, Ou JJ, Liu L, Zhang D, Zhao JP, Tang SY. Association Between Maternal Obesity and Autism Spectrum Disorder in Offspring: A Meta-analysis. J Autism Dev Disord 2016; 46:95-102. [PMID: 26254893 DOI: 10.1007/s10803-015-2549-8] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
As the link between maternal obesity and risk of autism among offspring is unclear, the present study assessed this association. A systematic search of an electronic database was performed to identify observational studies that examined the association between maternal obesity and autism. The outcome measures were odds ratios comparing offspring autism risk between obese and normal-weight mothers. Five observational studies were included in the meta-analysis. A fixed-effects model was used since low heterogeneity was observed between studies. The pooled adjusted odds ratio was 1.47 (95 % CI 1.24-1.74). The meta-analysis results support an increased risk of autism spectrum disorder in children of women who were obese during pregnancy. However, further study is warranted to confirm these results.
Collapse
Affiliation(s)
- Ya-Min Li
- The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China.,Xiangya Nursing School, Central South University, Changsha, Hunan Province, China
| | - Jian-Jun Ou
- The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Li Liu
- Xiangya Nursing School, Central South University, Changsha, Hunan Province, China
| | - Dan Zhang
- Xiangya Nursing School, Central South University, Changsha, Hunan Province, China
| | - Jing-Ping Zhao
- The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Si-Yuan Tang
- Xiangya Nursing School, Central South University, Changsha, Hunan Province, China.
| |
Collapse
|
138
|
Qasem H, Al-Ayadhi L, El-Ansary A. Cysteinyl leukotriene correlated with 8-isoprostane levels as predictive biomarkers for sensory dysfunction in autism. Lipids Health Dis 2016; 15:130. [PMID: 27530350 PMCID: PMC4988023 DOI: 10.1186/s12944-016-0298-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 07/28/2016] [Indexed: 12/18/2022] Open
Abstract
Background Autism is a neurodevelopmental disorder that clinically presented as cognitive deficits, social impairments and sensory dysfunction. An increasing body of evidence has shown that oxidative stress and inflammation are involved in the pathophysiology of autism. Recording biomarkers as measure of the severity of autistic features might help in understanding the pathophysiology of autism. Methods This study investigates the plasma levels of 8-isoprostane and Cysteinyl leukotrienes (CysLTs) in 44 autistic children and 40 healthy controls. The recruited autistic patients were assessed for behavior, cognitive and sensory deficits by using different autism severity rating scales, including the Childhood Autism Rating Scales (CARS), Social responsiveness scale (SRS) and Short Sensory Profile (SSP). Receiver Operating Characteristics analysis (ROC) of the obtained data was performed to measure the predictive value of 8-isoprostane and Cysteinyl leukotrienes (CysLTs) as oxidative stress- related parameters. Pearson’s correlations between the measured parameters was also performed. Results The concentrations of 8-isoprostane and CysLTs in autistic patients were significantly higher than those in controls. While cognitive and social impairments did not show any significant differences, the SSP results were strongly correlated with the levels of both of the biomarkers assessed. However, autistic children showed improvements in oxidative stress status (as determined by 8-isoprostane levels) at increasing ages. Conclusion This study indicates that 8-isoprostane and CysLTs can be used as markers for the early recognition of autistic patients through sensory deficits phenotypes which might help early intervention.
Collapse
Affiliation(s)
- Hanan Qasem
- Biochemistry Department, Science College, King Saud University, P.O. Box 22452, Riyadh, 11495, Saudi Arabia
| | - Laila Al-Ayadhi
- Autism Research and Treatment Center, Riyadh, Saudi Arabia.,Shaik AL-Amodi Autism Research Chair, King Saud University, Riyadh, Saudi Arabia.,Department of Physiology, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Afaf El-Ansary
- Central Laboratory, Female Center for Medical Studies and Scientific Section, King Saud University, Riyadh, KSA, Saudi Arabia. .,Autism Research and Treatment Center, Riyadh, Saudi Arabia. .,Shaik AL-Amodi Autism Research Chair, King Saud University, Riyadh, Saudi Arabia. .,Medicinal Chemistry Department, National Research Centre, Dokki, Cairo, Egypt.
| |
Collapse
|
139
|
Abstract
Microglia constitute the powerhouse of the innate immune system in the brain. It is now widely accepted that they are monocytic-derived cells that infiltrate the developing brain at the early embryonic stages, and acquire a resting phenotype characterized by the presence of dense branching processes, called ramifications. Microglia use these dynamic ramifications as sentinels to sense and detect any occurring alteration in brain homeostasis. Once a danger signal is detected, such as molecular factors associated to brain damage or infection, they get activated by acquiring a less ramified phenotype, and mount adequate responses that range from phagocyting cell debris to secreting inflammatory and trophic factors. Here, we review the origin of microglia and we summarize the main molecular signals involved in controlling their function under physiological conditions. In addition, their implication in the pathogenesis of multiple sclerosis and stress is discussed.
Collapse
Affiliation(s)
- Ayman ElAli
- Neuroscience Laboratory, CHU de Québec Research Center (CHUL), Department of Psychiatry and Neuroscience, Faculty of Medicine, Laval University Quebec, CA, Canada
| | - Serge Rivest
- Neuroscience Laboratory, CHU de Québec Research Center (CHUL), Department of Molecular Medicine, Faculty of Medicine, Laval University Quebec, CA, Canada
| |
Collapse
|
140
|
De Felice A, Greco A, Calamandrei G, Minghetti L. Prenatal exposure to the organophosphate insecticide chlorpyrifos enhances brain oxidative stress and prostaglandin E2 synthesis in a mouse model of idiopathic autism. J Neuroinflammation 2016; 13:149. [PMID: 27301868 PMCID: PMC4908699 DOI: 10.1186/s12974-016-0617-4] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 06/07/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Autism spectrum disorders (ASD) are emerging as polygenic and multifactorial disorders in which complex interactions between defective genes and early exposure to environmental stressors impact on the correct neurodevelopment and brain processes. Organophosphate insecticides, among which chlorpyrifos (CPF), are widely diffused environmental toxicants associated with neurobehavioral deficits and increased risk of ASD occurrence in children. Oxidative stress and dysregulated immune responses are implicated in both organophosphate neurodevelopmental effects and ASD etiopathogenesis. BTBR T+tf/J mice, a well-studied model of idiopathic autism, show several behavioral and immunological alterations found in ASD children, and we recently showed that CPF gestational exposure strengthened some of these autistic-like traits. In the present study, we aimed at investigating whether the behavioral effects of gestational CPF administration are associated with brain increased oxidative stress and altered lipid mediator profile. METHODS Brain levels of F2-isoprostanes (15-F2t-IsoP), as index of in vivo oxidative stress, and prostaglandin E2 (PGE2), a major arachidonic acid metabolite released by immune cells and by specific glutamatergic neuron populations mainly in cortex and hippocampus, were assessed by specific enzyme-immuno assays in brain homogenates from BTBR T+tf/J and C57Bl6/J mice, exposed during gestation to either vehicle or CPF. Measures were performed in mice of both sexes, at different postnatal stages (PNDs 1, 21, and 70). RESULTS At birth, BTBR T+tf/J mice exhibited higher baseline 15-F2t-IsoP levels as compared to C57Bl6/J mice, suggestive of greater oxidative stress processes. Gestational treatment with CPF-enhanced 15-F2t-IsoP and PGE2 levels in strain- and age-dependent manner, with 15-F2t-IsoP increased in BTBR T+tf/J mice at PNDs 1 and 21, and PGE2 elevated in BTBR T+tf/J mice at PNDs 21 and 70. At PND 21, CPF effects were sex-dependent being the increase of the two metabolites mainly associated with male mice. CPF treatment also induced a reduction of somatic growth, which reached statistical significance at PND 21. CONCLUSIONS These findings indicate that the autistic-like BTBR T+tf/J strain is highly vulnerable to environmental stressors during gestational period. The results further support the hypothesis that oxidative stress might be the link between environmental neurotoxicants such as CPF and ASD. The increased levels of oxidative stress during early postnatal life could result in delayed and long-lasting alterations in specific pathways relevant to ASD, of which PGE2 signaling represents an important one.
Collapse
Affiliation(s)
- Alessia De Felice
- Section of Neurotoxicology and Neuroendocrinology, Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Viale Regina Elena 299, I-00161, Rome, Italy
- Present address: Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, Via Bettini 31, 38068, Rovereto (TN), Italy
| | - Anita Greco
- Section of Experimental Neurology, Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Viale Regina Elena 299, I-00161, Rome, Italy
| | - Gemma Calamandrei
- Section of Neurotoxicology and Neuroendocrinology, Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Viale Regina Elena 299, I-00161, Rome, Italy
| | - Luisa Minghetti
- Section of Experimental Neurology, Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Viale Regina Elena 299, I-00161, Rome, Italy.
| |
Collapse
|
141
|
Prins BP, Abbasi A, Wong A, Vaez A, Nolte I, Franceschini N, Stuart PE, Guterriez Achury J, Mistry V, Bradfield JP, Valdes AM, Bras J, Shatunov A, Lu C, Han B, Raychaudhuri S, Bevan S, Mayes MD, Tsoi LC, Evangelou E, Nair RP, Grant SFA, Polychronakos C, Radstake TRD, van Heel DA, Dunstan ML, Wood NW, Al-Chalabi A, Dehghan A, Hakonarson H, Markus HS, Elder JT, Knight J, Arking DE, Spector TD, Koeleman BPC, van Duijn CM, Martin J, Morris AP, Weersma RK, Wijmenga C, Munroe PB, Perry JRB, Pouget JG, Jamshidi Y, Snieder H, Alizadeh BZ. Investigating the Causal Relationship of C-Reactive Protein with 32 Complex Somatic and Psychiatric Outcomes: A Large-Scale Cross-Consortium Mendelian Randomization Study. PLoS Med 2016; 13:e1001976. [PMID: 27327646 PMCID: PMC4915710 DOI: 10.1371/journal.pmed.1001976] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2015] [Accepted: 02/03/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND C-reactive protein (CRP) is associated with immune, cardiometabolic, and psychiatric traits and diseases. Yet it is inconclusive whether these associations are causal. METHODS AND FINDINGS We performed Mendelian randomization (MR) analyses using two genetic risk scores (GRSs) as instrumental variables (IVs). The first GRS consisted of four single nucleotide polymorphisms (SNPs) in the CRP gene (GRSCRP), and the second consisted of 18 SNPs that were significantly associated with CRP levels in the largest genome-wide association study (GWAS) to date (GRSGWAS). To optimize power, we used summary statistics from GWAS consortia and tested the association of these two GRSs with 32 complex somatic and psychiatric outcomes, with up to 123,865 participants per outcome from populations of European ancestry. We performed heterogeneity tests to disentangle the pleiotropic effect of IVs. A Bonferroni-corrected significance level of less than 0.0016 was considered statistically significant. An observed p-value equal to or less than 0.05 was considered nominally significant evidence for a potential causal association, yet to be confirmed. The strengths (F-statistics) of the IVs were 31.92-3,761.29 and 82.32-9,403.21 for GRSCRP and GRSGWAS, respectively. CRP GRSGWAS showed a statistically significant protective relationship of a 10% genetically elevated CRP level with the risk of schizophrenia (odds ratio [OR] 0.86 [95% CI 0.79-0.94]; p < 0.001). We validated this finding with individual-level genotype data from the schizophrenia GWAS (OR 0.96 [95% CI 0.94-0.98]; p < 1.72 × 10-6). Further, we found that a standardized CRP polygenic risk score (CRPPRS) at p-value thresholds of 1 × 10-4, 0.001, 0.01, 0.05, and 0.1 using individual-level data also showed a protective effect (OR < 1.00) against schizophrenia; the first CRPPRS (built of SNPs with p < 1 × 10-4) showed a statistically significant (p < 2.45 × 10-4) protective effect with an OR of 0.97 (95% CI 0.95-0.99). The CRP GRSGWAS showed that a 10% increase in genetically determined CRP level was significantly associated with coronary artery disease (OR 0.88 [95% CI 0.84-0.94]; p < 2.4 × 10-5) and was nominally associated with the risk of inflammatory bowel disease (OR 0.85 [95% CI 0.74-0.98]; p < 0.03), Crohn disease (OR 0.81 [95% CI 0.70-0.94]; p < 0.005), psoriatic arthritis (OR 1.36 [95% CI 1.00-1.84]; p < 0.049), knee osteoarthritis (OR 1.17 [95% CI 1.01-1.36]; p < 0.04), and bipolar disorder (OR 1.21 [95% CI 1.05-1.40]; p < 0.007) and with an increase of 0.72 (95% CI 0.11-1.34; p < 0.02) mm Hg in systolic blood pressure, 0.45 (95% CI 0.06-0.84; p < 0.02) mm Hg in diastolic blood pressure, 0.01 ml/min/1.73 m2 (95% CI 0.003-0.02; p < 0.005) in estimated glomerular filtration rate from serum creatinine, 0.01 g/dl (95% CI 0.0004-0.02; p < 0.04) in serum albumin level, and 0.03 g/dl (95% CI 0.008-0.05; p < 0.009) in serum protein level. However, after adjustment for heterogeneity, neither GRS showed a significant effect of CRP level (at p < 0.0016) on any of these outcomes, including coronary artery disease, nor on the other 20 complex outcomes studied. Our study has two potential limitations: the limited variance explained by our genetic instruments modeling CRP levels in blood and the unobserved bias introduced by the use of summary statistics in our MR analyses. CONCLUSIONS Genetically elevated CRP levels showed a significant potentially protective causal relationship with risk of schizophrenia. We observed nominal evidence at an observed p < 0.05 using either GRSCRP or GRSGWAS-with persistence after correction for heterogeneity-for a causal relationship of elevated CRP levels with psoriatic osteoarthritis, rheumatoid arthritis, knee osteoarthritis, systolic blood pressure, diastolic blood pressure, serum albumin, and bipolar disorder. These associations remain yet to be confirmed. We cannot verify any causal effect of CRP level on any of the other common somatic and neuropsychiatric outcomes investigated in the present study. This implies that interventions that lower CRP level are unlikely to result in decreased risk for the majority of common complex outcomes.
Collapse
Affiliation(s)
- Bram. P. Prins
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Human Genetics, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
- * E-mail: (BPP); (BZA)
| | - Ali Abbasi
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom
- Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Anson Wong
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Ahmad Vaez
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ilja Nolte
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Nora Franceschini
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Philip E. Stuart
- Department of Dermatology, Veterans Affairs Hospital, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Javier Guterriez Achury
- Department of Genetics, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Vanisha Mistry
- Metabolic Research Laboratories, Wellcome Trust–MRC Institute of Metabolic Science, Addenbrooke’s Hospital, University of Cambridge, Cambridge, United Kingdom
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Jonathan P. Bradfield
- Center for Applied Genomics, Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, United States of America
| | - Ana M. Valdes
- Department of Academic Rheumatology, University of Nottingham, Nottingham, United Kingdom
| | - Jose Bras
- Department of Molecular Neuroscience, Institute of Neurology, London, United Kingdom
| | - Aleksey Shatunov
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - PAGE Consortium
- Department of Dermatology, Veterans Affairs Hospital, University of Michigan, Ann Arbor, Michigan, United States of America
| | | | - Systemic Sclerosis consortium
- Division of Rheumatology and Clinical Immunogenetics, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
- Instituto de Parasitologia y Biomedicina Lopez-Neyra, Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - Treat OA consortium
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - DIAGRAM Consortium
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | | | - ALS consortium
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | | | | | - CKDGen consortium
- NHLBI’s Framingham Heart Study, Center for Population Studies and Harvard Medical School, Framingham, Massachusetts, United States of America
| | - GERAD1 Consortium
- Institute of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | | | | | | | - Chen Lu
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
| | - Buhm Han
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, United States of America
| | - Soumya Raychaudhuri
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, United States of America
- Division of Genetics, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Rheumatology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Partners HealthCare Center for Personalized Genetic Medicine, Boston, Massachusetts, United States of America
- Faculty of Medical and Human Sciences, University of Manchester, Manchester, United Kingdom
| | - Steve Bevan
- Neurology Unit, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Maureen D. Mayes
- Division of Rheumatology and Clinical Immunogenetics, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Lam C. Tsoi
- Department of Dermatology, Veterans Affairs Hospital, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Evangelos Evangelou
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
| | - Rajan P. Nair
- Department of Dermatology, Veterans Affairs Hospital, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Struan F. A. Grant
- Center for Applied Genomics, Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, United States of America
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States of America
| | - Constantin Polychronakos
- Endocrine Genetics Research Institute, McGill University Health Center, Montreal, Quebec, Canada
| | - Timothy R. D. Radstake
- Department of Rheumatology & Clinical Immunology and Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - David A. van Heel
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Melanie L. Dunstan
- Institute of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Nicholas W. Wood
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, United Kingdom
| | - Ammar Al-Chalabi
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- Complex Disease Genetics, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, United States of America
| | - Abbas Dehghan
- Department of Epidemiology, Erasmus University Rotterdam, University Medical Centre Rotterdam, Rotterdam, the Netherlands
| | - Hakon Hakonarson
- Center for Applied Genomics, Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, United States of America
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States of America
| | - Hugh S. Markus
- Neurology Unit, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - James T. Elder
- Department of Dermatology, Veterans Affairs Hospital, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Jo Knight
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Biostatistics Division, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Dan E. Arking
- McKusick-Nathans Institute of Genetic Medicine and Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Timothy D. Spector
- Department of Twin Research and Genetic Epidemiology, King’s College London, London, United Kingdom
| | - Bobby P. C. Koeleman
- Complex Genetic Section, Department of Medical Genetics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Cornelia M. van Duijn
- Department of Epidemiology, Erasmus University Rotterdam, University Medical Centre Rotterdam, Rotterdam, the Netherlands
| | - Javier Martin
- Instituto de Parasitologia y Biomedicina Lopez-Neyra, Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - Andrew P. Morris
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Department of Biostatistics, University of Liverpool, Liverpool, United Kingdom
| | - Rinse K. Weersma
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Cisca Wijmenga
- Department of Genetics, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Patricia B. Munroe
- NIHR Barts Cardiovascular Biomedical Research Unit, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
- Clinical Pharmacology, William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London, London, United Kingdom
| | - John R. B. Perry
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Jennie G. Pouget
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Yalda Jamshidi
- Cardiogenetics Lab, Cardiovascular and Cell Sciences Institute, St George’s Hospital Medical School, London, United Kingdom
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Behrooz Z. Alizadeh
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- * E-mail: (BPP); (BZA)
| |
Collapse
|
142
|
Şimşek Ş, Çetin İ, Çim A, Kaya S. Elevated levels of tissue plasminogen activator and E-selectin in male children with autism spectrum disorder. Autism Res 2016; 9:1241-1247. [DOI: 10.1002/aur.1638] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 03/01/2016] [Accepted: 04/04/2016] [Indexed: 12/30/2022]
Affiliation(s)
- Şeref Şimşek
- Department of Child Psychiatry; Dicle University, Medical School; Diyarbakır Turkey
| | - İhsan Çetin
- Department of Nutrition and Dietetics; Batman University, School of Health Sciences; Batman Turkey
| | - Abdullah Çim
- Department of Medical Genetics; Dicle University, Medical School; Diyarbakır Turkey
| | - Savaş Kaya
- Department of Immunology; Dicle University, Medical School; Diyarbakır Turkey
| |
Collapse
|
143
|
Lee JH, Espinera AR, Chen D, Choi KE, Caslin AY, Won S, Pecoraro V, Xu GY, Wei L, Yu SP. Neonatal inflammatory pain and systemic inflammatory responses as possible environmental factors in the development of autism spectrum disorder of juvenile rats. J Neuroinflammation 2016; 13:109. [PMID: 27184741 PMCID: PMC4867541 DOI: 10.1186/s12974-016-0575-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 05/06/2016] [Indexed: 02/28/2023] Open
Abstract
Background Autism spectrum disorder (ASD) affects many children and juveniles. The pathogenesis of ASD is not well understood. Environmental factors may play important roles in the development of ASD. We examined a possible relationship of inflammatory pain in neonates and the development of ASD in juveniles. Methods Acute inflammation pain was induced by 5 % formalin (5 μl/day) subcutaneous injection into two hindpaws of postnatal day 3 to 5 (P3–P5) rat pups. Western blot, immunohistochemical, and behavioral examinations were performed at different time points after the insult. Results Formalin injection caused acute and chronic inflammatory responses including transient local edema, increased levels of inflammatory cytokines, TNF-α, and IL-1β in the blood as well as in the brain, and increased microglia in the brain. One day after the pain insult, there was significant cell death in the cortex and hippocampus. Two weeks later, although the hindpaw local reaction subsided, impaired axonal growth and demyelization were seen in the brain of P21 juvenile rats. The number of bromodeoxyuridine (BrdU) and doublecortin (DCX) double-positive cells in the hippocampal dentate gyrus of P21 rats was significantly lower than that in controls, indicating reduced neurogenesis. In the P21 rat’s brain of the formalin group, the expression of autism-related gene neurexin 1 (NRXN1), fragile X mental retardation 1 (FMR1), and oxytocin was significantly downregulated, consistent with the gene alteration in ASD. Juvenile rats in the formalin group showed hyperalgesia, repetitive behaviors, abnormal locomotion, sleep disorder, and distinct deficits in social memory and social activities. These alterations in neuroinflammatory reactions, gene expression, and behaviors were more evident in male than in female rats. Importantly, an anti-inflammation treatment using indomethacin (10 mg/kg, i.p.) at the time of formalin injections suppressed inflammatory responses and neuronal cell death and prevented alterations in ASD-related genes and the development of abnormal behaviors. Conclusions These novel observations indicate that severe inflammatory pain in neonates and persistent inflammatory reactions may predispose premature infants to development delays and psychiatric disorders including ASD. The prevention of pain stimuli and prompt treatments of inflammation during development appear vitally important in disrupting possible evolution of ASD syndromes. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0575-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jin Hwan Lee
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Alyssa R Espinera
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Dongdong Chen
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA.,The Laboratory of Translational Pain Medicine, Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Ko-Eun Choi
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Asha Yoshiko Caslin
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Soonmi Won
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Valentina Pecoraro
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Guang-Yin Xu
- The Laboratory of Translational Pain Medicine, Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA.,Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Shan Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA. .,Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Atlanta, GA, 30033, USA. .,Emory University School of Medicine, 101 Woodruff Circle, WMB Suite 620, Atlanta, GA, 30322, USA.
| |
Collapse
|
144
|
Crowley T, Cryan JF, Downer EJ, O'Leary OF. Inhibiting neuroinflammation: The role and therapeutic potential of GABA in neuro-immune interactions. Brain Behav Immun 2016; 54:260-277. [PMID: 26851553 DOI: 10.1016/j.bbi.2016.02.001] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 01/22/2016] [Accepted: 02/02/2016] [Indexed: 12/25/2022] Open
Abstract
The central nervous system, once thought to be a site of immunological privilege, has since been found to harbour immunocompetent cells and to communicate with the peripheral nervous system. In the central nervous system (CNS), glial cells display immunological responses to pathological and physiological stimuli through pro- and anti-inflammatory cytokine and chemokine signalling, antigen presentation and the clearing of cellular debris through phagocytosis. While this neuroinflammatory signalling can act to reduce neuronal damage and comprises a key facet of CNS homeostasis, persistent inflammation or auto-antigen-mediated immunoreactivity can induce a positive feedback cycle of neuroinflammation that ultimately results in necrosis of glia and neurons. Persistent neuroinflammation has been recognised as a major pathological component of virtually all neurodegenerative diseases and has also been a focus of research into the pathology underlying psychiatric disorders. Thus, pharmacological strategies to curb the pathological effects of persistent neuroinflammation are of interest for many disorders of the CNS. Accumulating evidence suggests that GABAergic activities are closely bound to immune processes and signals, and thus the GABAergic neurotransmitter system might represent an important therapeutic target in modulating neuroinflammation. Here, we review evidence that inflammation induces changes in the GABA neurotransmitter system in the CNS and that GABAergic signalling exerts a reciprocal influence over neuroinflammatory processes. Together, the data support the hypothesis that the GABA system is a potential therapeutic target in the modulation of central inflammation.
Collapse
Affiliation(s)
- Tadhg Crowley
- Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Ireland; APC Microbiome Institute, University College Cork, Ireland
| | - Eric J Downer
- School of Medicine, Discipline of Physiology, Trinity Biomedical Sciences Institute, Trinity College, Dublin 2, Ireland.
| | - Olivia F O'Leary
- Department of Anatomy and Neuroscience, University College Cork, Ireland; APC Microbiome Institute, University College Cork, Ireland.
| |
Collapse
|
145
|
Kim KN, Kwon HJ, Hong YC. Low-level lead exposure and autistic behaviors in school-age children. Neurotoxicology 2016; 53:193-200. [PMID: 26877220 DOI: 10.1016/j.neuro.2016.02.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 01/07/2016] [Accepted: 02/08/2016] [Indexed: 01/30/2023]
Abstract
INTRODUCTION The association between lead exposure and autism spectrum disorder is inconclusive. We hypothesized an association between higher blood lead concentrations and more autistic behaviors, including impaired social interactions and communication, stereotypical behaviors, and restricted interests, among school-age children. METHODS Data from 2473 Korean children aged 7-8years who had no prior history of developmental disorders were analyzed. Two follow-up surveys were conducted biennially until the children reached 11-12years of age. Blood lead concentrations were measured at every survey, and autistic behaviors were evaluated at 11-12years of age using the Autism Spectrum Screening Questionnaire (ASSQ) and Social Responsiveness Scale (SRS). The associations of blood lead concentration with ASSQ and SRS scores were analyzed using negative binomial, logistic, and linear regression models. RESULTS Blood lead concentrations at 7-8years of age (geometric mean: 1.64μg/dL), but not at 9-10 and 11-12years of age, were associated with more autistic behaviors at 11-12years of age, according to the ASSQ (β=0.151; 95% confidence interval [CI]: 0.061, 0.242) and SRS (β=2.489; 95% CI: 1.378, 3.600). SRS subscale analysis also revealed associations between blood lead concentrations and social awareness, cognition, communication, motivation, and mannerisms. CONCLUSION Even low blood lead concentrations at 7-8years of age are associated with more autistic behaviors at 11-12years of age, underscoring the need for continued efforts to reduce lead exposure.
Collapse
Affiliation(s)
- Kyoung-Nam Kim
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ho-Jang Kwon
- Department of Preventive Medicine and Public Health, Dankook University College of Medicine, Cheonan, Republic of Korea
| | - Yun-Chul Hong
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea; Institute of Environmental Medicine, Medical Research Center, Seoul, Republic of Korea; Environmental Health Center, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
146
|
Young AMH, Chakrabarti B, Roberts D, Lai MC, Suckling J, Baron-Cohen S. From molecules to neural morphology: understanding neuroinflammation in autism spectrum condition. Mol Autism 2016; 7:9. [PMID: 26793298 PMCID: PMC4719563 DOI: 10.1186/s13229-016-0068-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 01/04/2016] [Indexed: 12/31/2022] Open
Abstract
Growing evidence points toward a critical role for early (prenatal) atypical neurodevelopmental processes in the aetiology of autism spectrum condition (ASC). One such process that could impact early neural development is inflammation. We review the evidence for atypical expression of molecular markers in the amniotic fluid, serum, cerebrospinal fluid (CSF), and the brain parenchyma that suggest a role for inflammation in the emergence of ASC. This is complemented with a number of neuroimaging and neuropathological studies describing microglial activation. Implications for treatment are discussed.
Collapse
Affiliation(s)
- Adam M H Young
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Douglas House, 18B Trumpington Road, Cambridge, UK ; School of Clinical Medicine, Addenbrookes Hospital, University of Cambridge, Cambridge, UK
| | - Bhismadev Chakrabarti
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Douglas House, 18B Trumpington Road, Cambridge, UK ; Centre for Integrative Neuroscience and Neurodynamics, School of Psychology and Clinical Language Science, University of Reading, Reading, UK
| | - David Roberts
- School of Clinical Medicine, Addenbrookes Hospital, University of Cambridge, Cambridge, UK
| | - Meng-Chuan Lai
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Douglas House, 18B Trumpington Road, Cambridge, UK ; Centre for Addiction and Mental Health and Department of Psychiatry, University of Toronto, Toronto, Canada ; Department of Psychiatry, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - John Suckling
- Brain Mapping Unit, Department of Psychiatry, University of Cambridge, Cambridge, UK ; Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
| | - Simon Baron-Cohen
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Douglas House, 18B Trumpington Road, Cambridge, UK ; CLASS Clinic, Cambridgeshire and Peterborough NHS Foundation Trust, Cambridge, UK
| |
Collapse
|
147
|
Kern JK, Geier DA, Sykes LK, Geier MR. Relevance of Neuroinflammation and Encephalitis in Autism. Front Cell Neurosci 2016; 9:519. [PMID: 26834565 PMCID: PMC4717322 DOI: 10.3389/fncel.2015.00519] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Accepted: 12/24/2015] [Indexed: 12/12/2022] Open
Abstract
In recent years, many studies indicate that children with an autism spectrum disorder (ASD) diagnosis have brain pathology suggestive of ongoing neuroinflammation or encephalitis in different regions of their brains. Evidence of neuroinflammation or encephalitis in ASD includes: microglial and astrocytic activation, a unique and elevated proinflammatory profile of cytokines, and aberrant expression of nuclear factor kappa-light-chain-enhancer of activated B cells. A conservative estimate based on the research suggests that at least 69% of individuals with an ASD diagnosis have microglial activation or neuroinflammation. Encephalitis, which is defined as inflammation of the brain, is medical diagnosis code G04.90 in the International Classification of Disease, 10th revision; however, children with an ASD diagnosis are not generally assessed for a possible medical diagnosis of encephalitis. This is unfortunate because if a child with ASD has neuroinflammation, then treating the underlying brain inflammation could lead to improved outcomes. The purpose of this review of the literature is to examine the evidence of neuroinflammation/encephalitis in those with an ASD diagnosis and to address how a medical diagnosis of encephalitis, when appropriate, could benefit these children by driving more immediate and targeted treatments.
Collapse
Affiliation(s)
- Janet K. Kern
- Institute of Chronic Illnesses, Inc., Silver SpringMD, USA
| | - David A. Geier
- Institute of Chronic Illnesses, Inc., Silver SpringMD, USA
| | | | - Mark R. Geier
- Institute of Chronic Illnesses, Inc., Silver SpringMD, USA
| |
Collapse
|
148
|
Zaman S, Yazdani U, Deng Y, Li W, Gadad BS, Hynan L, Karp D, Roatch N, Schutte C, Nathan Marti C, Hewitson L, German DC. A Search for Blood Biomarkers for Autism: Peptoids. Sci Rep 2016; 6:19164. [PMID: 26764136 PMCID: PMC4725892 DOI: 10.1038/srep19164] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 11/12/2015] [Indexed: 12/18/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by impairments in social interaction and communication, and restricted, repetitive patterns of behavior. In order to identify individuals with ASD and initiate interventions at the earliest possible age, biomarkers for the disorder are desirable. Research findings have identified widespread changes in the immune system in children with autism, at both systemic and cellular levels. In an attempt to find candidate antibody biomarkers for ASD, highly complex libraries of peptoids (oligo-N-substituted glycines) were screened for compounds that preferentially bind IgG from boys with ASD over typically developing (TD) boys. Unexpectedly, many peptoids were identified that preferentially bound IgG from TD boys. One of these peptoids was studied further and found to bind significantly higher levels (>2-fold) of the IgG1 subtype in serum from TD boys (n = 60) compared to ASD boys (n = 74), as well as compared to older adult males (n = 53). Together these data suggest that ASD boys have reduced levels (>50%) of an IgG1 antibody, which resembles the level found normally with advanced age. In this discovery study, the ASD1 peptoid was 66% accurate in predicting ASD.
Collapse
Affiliation(s)
- Sayed Zaman
- Department of Psychiatry, UT Southwestern Medical Center, Dallas TX
| | - Umar Yazdani
- Department of Psychiatry, UT Southwestern Medical Center, Dallas TX
| | - Yan Deng
- Department of Psychiatry, UT Southwestern Medical Center, Dallas TX
| | - Wenhao Li
- Department of Psychiatry, UT Southwestern Medical Center, Dallas TX
| | - Bharathi S Gadad
- Department of Psychiatry, UT Southwestern Medical Center, Dallas TX
| | - Linda Hynan
- Department of Clinical Sciences, UT Southwestern Medical Center, Dallas TX
| | - David Karp
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas TX
| | - Nichole Roatch
- Johnson Center for Child Health and Development, Austin TX
| | - Claire Schutte
- Johnson Center for Child Health and Development, Austin TX
| | | | - Laura Hewitson
- Department of Psychiatry, UT Southwestern Medical Center, Dallas TX.,Johnson Center for Child Health and Development, Austin TX
| | - Dwight C German
- Department of Psychiatry, UT Southwestern Medical Center, Dallas TX
| |
Collapse
|
149
|
Williamson LL, McKenney EA, Holzknecht ZE, Belliveau C, Rawls JF, Poulton S, Parker W, Bilbo SD. Got worms? Perinatal exposure to helminths prevents persistent immune sensitization and cognitive dysfunction induced by early-life infection. Brain Behav Immun 2016; 51:14-28. [PMID: 26162711 DOI: 10.1016/j.bbi.2015.07.006] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 07/06/2015] [Accepted: 07/06/2015] [Indexed: 02/08/2023] Open
Abstract
The incidence of autoimmune and inflammatory diseases has risen dramatically in post-industrial societies. "Biome depletion" - loss of commensal microbial and multicellular organisms such as helminths (intestinal worms) that profoundly modulate the immune system - may contribute to these increases. Hyperimmune-associated disorders also affect the brain, especially neurodevelopment, and increasing evidence links early-life infection to cognitive and neurodevelopmental disorders. We have demonstrated previously that rats infected with bacteria as newborns display life-long vulnerabilities to cognitive dysfunction, a vulnerability that is specifically linked to long-term hypersensitivity of microglial cell function, the resident immune cells of the brain. Here, we demonstrate that helminth colonization of pregnant dams attenuated the exaggerated brain cytokine response of their offspring to bacterial infection, and that combined with post-weaning colonization of offspring with helminths (consistent with their mothers treatment) completely prevented enduring microglial sensitization and cognitive dysfunction in adulthood. Importantly, helminths had no overt impact on adaptive immune cell subsets, whereas exaggerated innate inflammatory responses in splenic macrophages were prevented. Finally, helminths altered the effect of neonatal infection on the gut microbiome; neonatal infection with Escherichia coli caused a shift from genera within the Actinobacteria and Tenericutes phyla to genera in the Bacteroidetes phylum in rats not colonized with helminths, but helminths attenuated this effect. In sum, these data point toward an inter-relatedness of various components of the biome, and suggest potential mechanisms by which this helminth might exert therapeutic benefits in the treatment of neuroinflammatory and cognitive disorders.
Collapse
Affiliation(s)
- Lauren L Williamson
- Department of Psychology & Neuroscience, Duke Institute for Brain Sciences, Duke University, United States
| | | | - Zoie E Holzknecht
- Department of Surgery, Duke University Medical Center, United States
| | - Christine Belliveau
- Department of Psychology & Neuroscience, Duke Institute for Brain Sciences, Duke University, United States
| | - John F Rawls
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, United States
| | - Susan Poulton
- Department of Surgery, Duke University Medical Center, United States
| | - William Parker
- Department of Surgery, Duke University Medical Center, United States
| | - Staci D Bilbo
- Department of Psychology & Neuroscience, Duke Institute for Brain Sciences, Duke University, United States.
| |
Collapse
|
150
|
Klyushnik TP, Androsova LV, Simashkova NV, Zozulya SA, Otman IN, Shushpanova OV, Brusov OS. Clinical and laboratory diagnosis of autism spectrum disorders in children. ACTA ACUST UNITED AC 2016. [DOI: 10.17116/labs20165222-27] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|