51
|
Prosperi M, Guiducci L, Peroni DG, Narducci C, Gaggini M, Calderoni S, Tancredi R, Morales MA, Gastaldelli A, Muratori F, Santocchi E. Inflammatory Biomarkers are Correlated with Some Forms of Regressive Autism Spectrum Disorder. Brain Sci 2019; 9:brainsci9120366. [PMID: 31835709 PMCID: PMC6955787 DOI: 10.3390/brainsci9120366] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/06/2019] [Accepted: 12/09/2019] [Indexed: 02/07/2023] Open
Abstract
Background: Several studies have tried to investigate the role of inflammatory biomarkers in Autism Spectrum Disorder (ASD), and their correlations with clinical phenotypes. Despite the growing research in this topic, existing data are mostly contradictory. Methods: Eighty-five ASD preschoolers were assessed for developmental level, adaptive functioning, gastrointestinal (GI), socio-communicative and psychopathological symptoms. Plasma levels of leptin, resistin, plasminogen activator inhibitor-1 (PAI-1), macrophage chemoattractant protein-1 (CCL2), tumor necrosis factor-alfa (TNF-α), and interleukin-6 (IL-6) were correlated with clinical scores and were compared among different ASD subgroups according to the presence or absence of: (i) GI symptoms, (ii) regressive onset of autism. Results: Proinflammatory cytokines (TNF-α, IL-6 and CCL2) were lower than those reported in previous studies in children with systemic inflammatory conditions. GI symptoms were not correlated with levels of inflammatory biomarkers except for resistin that was lower in ASD-GI children (p = 0.032). Resistin and PAI-1 levels were significantly higher in the group with “regression plus a developmental delay” onset (Reg+DD group) compared to groups without regression or with regression without a developmental delay (p < 0.01 for all). Conclusions: Our results did not highlight the presence of any systemic inflammatory state in ASD subjects neither disentangling children with/without GI symptoms. The Reg + DD group significantly differed from others in some plasmatic values, but these differences failed to discriminate the subgroups as possible distinct ASD endo-phenotypes.
Collapse
Affiliation(s)
- Margherita Prosperi
- IRCCS Fondazione Stella Maris, Calambrone, 56128 Pisa, Italy; (M.P.); (S.C.); (R.T.); (F.M.); (E.S.)
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy;
| | - Letizia Guiducci
- Institute of Clinical Physiology, CNR, 56124 Pisa, Italy; (L.G.); (M.G.); (M.A.M.)
| | - Diego G. Peroni
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy;
| | - Chiara Narducci
- Child and Adolescent Neuropsychiatry Unit, Department of Biomedical Science, University of Cagliari & “Antonio Cao” Paediatric Hospital, “G. Brotzu” Hospital trust, 09124 Cagliari, Italy;
| | - Melania Gaggini
- Institute of Clinical Physiology, CNR, 56124 Pisa, Italy; (L.G.); (M.G.); (M.A.M.)
| | - Sara Calderoni
- IRCCS Fondazione Stella Maris, Calambrone, 56128 Pisa, Italy; (M.P.); (S.C.); (R.T.); (F.M.); (E.S.)
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy;
| | - Raffaella Tancredi
- IRCCS Fondazione Stella Maris, Calambrone, 56128 Pisa, Italy; (M.P.); (S.C.); (R.T.); (F.M.); (E.S.)
| | - Maria Aurora Morales
- Institute of Clinical Physiology, CNR, 56124 Pisa, Italy; (L.G.); (M.G.); (M.A.M.)
| | - Amalia Gastaldelli
- Institute of Clinical Physiology, CNR, 56124 Pisa, Italy; (L.G.); (M.G.); (M.A.M.)
- Correspondence: ; Tel.: +39-0503-152-679
| | - Filippo Muratori
- IRCCS Fondazione Stella Maris, Calambrone, 56128 Pisa, Italy; (M.P.); (S.C.); (R.T.); (F.M.); (E.S.)
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy;
| | - Elisa Santocchi
- IRCCS Fondazione Stella Maris, Calambrone, 56128 Pisa, Italy; (M.P.); (S.C.); (R.T.); (F.M.); (E.S.)
| |
Collapse
|
52
|
Ahmad SF, Ansari MA, Nadeem A, Bakheet SA, Alsanea S, Al-Hosaini KA, Mahmood HM, Alzahrani MZ, Attia SM. Inhibition of tyrosine kinase signaling by tyrphostin AG126 downregulates the IL-21/IL-21R and JAK/STAT pathway in the BTBR mouse model of autism. Neurotoxicology 2019; 77:1-11. [PMID: 31811869 DOI: 10.1016/j.neuro.2019.12.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 12/02/2019] [Accepted: 12/03/2019] [Indexed: 01/01/2023]
Abstract
Autism spectrum disorder (ASD) comprises a broad range of neurodevelopmental disorders that are associated with deficits in social interaction and communication. The tyrosine kinase inhibitor tyrphostin AG126 represents a promising therapeutic agent for several neuroinflammatory disorders. There are currently no treatments available that can improve ASD and we previously showed that AG126 treatment exerts beneficial effects on BTBR T+ Itpr3tf/J (BTBR) mice, a model for autism that shows the core features of ASD; however, the immunological mechanisms and molecular targets associated with this effect were previously unclear. This study was undertaken to delineate the neuroprotective effect of AG126 on BTBR mice. Here, using this mouse model, we investigated the effects of AG126 administration on IL-21R, IL-21, IL-22, TNF-α, NOS2, STAT3, IL-27, and Foxp3 production by CD8+ T cells in the spleen by flow cytometry. We further explored the mRNA and protein expression of IL-21, IL-22, IL-1β, TNF-α, NOS2, JAK1, STAT3, IL-27, and Foxp3 in brain tissue by RT-PCR, and western blotting. We found that BTBR mice treated with AG126 exhibited significant decreases in IL-21R-, IL-21-, IL-22-, TNF-α-, NOS2-, STAT3-producing, and increases in IL-27- and Foxp3-producing, CD8+ T cells. Our results further demonstrated that AG126 treatment effectively decreased IL-21, IL-22, IL-1β, TNF-α, NOS2, JAK1, and STAT3, and increased IL-27 and Foxp3 mRNA and protein expression in brain tissues. Our findings suggest that AG126 elicits a neuroprotective response through downregulation of the IL-21/IL-21R and JAK/STAT pathway in BTBR mice, which could represent a promising novel therapeutic target for ASD treatment.
Collapse
Affiliation(s)
- Sheikh F Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.
| | - Mushtaq A Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Saleh A Bakheet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Sary Alsanea
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Khaled A Al-Hosaini
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Hafiz M Mahmood
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohammad Z Alzahrani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
53
|
Chaganti S, Welty VF, Taylor W, Albert K, Failla MD, Cascio C, Smith S, Mawn L, Resnick SM, Beason-Held LL, Bagnato F, Lasko T, Blume JD, Landman BA. Discovering novel disease comorbidities using electronic medical records. PLoS One 2019; 14:e0225495. [PMID: 31774837 PMCID: PMC6880990 DOI: 10.1371/journal.pone.0225495] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 09/22/2019] [Indexed: 11/18/2022] Open
Abstract
Increasing reliance on electronic medical records at large medical centers provides unique opportunities to perform population level analyses exploring disease progression and etiology. The massive accumulation of diagnostic, procedure, and laboratory codes in one place has enabled the exploration of co-occurring conditions, their risk factors, and potential prognostic factors. While most of the readily identifiable associations in medical records are (now) well known to the scientific community, there is no doubt many more relationships are still to be uncovered in EMR data. In this paper, we introduce a novel finding index to help with that task. This new index uses data mined from real-time PubMed abstracts to indicate the extent to which empirically discovered associations are already known (i.e., present in the scientific literature). Our methods leverage second-generation p-values, which better identify associations that are truly clinically meaningful. We illustrate our new method with three examples: Autism Spectrum Disorder, Alzheimer's Disease, and Optic Neuritis. Our results demonstrate wide utility for identifying new associations in EMR data that have the highest priority among the complex web of correlations and causalities. Data scientists and clinicians can work together more effectively to discover novel associations that are both empirically reliable and clinically understudied.
Collapse
Affiliation(s)
- Shikha Chaganti
- Department of Electrical Engineering and Computer Science, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Valerie F. Welty
- Department of Biostatistics, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Warren Taylor
- Department of Psychiatry & Behavioral Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Kimberly Albert
- Department of Psychiatry & Behavioral Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Michelle D. Failla
- Department of Psychiatry & Behavioral Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Carissa Cascio
- Department of Psychiatry & Behavioral Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Seth Smith
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Louise Mawn
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Susan M. Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Lori L. Beason-Held
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Francesca Bagnato
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Thomas Lasko
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Jeffrey D. Blume
- Department of Biostatistics, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Bennett A. Landman
- Department of Electrical Engineering and Computer Science, Vanderbilt University, Nashville, Tennessee, United States of America
| |
Collapse
|
54
|
Ristori MV, Quagliariello A, Reddel S, Ianiro G, Vicari S, Gasbarrini A, Putignani L. Autism, Gastrointestinal Symptoms and Modulation of Gut Microbiota by Nutritional Interventions. Nutrients 2019; 11:nu11112812. [PMID: 31752095 PMCID: PMC6893818 DOI: 10.3390/nu11112812] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 11/07/2019] [Accepted: 11/13/2019] [Indexed: 02/07/2023] Open
Abstract
Autism spectrum disorder (ASD) is a complex behavioral syndrome that is characterized by speech and language disorders, intellectual impairment, learning and motor dysfunctions. Several genetic and environmental factors are suspected to affect the ASD phenotype including air pollution, exposure to pesticides, maternal infections, inflammatory conditions, dietary factors or consumption of antibiotics during pregnancy. Many children with ASD shows abnormalities in gastrointestinal (GI) physiology, including increased intestinal permeability, overall microbiota alterations, and gut infection. Moreover, they are "picky eaters" and the existence of specific sensory patterns in ASD patients could represent one of the main aspects in hampering feeding. GI disorders are associated with an altered composition of the gut microbiota. Gut microbiome is able to communicate with brain activities through microbiota-derived signaling molecules, immune mediators, gut hormones as well as vagal and spinal afferent neurons. Since the diet induces changes in the intestinal microbiota and in the production of molecules, such as the SCFA, we wanted to investigate the role that nutritional intervention can have on GI microbiota composition and thus on its influence on behavior, GI symptoms and microbiota composition and report which are the beneficial effect on ASD conditions.
Collapse
Affiliation(s)
- Maria Vittoria Ristori
- Unit of Human Microbiome, Children’s Hospital and Research Institute “Bambino Gesù”, IRCCS, Piazza Sant’Onofrio 4, 00165 Rome, Italy; (M.V.R.); (A.Q.); (S.R.)
| | - Andrea Quagliariello
- Unit of Human Microbiome, Children’s Hospital and Research Institute “Bambino Gesù”, IRCCS, Piazza Sant’Onofrio 4, 00165 Rome, Italy; (M.V.R.); (A.Q.); (S.R.)
| | - Sofia Reddel
- Unit of Human Microbiome, Children’s Hospital and Research Institute “Bambino Gesù”, IRCCS, Piazza Sant’Onofrio 4, 00165 Rome, Italy; (M.V.R.); (A.Q.); (S.R.)
| | - Gianluca Ianiro
- Dipartimento di Gastroenterologia, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy;
| | - Stefano Vicari
- Neuropsichiatria dell’infanzia e dell’adolescenza, Children’s Hospital and Research Institute “Bambino Gesù”, IRCCS, Piazza Sant’Onofrio 4, 00165 Rome, Italy;
| | - Antonio Gasbarrini
- Istituto di Patologia Speciale Medica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- UOC Medicina Interna e Gastroenterologia, Area Gastroenterologia ed Oncologia Medica, Dipartimento di Scienze Gastroenterologiche, Endocrino-Metaboliche e Nefro-Urologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Correspondence: (A.G.); (L.P.); Tel.: +39-0668-59-4127 (L.P.)
| | - Lorenza Putignani
- Units of Parasitology and Human Microbiome, Children’s Hospital and Research Institute “Bambino Gesù”, IRCCS, Piazza Sant’Onofrio 4, 00165 Rome, Italy
- Correspondence: (A.G.); (L.P.); Tel.: +39-0668-59-4127 (L.P.)
| |
Collapse
|
55
|
Ahmad SF, Nadeem A, Ansari MA, Bakheet SA, Alasmari F, Alasmari AF, Al-Kharashi LA, Al-Qahtani Q, Attia SM. The potent immunomodulatory compound VGX-1027 regulates inflammatory mediators in CD4+ T cells, which are concomitant with the prevention of neuroimmune dysregulation in BTBR T+ Itpr3tf/J mice. Life Sci 2019; 237:116930. [DOI: 10.1016/j.lfs.2019.116930] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 10/01/2019] [Accepted: 10/02/2019] [Indexed: 12/14/2022]
|
56
|
Graciarena M. Cytokines and Chemokines in Novel Roles: Exploring Their Potential as Predictors of Autism Spectrum Disorder. Biol Psychiatry 2019; 86:e11-e12. [PMID: 31370965 DOI: 10.1016/j.biopsych.2019.06.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 06/19/2019] [Indexed: 11/28/2022]
Affiliation(s)
- Mariana Graciarena
- Department of Physiology, Molecular and Cellular Biology, Faculty of Exact and Natural Sciences, University of Buenos Aires, Buenos Aires, Argentina; Institute of Physiology, Molecular Biology, and Neurosciences, National Council of Scientific Research, University of Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
57
|
Heuer LS, Croen LA, Jones KL, Yoshida CK, Hansen RL, Yolken R, Zerbo O, DeLorenze G, Kharrazi M, Ashwood P, Van de Water J. An Exploratory Examination of Neonatal Cytokines and Chemokines as Predictors of Autism Risk: The Early Markers for Autism Study. Biol Psychiatry 2019; 86:255-264. [PMID: 31279535 PMCID: PMC6677631 DOI: 10.1016/j.biopsych.2019.04.037] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 04/09/2019] [Accepted: 04/27/2019] [Indexed: 01/17/2023]
Abstract
BACKGROUND The identification of an early biomarker for autism spectrum disorder (ASD) would improve the determination of risk, leading to earlier diagnosis and, potentially, earlier intervention and improved outcomes. METHODS Data were generated from the Early Markers for Autism study, a population-based case-control study of prenatal and neonatal biomarkers of ASD. Newborn bloodspots of children with ASD (n = 370), children with developmental delay (n = 140), and general population (GP) controls (n = 378) were analyzed for 42 different immune markers using a Luminex multiplex platform. Comparisons of immune marker concentrations between groups were examined using logistic regression and partial least squares discriminant analysis. RESULTS Children with ASD had significantly increased neonatal levels of interleukin-6 (IL-6) and IL-8 compared with GP controls. An increase in IL-8 was especially significant in the ASD group with early onset compared with the GP group, with an adjusted odds ratio of 1.97 (95% confidence interval, 1.39-2.83; p = .00014). In addition, children with ASD had significantly elevated levels of eotaxin-1, interferon-γ, and IL-12p70 relative to children with developmental delay. We observed no significant differences in levels of immune markers between the developmental delay and GP groups. CONCLUSIONS Elevated levels of some inflammatory markers in newborn bloodspots indicated a higher degree of immune activation at birth in children who were subsequently diagnosed with ASD. The data from this exploratory study suggest that with further expansion, the development of neonatal bloodspot testing for cytokine/chemokine levels might lead to the identification of biomarkers that provide an accurate assessment of ASD risk at birth.
Collapse
Affiliation(s)
- Luke S Heuer
- Division of Rheumatology, Allergy, and Clinical Immunology, Department of Internal Medicine, University of California, Davis, Davis, California; MIND Institute, University of California, Davis, Davis, California
| | - Lisa A Croen
- Division of Research, Kaiser Permanente Northern California, Oakland, California
| | - Karen L Jones
- Division of Rheumatology, Allergy, and Clinical Immunology, Department of Internal Medicine, University of California, Davis, Davis, California; MIND Institute, University of California, Davis, Davis, California
| | - Cathleen K Yoshida
- Division of Research, Kaiser Permanente Northern California, Oakland, California
| | - Robin L Hansen
- MIND Institute, University of California, Davis, Davis, California; Department of Pediatrics, University of California, Davis, Davis, California
| | - Robert Yolken
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ousseny Zerbo
- Division of Research, Kaiser Permanente Northern California, Oakland, California
| | - Gerald DeLorenze
- Division of Research, Kaiser Permanente Northern California, Oakland, California
| | - Martin Kharrazi
- Environmental Health Investigations Branch, California Department of Public Health, Richmond, California
| | - Paul Ashwood
- MIND Institute, University of California, Davis, Davis, California; Department of Medical Microbiology and Immunology, University of California, Davis, Davis, California
| | - Judy Van de Water
- Division of Rheumatology, Allergy, and Clinical Immunology, Department of Internal Medicine, University of California, Davis, Davis, California; MIND Institute, University of California, Davis, Davis, California.
| |
Collapse
|
58
|
Alhowikan AM, AL-Ayadhi LY, Halepoto DM. Impact of environmental pollution, dietary factors and diabetes mellitus on Autism Spectrum Disorder (ASD). Pak J Med Sci 2019; 35:1179-1184. [PMID: 31372164 PMCID: PMC6659068 DOI: 10.12669/pjms.35.4.269] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 02/05/2019] [Accepted: 05/26/2019] [Indexed: 02/06/2023] Open
Abstract
Autism spectrum disorder (ASD) is complex neurodevelopmental condition described by impairments in three main behavioral areas: social deficits, impaired communication, and repetitive behaviors. Despite many years of vast study, the causes of ASD are still unknown. Various risk factors including genetic, infectious, metabolic and immunological have been investigated however, environmental, nutritional and diabetes related risk factors have not received sufficient attention. This study has provided an insight into the comprehensive interaction between environmental pollution, dietary factors and diabetes mellitus that could lead to the advancement of this debilitating neurodevelopment disorder. The literature search was done using PubMed and Google Scholar databases up to October 2018. Key words "Environmental Pollution", "Nutritional Factors", "Diabetes Mellitus", "Autism Spectrum Disorder" were selected.
Collapse
Affiliation(s)
- Abdulrahman Mohammed Alhowikan
- Abdulrahman Mohammed Alhowikan, PhD. Department of Physiology, Faculty of Medicine, King Saud University, P O Box 2925, Riyadh 11461 and Saudi Arabia
| | - Laila Yousef AL-Ayadhi
- Laila Yousef AL-Ayadhi, MBBS, PhD. Autism Research and Treatment Center, Department of physiology, Faculty of Medicine, King Saud University, P O Box 2925, Riyadh 11461 and Saudi Arabia
| | - Dost Muhammad Halepoto
- Dost Muhammad Halepoto, PhD. Autism Research and Treatment Center, Department of Physiology, Faculty of Medicine, King Saud University, P O Box 2925, Riyadh 11461 and Saudi Arabia
| |
Collapse
|
59
|
Rose D, Ashwood P. Rapid Communication: Plasma Interleukin-35 in Children with Autism. Brain Sci 2019; 9:E152. [PMID: 31252635 PMCID: PMC6680732 DOI: 10.3390/brainsci9070152] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 06/21/2019] [Accepted: 06/24/2019] [Indexed: 12/29/2022] Open
Abstract
In autism spectrum disorders (ASD) many individuals have co-morbid immune dysregulation that can lead to inflammation in the brain and periphery. The novel cytokine interleukin (IL)-35 has described anti-inflammatory properties; however, the plasma levels of IL-35 in children with ASD have never been investigated. The plasma levels of IL-35 were measured by an enzyme-linked immunosorbent assay in 30 children with ASD and 39 typically developing (TD) controls. In the current study, we found that plasma IL-35 levels were significantly decreased in children with ASD compared with TD children. Furthermore, lower IL-35 levels were associated with worse behaviors as assessed using the aberrant behavior checklist. These findings are in line with other observations of decreased regulatory cytokines such as transforming growth factor beta and IL-10 in ASD, and associations with severity of behaviors. In conclusion, regulating the expression of IL-35 may provide a new possible target for the treatment of immune issues in ASD to address an imbalance between pro- and anti-inflammatory signals that alter the behavioral phenotype.
Collapse
Affiliation(s)
- Destanie Rose
- Department of Medical Microbiology and Immunology, and The Medical Investigation of Neurodevelopmental Disorders Institute, University of California, Davis, CA 95817, USA
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, and The Medical Investigation of Neurodevelopmental Disorders Institute, University of California, Davis, CA 95817, USA.
| |
Collapse
|
60
|
Marchezan J. Editorial: Autism Spectrum Disorder and Autoimmune Diseases: A Pathway in Common? J Am Acad Child Adolesc Psychiatry 2019; 58:481-483. [PMID: 30902666 DOI: 10.1016/j.jaac.2019.03.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Accepted: 03/13/2019] [Indexed: 10/27/2022]
Abstract
The diagnosis of autism spectrum disorder (ASD) has increased significantly in the past decade. A rare condition at the time Kanner (1942) initially described it, ASD has become a public health issue with great social and financial burdens. In 2018, the Centers for Disease Control and Prevention estimated the prevalence of autism at 16.8 for every 1,000 (1:59) children by 8 years of age, affecting 26.6 of 1,000 boys and 6.6 of 1,000 girls. These numbers represent an increase of approximately 150% from 2000 to 2014.1.
Collapse
Affiliation(s)
- Josemar Marchezan
- School of Medicine, Universidade do Vale do Taquari (UNIVATES), Lajeado, Brazil; National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Porto Alegre, Brazil; Translational Research Group in Autism Spectrum Disorders GETTEA, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
| |
Collapse
|
61
|
Nadeem A, Ahmad SF, Al-Harbi NO, Attia SM, Bakheet SA, Ibrahim KE, Alqahtani F, Alqinyah M. Nrf2 activator, sulforaphane ameliorates autism-like symptoms through suppression of Th17 related signaling and rectification of oxidant-antioxidant imbalance in periphery and brain of BTBR T+tf/J mice. Behav Brain Res 2019; 364:213-224. [DOI: 10.1016/j.bbr.2019.02.031] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 02/17/2019] [Accepted: 02/17/2019] [Indexed: 12/11/2022]
|
62
|
Abraham JR, Barnard J, Wang H, Noritz GH, Yeganeh M, Buhas D, Natowicz MR. Proteomic investigations of human HERC2 mutants: Insights into the pathobiology of a neurodevelopmental disorder. Biochem Biophys Res Commun 2019; 512:421-427. [PMID: 30902390 DOI: 10.1016/j.bbrc.2019.02.149] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 02/27/2019] [Indexed: 01/11/2023]
Abstract
HERC2 is a giant protein with E3 ubiquitin ligase activity and other known and suspected functions. Mutations of HERC2 are implicated in the pathogenesis of various cancers and result in severe neurological conditions in Herc2-mutant mice. Recently, a pleotropic autosomal recessive HERC2-associated syndrome of intellectual disability, autism and variable neurological deficits was described; its pathogenetic basis is largely unknown. Using peripheral blood-derived lymphoblasts from 3 persons with homozygous HERC2 variants and 14 age- and gender-matched controls, we performed label-free unbiased HPLC-tandem mass spectrometry-based proteomic analyses to provide insights into HERC2-mediated pathobiology. We found that out of 3427 detected proteins, there were 812 differentially expressed proteins between HERC2-cases vs. controls. 184 canonical pathways were enriched after FDR adjustment, including mitochondrial function, energy metabolism, EIF2 signaling, immune functions, ubiquitination and DNA repair. Ingenuity Pathway Analysis® identified 209 upstream regulators that could drive the differential expression, prominent amongst which were neurodegeneration-associated proteins. Differentially expressed protein interaction networks highlighted themes of immune function/dysfunction, regulation of cell cycle/cell death, and energy metabolism. Overall, the analysis of the HERC2-associated proteome revealed striking differential protein expression between cases and controls. The large number of differentially expressed proteins likely reflects HERC2's multiple domains and numerous interacting proteins. Our canonical pathway and protein interaction network findings suggest derangements of multiple pathways in HERC2-associated disease.
Collapse
Affiliation(s)
- Joseph R Abraham
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - John Barnard
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Heng Wang
- DDC Clinic, Center for Special Needs Children, Middlefield, OH, USA
| | - Garey H Noritz
- Complex Health Care Program, Nationwide Children's Hospital, Columbus, OH, USA
| | - Mehdi Yeganeh
- Department of Medical Genetics, McGill University Health Centre, Montreal, Canada
| | - Daniela Buhas
- Department of Medical Genetics, McGill University Health Centre, Montreal, Canada
| | - Marvin R Natowicz
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA; Pathology and Laboratory Medicine, Genomic Medicine, Neurological and Pediatrics Institutes, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
63
|
Plaza-Díaz J, Gómez-Fernández A, Chueca N, Torre-Aguilar MJDL, Gil Á, Perez-Navero JL, Flores-Rojas K, Martín-Borreguero P, Solis-Urra P, Ruiz-Ojeda FJ, Garcia F, Gil-Campos M. Autism Spectrum Disorder (ASD) with and without Mental Regression is Associated with Changes in the Fecal Microbiota. Nutrients 2019; 11:337. [PMID: 30764497 PMCID: PMC6412819 DOI: 10.3390/nu11020337] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 01/22/2019] [Accepted: 02/01/2019] [Indexed: 02/06/2023] Open
Abstract
New microbiome sequencing technologies provide novel information about the potential interactions among intestinal microorganisms and the host in some neuropathologies as autism spectrum disorders (ASD). The microbiota⁻gut⁻brain axis is an emerging aspect in the generation of autistic behaviors; evidence from animal models suggests that intestinal microbial shifts may produce changes fitting the clinical picture of autism. The aim of the present study was to evaluate the fecal metagenomic profiles in children with ASD and compare them with healthy participants. This comparison allows us to ascertain how mental regression (an important variable in ASD) could influence the intestinal microbiota profile. For this reason, a subclassification in children with ASD by mental regression (AMR) and no mental regression (ANMR) phenotype was performed. The present report was a descriptive observational study. Forty-eight children aged 2⁻6 years with ASD were included: 30 with ANMR and 18 with AMR. In addition, a control group of 57 normally developing children was selected and matched to the ASD group by sex and age. Fecal samples were analyzed with a metagenomic approach using a next-generation sequencing platform. Several differences between children with ASD, compared with the healthy group, were detected. Namely, Actinobacteria and Proteobacteria at phylum level, as well as, Actinobacteria, Bacilli, Erysipelotrichi, and Gammaproteobacteria at class level were found at higher proportions in children with ASD. Additionally, Proteobacteria levels showed to be augmented exclusively in AMR children. Preliminary results, using a principal component analysis, showed differential patterns in children with ASD, ANMR and AMR, compared to healthy group, both for intestinal microbiota and food patterns. In this study, we report, higher levels of Actinobacteria, Proteobacteria and Bacilli, aside from Erysipelotrichi, and Gammaproteobacteria in children with ASD compared to healthy group. Furthermore, AMR children exhibited higher levels of Proteobacteria. Further analysis using these preliminary results and mixing metagenomic and other "omic" technologies are needed in larger cohorts of children with ASD to confirm these intestinal microbiota changes.
Collapse
Affiliation(s)
- Julio Plaza-Díaz
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain;.
- Institute of Nutrition and Food Technology "José Mataix", Center of Biomedical Research,University of Granada, 18016 Armilla, Granada, Spain.
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada,18014 Granada, Spain.
| | - Antonio Gómez-Fernández
- Pediatric Research and Metabolism Unit, Reina Sofia University Hospital, Maimónides Institute forBiomedical Research of Córdoba (IMIBIC), University of Córdoba, 14010 Córdoba, Spain.
| | - Natalia Chueca
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada,18014 Granada, Spain.
| | - María José de la Torre-Aguilar
- Pediatric Research and Metabolism Unit, Reina Sofia University Hospital, Maimónides Institute forBiomedical Research of Córdoba (IMIBIC), University of Córdoba, 14010 Córdoba, Spain.
| | - Ángel Gil
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain;.
- Institute of Nutrition and Food Technology "José Mataix", Center of Biomedical Research,University of Granada, 18016 Armilla, Granada, Spain.
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada,18014 Granada, Spain.
- CIBEROBN (CIBER Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III,28029 Madrid, Spain.
| | - Juan Luis Perez-Navero
- Pediatric Research and Metabolism Unit, Reina Sofia University Hospital, Maimónides Institute forBiomedical Research of Córdoba (IMIBIC), University of Córdoba, 14010 Córdoba, Spain.
| | - Katherine Flores-Rojas
- Pediatric Research and Metabolism Unit, Reina Sofia University Hospital, Maimónides Institute forBiomedical Research of Córdoba (IMIBIC), University of Córdoba, 14010 Córdoba, Spain.
- CIBEROBN (CIBER Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III,28029 Madrid, Spain.
| | - Pilar Martín-Borreguero
- Department of Child and Adolescent Clinical Psychiatry and Psychology, Reina Sofia University Hospital,Maimónides Institute for Biomedical Research of Córdoba (IMIBIC), 14010 Cordoba, Spain.
| | - Patricio Solis-Urra
- PROFITH "PROmoting FITness and Health through physical activity" research group, Department ofPhysical Education and Sport, Faculty of Sport Sciences, University of Granada, 18071 Granada, Spain.
- IRyS Research Group, School of Physical Education, Pontificia Universidad Católica de Valparaíso,Valparaiso 2374631, Chile.
| | - Francisco Javier Ruiz-Ojeda
- RG Adipocytes and metabolism, Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Center Munich, 85748 Garching, Munich, Germany.
| | - Federico Garcia
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada,18014 Granada, Spain.
| | - Mercedes Gil-Campos
- Pediatric Research and Metabolism Unit, Reina Sofia University Hospital, Maimónides Institute forBiomedical Research of Córdoba (IMIBIC), University of Córdoba, 14010 Córdoba, Spain.
- CIBEROBN (CIBER Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III,28029 Madrid, Spain.
| |
Collapse
|
64
|
Ahmad SF, Ansari MA, Nadeem A, Bakheet SA, Alotaibi MR, Alasmari AF, Alshammari MA, Al-Mazroua HA, Attia SM. DAPTA, a C-C chemokine receptor 5 (CCR5) antagonist attenuates immune aberrations by downregulating Th9/Th17 immune responses in BTBR T + Itpr3tf/J mice. Eur J Pharmacol 2019; 846:100-108. [PMID: 30658114 DOI: 10.1016/j.ejphar.2019.01.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 01/14/2019] [Accepted: 01/14/2019] [Indexed: 01/06/2023]
Abstract
Autism spectrum disorder (ASD) is a prevalent neurodevelopmental disorder characterized by deficits in social interaction, communication, and repetitive behaviors. BTBR T+ Itpr3tf/J (BTBR) mice, a preclinical autistic model featuring ASD symptoms as defined by social relations, was used in this study. We evaluated the potentially protective effect of D-Ala-peptide T-amide (DAPTA), a selective C-C chemokine receptor 5 (CCR5) antagonist, in BTBR mice. CCR5 is considered a potential therapeutic target in different neurodegenerative disorders. BTBR and C57 mice were intraperitoneally (i.p) treated with the DAPTA (0.01 mg/kg, i.p, once daily) for 7 days. We examined the effect of DAPTA by evaluating marble burying and administering repetitive behavior tests. We employed flow cytometry to assess the effect of DAPTA on CCR5+, CD4+CCR5+, CCR5+IL-6+, CCR5+IL-9+, CCR5+IL-17A+, CCR5+RORγT+, CCR5+IL-10+, and CCR5+Foxp3+ in spleen cells. We further explored the effects of DAPTA on IL-6, IL-9, IL-17A, RORγT, IL-10, and Foxp3 protein and mRNA expression levels in the brain tissues. DAPTA administration significantly decreased marble burying and repetitive behavior in BTBR mice. Additionally, DAPTA treatment inhibited CCR5+, CD4+CCR5+, CCR5+IL-6+, CCR5+IL-9+, CCR5+IL-17A+, CCR5+RORγT+, and upregulated CCR5+IL-10+, and CCR5+Foxp3+ production. We further observed that DAPTA downregulated IL-6, IL-9, IL-17A, and RORγT, and increased IL-10 and Foxp3 protein and mRNA expression. Therefore, our results suggest that DAPTA administration represents a potential treatment strategy for patients with ASD.
Collapse
Affiliation(s)
- Sheikh F Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.
| | - Mushtaq A Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Saleh A Bakheet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Moureq R Alotaibi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdullah F Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Musaad A Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Haneen A Al-Mazroua
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia; Department of Pharmacology and Toxicology, College of Pharmacy, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
65
|
Carpenter KLH, Major S, Tallman C, Chen LW, Franz L, Sun J, Kurtzberg J, Song A, Dawson G. White Matter Tract Changes Associated with Clinical Improvement in an Open-Label Trial Assessing Autologous Umbilical Cord Blood for Treatment of Young Children with Autism. Stem Cells Transl Med 2019; 8:138-147. [PMID: 30620122 PMCID: PMC6344899 DOI: 10.1002/sctm.18-0251] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 11/19/2018] [Indexed: 12/26/2022] Open
Abstract
Autism spectrum disorder (ASD) is a heterogeneous neurodevelopmental disorder characterized by social communication deficits and the presence of restricted interests and repetitive behaviors. We have previously reported significant improvements in behavior, including increased social functioning, improved communication abilities, and decreased clinical symptoms in children with ASD, following treatment with a single infusion of autologous cord blood in a phase I open‐label trial. In the current study, we aimed to understand whether these improvements were associated with concurrent changes in brain structural connectivity. Twenty‐five 2‐ to 6‐year‐old children with ASD participated in this trial. Clinical outcome measures included the Vineland Adaptive Behavior Scales‐II Socialization Subscale, Expressive One‐Word Picture Vocabulary Test‐4, and the Clinical Global Impression‐Improvement Scale. Structural connectivity was measured at baseline and at 6 months in a subset of 19 children with 25‐direction diffusion tensor imaging and deterministic tractography. Behavioral improvements were associated with increased white matter connectivity in frontal, temporal, and subcortical regions (hippocampus and basal ganglia) that have been previously shown to show anatomical, connectivity, and functional abnormalities in ASD. The current results suggest that improvements in social communication skills and a reduction in symptoms in children with ASD following treatment with autologous cord blood infusion were associated with increased structural connectivity in brain networks supporting social, communication, and language abilities. stem cells translational medicine2019;8:138&10
Collapse
Affiliation(s)
- Kimberly L H Carpenter
- Duke Center for Autism and Brain Development, Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, North Carolina, USA
| | - Samantha Major
- Duke Center for Autism and Brain Development, Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, North Carolina, USA
| | - Catherine Tallman
- Brain Imaging and Analysis Center, Duke University Medical Center, Duke University School of Medicine, Durham, North Carolina, USA
| | - Lyon W Chen
- Brain Imaging and Analysis Center, Duke University Medical Center, Duke University School of Medicine, Durham, North Carolina, USA
| | - Lauren Franz
- Duke Center for Autism and Brain Development, Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, North Carolina, USA.,Duke Global Health Institute, Duke University School of Medicine, Duke University, Durham, North Carolina, USA
| | - Jessica Sun
- Marcus Center for Cellular Cures, Duke University Medical Center, Duke University School of Medicine, Durham, North Carolina, USA
| | - Joanne Kurtzberg
- Marcus Center for Cellular Cures, Duke University Medical Center, Duke University School of Medicine, Durham, North Carolina, USA
| | - Allen Song
- Brain Imaging and Analysis Center, Duke University Medical Center, Duke University School of Medicine, Durham, North Carolina, USA
| | - Geraldine Dawson
- Duke Center for Autism and Brain Development, Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, North Carolina, USA.,Marcus Center for Cellular Cures, Duke University Medical Center, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
66
|
Proteomic Investigations of Autism Spectrum Disorder: Past Findings, Current Challenges, and Future Prospects. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1118:235-252. [DOI: 10.1007/978-3-030-05542-4_12] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
67
|
Bryn V, Verkerk R, Skjeldal OH, Saugstad OD, Ormstad H. Kynurenine Pathway in Autism Spectrum Disorders in Children. Neuropsychobiology 2018; 76:82-88. [PMID: 29694960 DOI: 10.1159/000488157] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 03/04/2018] [Indexed: 12/27/2022]
Abstract
BACKGROUND There is increasing evidence that altered immune responses play a role in the pathogenesis of autism spectrum disorders (ASD), together with dysfunction of the serotonergic and glutamatergic systems. Since the kynurenine (KYN) pathway that degrades tryptophan (TRP) is activated in various neuroinflammatory states, we aimed to determine whether this pathway is activated in ASD. METHODS Sixty-five pediatric ASD patients (including 52 boys) were enrolled from an epidemiological survey covering 2 counties in Norway; 30 (46.5%) of these patients were diagnosed with childhood autism, 16 (24.6%) with Asperger syndrome, 12 (18.5%) with atypical autism, 1 (1.5%) with Rett syndrome, and 6 (9.2%) with other ASD. The serum levels of the following markers were measured in the children with ASD and compared to those in 30 healthy children: TRP, KYN, kynurenic acid (KA), 3-hydroxykynurenine, and quinolinic acid. RESULTS The mean serum level of KA was significantly lower in the ASD group than in the healthy controls (28.97 vs. 34.44 nM, p = 0.040), while the KYN/KA ratio was significantly higher in the ASD group (61.12 vs. 50.39, p = 0.006). The same relative values were found when comparing the childhood autism subgroup with the controls. Also, the mean serum level of TRP was significantly lower in children with a subdiagnosis of childhood autism than in those with Asperger syndrome (67.26 vs. 77.79 μM, p = 0.020). CONCLUSION Our study indicates that there is an increased neurotoxic potential and also a possible lower KYN aminotransferase activity in ASD.
Collapse
Affiliation(s)
- Vesna Bryn
- Department of Pediatrics, Innlandet Hospital Trust, Lillehammer, Norway
| | - Robert Verkerk
- Laboratory of Medical Biochemistry, University of Antwerp, Antwerp, Belgium
| | - Ola H Skjeldal
- Gillberg Neuropsychiatry Centre, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ola Didrik Saugstad
- Pediatric Research Institute, Rikshospitalet Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Heidi Ormstad
- Faculty of Health Sciences, Buskerud University College, Drammen, Norway
| |
Collapse
|
68
|
Hughes HK, Rose D, Ashwood P. The Gut Microbiota and Dysbiosis in Autism Spectrum Disorders. Curr Neurol Neurosci Rep 2018; 18:81. [PMID: 30251184 DOI: 10.1007/s11910-018-0887-6] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW There is a growing body of evidence indicating the gut microbiota influence neurodevelopment and behavior. The purposes of this review are to provide an overview of studies analyzing the microbiota and their metabolites in autism spectrum disorders (ASD) and to discuss the possible mechanisms of action involved in microbial influence on the brain and behavior. RECENT FINDINGS The microbiota-gut-brain (MGB) axis has been extensively studied in animal models, and it is clear that alterations in the composition of microbiota alter neurological and behavioral outcomes. However, findings in human studies are less abundant. Although there are several studies so far showing altered microbiota (dysbiosis) in ASD, the results are heterogeneous and often contradictory. Intervention studies such as fecal microbiota transplant therapies show promise and lend credence to the involvement of the microbiota in ASD. A role for the microbiota in ASD is likely; however, further studies elucidating microbial or metabolomic signatures and mechanisms of action are needed. Future research should focus on intervention studies that can identify specific metabolites and immune mediators that improve with treatment to help identify etiologies and pathological mechanisms of ASD.
Collapse
Affiliation(s)
- Heather K Hughes
- Department of Medical Microbiology and Immunology, UC Davis, 2805, 50th Street, Sacramento, CA, 95817, USA.,The M.I.N.D. Institute, University of California at Davis, Davis, CA, USA
| | - Destanie Rose
- Department of Medical Microbiology and Immunology, UC Davis, 2805, 50th Street, Sacramento, CA, 95817, USA.,The M.I.N.D. Institute, University of California at Davis, Davis, CA, USA
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, UC Davis, 2805, 50th Street, Sacramento, CA, 95817, USA. .,The M.I.N.D. Institute, University of California at Davis, Davis, CA, USA.
| |
Collapse
|
69
|
Traglia M, Croen LA, Jones KL, Heuer LS, Yolken R, Kharrazi M, DeLorenze GN, Ashwood P, Van de Water J, Weiss LA. Cross-genetic determination of maternal and neonatal immune mediators during pregnancy. Genome Med 2018; 10:67. [PMID: 30134952 PMCID: PMC6106874 DOI: 10.1186/s13073-018-0576-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 08/03/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The immune system plays a fundamental role in development during pregnancy and early life. Alterations in circulating maternal and neonatal immune mediators have been associated with pregnancy complications as well as susceptibility to autoimmune and neurodevelopmental conditions in later life. Evidence suggests that the immune system in adults not only responds to environmental stimulation but is also under strong genetic control. METHODS This is the first genetic study of > 700 mother-infant pairs to analyse the circulating levels of 22 maternal mid-gestational serum-derived and 42 neonatal bloodspot-derived immune mediators (cytokines/chemokines) in the context of maternal and fetal genotype. We first estimated the maternal and fetal genome-wide SNP-based heritability (h2g) for each immune molecule and then performed genome-wide association studies (GWAS) to identify specific loci contributing to individual immune mediators. Finally, we assessed the relationship between genetic immune determinants and ASD outcome. RESULTS We show maternal and neonatal cytokines/chemokines displaying genetic regulation using independent methodologies. We demonstrate that novel fetal loci for immune function independently affect the physiological levels of maternal immune mediators and vice versa. The cross-associated loci are in distinct genomic regions compared with individual-specific immune mediator loci. Finally, we observed an interaction between increased IL-8 levels at birth, autism spectrum disorder (ASD) status, and a specific maternal genotype. CONCLUSIONS Our results suggest that maternal and fetal genetic variation influences the immune system during pregnancy and at birth via distinct mechanisms and that a better understanding of immune factor determinants in early development may shed light on risk factors for developmental disorders.
Collapse
Affiliation(s)
- Michela Traglia
- Department of Psychiatry and Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | - Lisa A Croen
- Divison of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Karen L Jones
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, University of California Davis, Davis, CA, USA
- MIND Institute, University of California Davis, Davis, CA, USA
| | - Luke S Heuer
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, University of California Davis, Davis, CA, USA
- MIND Institute, University of California Davis, Davis, CA, USA
| | - Robert Yolken
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Martin Kharrazi
- Division of Environmental and Occupational Disease Control, California Department of Public Health, Richmond, CA, USA
| | - Gerald N DeLorenze
- Divison of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Paul Ashwood
- MIND Institute, University of California Davis, Davis, CA, USA
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
| | - Judy Van de Water
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, University of California Davis, Davis, CA, USA
- MIND Institute, University of California Davis, Davis, CA, USA
| | - Lauren A Weiss
- Department of Psychiatry and Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
70
|
Sanctuary MR, Kain JN, Angkustsiri K, German JB. Dietary Considerations in Autism Spectrum Disorders: The Potential Role of Protein Digestion and Microbial Putrefaction in the Gut-Brain Axis. Front Nutr 2018; 5:40. [PMID: 29868601 PMCID: PMC5968124 DOI: 10.3389/fnut.2018.00040] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 04/30/2018] [Indexed: 12/13/2022] Open
Abstract
Children with autism spectrum disorders (ASD), characterized by a range of behavioral abnormalities and social deficits, display high incidence of gastrointestinal (GI) co-morbidities including chronic constipation and diarrhea. Research is now increasingly able to characterize the “fragile gut” in these children and understand the role that impairment of specific GI functions plays in the GI symptoms associated with ASD. This mechanistic understanding is extending to the interactions between diet and ASD, including food structure and protein digestive capacity in exacerbating autistic symptoms. Children with ASD and gut co-morbidities exhibit low digestive enzyme activity, impaired gut barrier integrity and the presence of antibodies specific for dietary proteins in the peripheral circulation. These findings support the hypothesis that entry of dietary peptides from the gut lumen into the vasculature are associated with an aberrant immune response. Furthermore, a subset of children with ASD exhibit high concentrations of metabolites originating from microbial activity on proteinaceous substrates. Taken together, the combination of specific protein intakes poor digestion, gut barrier integrity, microbiota composition and function all on a background of ASD represents a phenotypic pattern. A potential consequence of this pattern of conditions is that the fragile gut of some children with ASD is at risk for GI symptoms that may be amenable to improvement with specific dietary changes. There is growing evidence that shows an association between gut dysfunction and dysbiosis and ASD symptoms. It is therefore urgent to perform more experimental and clinical research on the “fragile gut” in children with ASD in order to move toward advancements in clinical practice. Identifying those factors that are of clinical value will provide an evidence-based path to individual management and targeted solutions; from real time sensing to the design of diets with personalized protein source/processing, all to improve GI function in children with ASD.
Collapse
Affiliation(s)
- Megan R Sanctuary
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Jennifer N Kain
- Department of Neurobiology, Physiology and Behavior Department, University of California, Davis, Davis, CA, United States
| | - Kathleen Angkustsiri
- School of Medicine, Department of Pediatrics, University of California, Davis, Sacramento, CA, United States.,Department of Pediatrics, UC Davis MIND Institute, Sacramento, CA, United States
| | - J Bruce German
- Department of Food Science and Technology, University of California, Davis, Davis, CA, United States.,Foods for Health Institute, University of California, Davis, Davis, CA, United States
| |
Collapse
|
71
|
Gómez-Giménez B, Llansola M, Cabrera-Pastor A, Hernández-Rabaza V, Agustí A, Felipo V. Endosulfan and Cypermethrin Pesticide Mixture Induces Synergistic or Antagonistic Effects on Developmental Exposed Rats Depending on the Analyzed Behavioral or Neurochemical End Points. ACS Chem Neurosci 2018; 9:369-380. [PMID: 29094921 DOI: 10.1021/acschemneuro.7b00364] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Exposure to pesticides has been associated with neurodevelopmental toxicity. Usually people are exposed to mixtures of pesticides. However, most studies analyze the effects of individual pesticides. Developmental exposure to mixtures of pesticides may result in additive effects or in antagonistic or synergistic effects. The aim of this work was to compare the effects of developmental exposure of rats to cypermethrin or endosulfan with the effects of its mixture on cognitive and motor function and on some underlying mechanisms. Exposure to individual pesticides or the mixture was from gestational day 7 to postnatal day 21. We analyzed the effects, in males and females, on spatial learning and memory, associative learning, anxiety, motor coordination, and spontaneous motor activity. We also analyzed neuroinflammation and NMDA receptor subunits in hippocampus and extracellular GABA in cerebellum. Exposure to the mixture, but not to individual pesticides, impaired spatial memory in males, associative learning in females, and increased motor activity in males and females. This indicates a synergistic effect of cypermethrin and endolsufan exposure on these end points. In contrast, motor coordination was impaired by individual exposure to endosulfan or cypermethrin, associated with increased extracellular GABA in cerebellum, but these effects were prevented in rats exposed to the mixture, indicating an antagonistic effect of cypermethrin and endolsufan exposure on these end points. The results show different interaction modes (synergism or antagonism) of the pesticides, depending on the end point analyzed and the sex of the rats.
Collapse
Affiliation(s)
- Belén Gómez-Giménez
- Laboratorio
de Neurobiología, Centro Investigación Príncipe Felipe, Valencia 46012, Spain
| | - Marta Llansola
- Laboratorio
de Neurobiología, Centro Investigación Príncipe Felipe, Valencia 46012, Spain
| | - Andrea Cabrera-Pastor
- Laboratorio
de Neurobiología, Centro Investigación Príncipe Felipe, Valencia 46012, Spain
| | - Vicente Hernández-Rabaza
- Laboratorio
de Neurobiología, Centro Investigación Príncipe Felipe, Valencia 46012, Spain
- Department
of Biomedical Sciences, Cardenal Herrera University-CEU, CEU Universities, Avda del Pozo s/n, Alfara del Patriarca 46115, Spain
| | - Ana Agustí
- Laboratorio
de Neurobiología, Centro Investigación Príncipe Felipe, Valencia 46012, Spain
| | - Vicente Felipo
- Laboratorio
de Neurobiología, Centro Investigación Príncipe Felipe, Valencia 46012, Spain
| |
Collapse
|
72
|
Gomez-Fernandez A, de la Torre-Aguilar MJ, Gil-Campos M, Flores-Rojas K, Cruz-Rico MD, Martin-Borreguero P, Perez-Navero JL. Children With Autism Spectrum Disorder With Regression Exhibit a Different Profile in Plasma Cytokines and Adhesion Molecules Compared to Children Without Such Regression. Front Pediatr 2018; 6:264. [PMID: 30320048 PMCID: PMC6169449 DOI: 10.3389/fped.2018.00264] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 09/05/2018] [Indexed: 12/27/2022] Open
Abstract
Background: In the etiopathogenesis of autism spectrum disorder (ASD), it has been suggested that a proinflammatory condition, as well as an alteration in adhesion molecules in the early stages of neurodevelopment, may play a role in the pathophysiology of the disorder. This study set out to evaluate the plasma levels of certain inflammatory cytokines, adhesion molecules, and growth factors in a sample of pediatric patients with ASD and compare them to the levels in a control group of healthy children. Methods: Fifty-four children (45 males and nine females) aged 2-6, who were diagnosed with ASD, and a control group of 54 typically-developing children of similar ages were selected. The diagnosis of ASD was carried out in accordance with the DSM-5 criteria and the data obtained from a developmental semi-structured clinical interview and the ADOS evaluation test. Additional testing was carried out to identify the children's developmental level and severity of ASD symptomatology. Patients with ASD were further divided into two subgroups based on developmental parameters: ASD children with neurodevelopmental regression (AMR) and ASD children without neurodevelopmental regression (ANMR). Analyses of plasma molecules, such as cathepsin, IL1β, IL6, IL8, MPO, RANTES, MCP, BDNF, PAI NCAM, sICAM, sVCAM and NGF, were performed. Results: Higher levels of NGF were observed in the ASD group compared with the levels in the control group (p < 0.05). However, in the analysis of the ASD subgroups, lower plasma levels of NCAM and higher levels of NGF were found in the group of ASD children without developmental regression compared to the levels in the group of typically-developing children. Conclusions: These results suggest differences that could be related to different pathophysiological mechanisms in ASD. There is not a specific profile for the expression of relevant plasma cytokines, adhesion molecules or growth factors in children with ASD compared with that in typically-developing children. However, in the ANMR and AMR subgroups, some of the adhesion molecules and neuronal growth factors show differences that may be related to synaptogenesis.
Collapse
Affiliation(s)
- Antonio Gomez-Fernandez
- Department of Pediatrics, Reina Sofia University Hospital, University of Córdoba, Maimónides Institute for Biomedical Research of Córdoba (IMIBIC), Cordoba, Spain
| | - Maria J de la Torre-Aguilar
- Department of Pediatrics, Reina Sofia University Hospital, University of Córdoba, Maimónides Institute for Biomedical Research of Córdoba (IMIBIC), Cordoba, Spain
| | - Mercedes Gil-Campos
- Pediatric Metabolism Unit, Reina Sofia University Hospital, Maimónides Institute for Biomedical Research of Córdoba (IMIBIC), University of Córdoba, Centro de Investigación Biomédica en Red-Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Cordoba, Spain
| | - Katherine Flores-Rojas
- Pediatric Metabolism Unit, Reina Sofia University Hospital, Maimónides Institute for Biomedical Research of Córdoba (IMIBIC), University of Córdoba, Centro de Investigación Biomédica en Red-Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Cordoba, Spain
| | - Maria D Cruz-Rico
- Department of Biochemistry and Molecular Biology II, Institute of Nutrition and Food Technology, Center of Biomedical Research, University of Granada, Granada, Spain
| | - Pilar Martin-Borreguero
- Department of Child and Adolescent Clinical Psychiatry and Psychology, Reina Sofia University Hospital, Maimónides Institute for Biomedical Research of Córdoba (IMIBIC), Cordoba, Spain
| | - Juan Luis Perez-Navero
- Department of Pediatrics, Reina Sofia University Hospital, University of Córdoba, Maimónides Institute for Biomedical Research of Córdoba (IMIBIC), Cordoba, Spain
| |
Collapse
|
73
|
Ashwood P. Differential T Cell Levels of Tumor Necrosis Factor Receptor-II in Children With Autism. Front Psychiatry 2018; 9:543. [PMID: 30524316 PMCID: PMC6256095 DOI: 10.3389/fpsyt.2018.00543] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 10/11/2018] [Indexed: 11/13/2022] Open
Abstract
Autism spectrum disorders (ASD) are characterized by impairments in verbal and non-verbal communication, in social interactions, and often accompanied by stereotypical interests and behaviors. A role for immune dysfunction has long been implicated in ASD pathophysiology, behavioral severity, and co-morbidities. The pro-inflammatory cytokine tumor necrosis factor alpha (TNFα) has been associated with ASD in some studies but little is known about its receptors. There are two receptors for TNFα, with TNFRI relaying many of the signals from TNFα, especially those that are rapid, whilst TNFRII relays later more long-term effects of TNFα. Proteolytic cleavage can lead to the soluble versions of these receptors which can neutralize the effects of TNFα. Here, we determined levels of TNFα and its receptors in 36 children with a confirmed diagnosis of ASD and 27 confirmed typically developing (TD) controls, 2-5 years-of-age. Children with ASD had higher levels of TNFRII on T cells compared to controls following cell stimulation. Levels of sTNFRII were decreased in cell supernatants following stimulation in ASD. Overall these data corroborate the role of inflammatory events in ASD and align with previous studies that have shown differential changes in cellular adaptive immunity in children with ASD. Future longitudinal analyzes of cellular immune function and downstream signaling from immune receptors will help further delineate the role of inflammation in ASD.
Collapse
Affiliation(s)
- Paul Ashwood
- Department of Medical Microbiology and Immunology, and The Medical Investigation of Neurodevelopmental Disorders Institute, University of California, Davis, CA, United States
| |
Collapse
|
74
|
Hughes HK, Ashwood P. Anti- Candida albicans IgG Antibodies in Children With Autism Spectrum Disorders. Front Psychiatry 2018; 9:627. [PMID: 30534090 PMCID: PMC6275220 DOI: 10.3389/fpsyt.2018.00627] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 11/05/2018] [Indexed: 12/20/2022] Open
Abstract
The gut microbiota are known to have a profound influence on both mucosal and systemic immunity and are important for gastrointestinal (GI) function. In addition, new evidence shows that the microbiota significantly influence neurodevelopment and behavior. Immune dysfunction and GI distress are extremely common in individuals with autism spectrum disorders (ASD). A growing body of evidence suggests that individuals with ASD have significant aberrations in the composition of their gut microbiota, known as dysbiosis. However, these studies have focused on the bacterial components of the microbiota, leaving the fungal microbiota in ASD poorly studied. Increases in fungal species such as Candida albicans are associated with inflammatory bowel disorders, and have recently been implicated in several neurological disorders including schizophrenia. We aimed to determine if children with ASD exhibit elevations in antibodies that target C. albicans, indicating current or previous overgrowth of this fungal species. We measured anti-C. albicans immunoglobulin (IgG) in plasma from 80 children enrolled in the UC Davis MIND Institute CHARGE study. Measurements were acquired using a commercial ELISA kit. Plasma anti-C. albicans antibody positivity was found in 36.5% (19/52) of children with ASD. Anti-C. albicans antibodies in typically developing controls was (14.3%; 4/28). Overall, ASD children had a higher rate of high-positive values compared to typically developed children with an unadjusted odds ratio of 3.45 (95% confidence interval, 1.0409 to 11.4650; p = 0.041, two-tailed). GI dysfunction was found in about half of the ASD children who were positive for anti-Candida IgG. This study provides evidence of a new microbial risk factor for ASD.
Collapse
Affiliation(s)
- Heather K Hughes
- Department of Medical Microbiology and Immunology, and The Medical Investigation of Neurodevelopmental Disorders (M.I.N.D.) Institute, University of California, Davis, Davis, CA, United States
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, and The Medical Investigation of Neurodevelopmental Disorders (M.I.N.D.) Institute, University of California, Davis, Davis, CA, United States
| |
Collapse
|
75
|
Masi A, Breen EJ, Alvares GA, Glozier N, Hickie IB, Hunt A, Hui J, Beilby J, Ravine D, Wray J, Whitehouse AJO, Guastella AJ. Cytokine levels and associations with symptom severity in male and female children with autism spectrum disorder. Mol Autism 2017; 8:63. [PMID: 29214007 PMCID: PMC5712192 DOI: 10.1186/s13229-017-0176-2] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 10/31/2017] [Indexed: 01/22/2023] Open
Abstract
Background Autism spectrum disorders (ASDs) are complex, pervasive, and heterogeneous neurodevelopmental conditions with varying trajectories, significant male bias and largely unknown etiology. However, an understanding of the biological mechanisms driving pathophysiology is evolving. Immune system aberrations, as identified through cytokine profiles, are believed to have a role in ASD. Altered cytokine levels may facilitate identification of ASD subtypes as well as provide biological markers of response to effective treatments. Research exploring the relationship between cytokine profiles and ASD symptoms is, however, in its infancy. The objective of this study was to explore relationships between cytokine levels and the severity of ASD and other clinical traits. Methods Multiplex assay techniques were used to measure levels of 27 cytokines in plasma samples from a cohort of 144 children diagnosed with ASD. Results Overall, results showed a significant negative association between platelet-derived growth factor (PDGF)-BB, and the severity of ASD symptoms. Furthermore, a significant interaction with sex suggested a different immune profile for females compared to males. ASD symptom severity was negatively associated with levels of 4 cytokines, IL-1β, IL-8, MIP-1β, and VEGF, in females, but not in males. Conclusions Results of the present study suggest that an altered cytokine response or profile is associated with the severity of ASD-related symptoms, with sex a potential modifier of this relationship. Further research in larger populations which recognizes the importance of sex comparisons and longitudinal assessments are now required to extend and further describe the role of the immune system in ASD. Electronic supplementary material The online version of this article (10.1186/s13229-017-0176-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anne Masi
- Autism Clinic for Translational Research, Brain and Mind Centre, Central Clinical School, Sydney Medical School, University of Sydney, 100 Mallett Street, Camperdown, New South Wales 2050 Australia
| | - Edmond J Breen
- Australian Proteome Analysis Facility, Macquarie University, Sydney, Australia
| | - Gail A Alvares
- Telethon Kids Institute, University of Western Australia, Perth, Western Australia Australia.,Cooperative Research Centre for Living with Autism (Autism CRC), Long Pocket, Brisbane, Queensland Australia
| | - Nicholas Glozier
- Autism Clinic for Translational Research, Brain and Mind Centre, Central Clinical School, Sydney Medical School, University of Sydney, 100 Mallett Street, Camperdown, New South Wales 2050 Australia
| | - Ian B Hickie
- Autism Clinic for Translational Research, Brain and Mind Centre, Central Clinical School, Sydney Medical School, University of Sydney, 100 Mallett Street, Camperdown, New South Wales 2050 Australia
| | - Anna Hunt
- Telethon Kids Institute, University of Western Australia, Perth, Western Australia Australia
| | - Jennie Hui
- Pathwest Laboratory Medicine WA, Nedlands, Western Australia Australia.,School of Pathology and Laboratory Medicine, University of Western Australia, Perth, Western Australia Australia.,School of Population Health, University of Western Australia, Perth, Western Australia Australia
| | - John Beilby
- Pathwest Laboratory Medicine WA, Nedlands, Western Australia Australia.,School of Pathology and Laboratory Medicine, University of Western Australia, Perth, Western Australia Australia
| | - David Ravine
- Pathwest Laboratory Medicine WA, Nedlands, Western Australia Australia.,School of Pathology and Laboratory Medicine, University of Western Australia, Perth, Western Australia Australia
| | - John Wray
- School of Pediatrics and Child Health, University of Western Australia, Perth, Western Australia Australia
| | - Andrew J O Whitehouse
- Telethon Kids Institute, University of Western Australia, Perth, Western Australia Australia.,Cooperative Research Centre for Living with Autism (Autism CRC), Long Pocket, Brisbane, Queensland Australia
| | - Adam J Guastella
- Autism Clinic for Translational Research, Brain and Mind Centre, Central Clinical School, Sydney Medical School, University of Sydney, 100 Mallett Street, Camperdown, New South Wales 2050 Australia
| |
Collapse
|
76
|
Yanguas-Casás N, Crespo-Castrillo A, de Ceballos ML, Chowen JA, Azcoitia I, Arevalo MA, Garcia-Segura LM. Sex differences in the phagocytic and migratory activity of microglia and their impairment by palmitic acid. Glia 2017; 66:522-537. [PMID: 29139169 DOI: 10.1002/glia.23263] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 10/25/2017] [Accepted: 10/30/2017] [Indexed: 12/11/2022]
Abstract
Sex differences in the incidence, clinical manifestation, disease course, and prognosis of neurological diseases, such as autism spectrum disorders or Alzheimer's disease, have been reported. Obesity has been postulated as a risk factor for cognitive decline and Alzheimer's disease and, during pregnancy, increases the risk of autism spectrum disorders in the offspring. Obesity is associated with increased serum and brain levels of free fatty acids, such as palmitic acid, which activate microglial cells triggering a potent inflammatory cascade. In this study, we have determined the effect of palmitic acid in the inflammatory profile, motility, and phagocytosis of primary male and female microglia, both in basal conditions and in the presence of a pro-inflammatory stimulus (interferon-γ). Male microglia in vitro showed higher migration than female microglia under basal and stimulated conditions. In contrast, female microglia had higher basal and stimulated phagocytic activity than male microglia. Palmitic acid did not affect basal migration or phagocytosis, but abolished the migration and phagocytic activity of male and female microglia in response to interferon-γ. These findings extend previous observations of sex differences in microglia and suggest that palmitic acid impairs the protective responses of these cells.
Collapse
Affiliation(s)
| | | | | | - Julie A Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, Madrid, 28009, Spain.,CIBER de Investigación Biomédica en Red: Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Iñigo Azcoitia
- Department of Cell Biology, Faculty of Biology, Universidad Complutense, Madrid, 28040, Spain.,CIBER de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Maria Angeles Arevalo
- Instituto Cajal, CSIC, Madrid, 28002, Spain.,CIBER de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Luis M Garcia-Segura
- Instituto Cajal, CSIC, Madrid, 28002, Spain.,CIBER de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
77
|
Morris G, Puri BK, Frye RE. The putative role of environmental aluminium in the development of chronic neuropathology in adults and children. How strong is the evidence and what could be the mechanisms involved? Metab Brain Dis 2017; 32:1335-1355. [PMID: 28752219 PMCID: PMC5596046 DOI: 10.1007/s11011-017-0077-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Accepted: 07/19/2017] [Indexed: 02/06/2023]
Abstract
The conceptualisation of autistic spectrum disorder and Alzheimer's disease has undergone something of a paradigm shift in recent years and rather than being viewed as single illnesses with a unitary pathogenesis and pathophysiology they are increasingly considered to be heterogeneous syndromes with a complex multifactorial aetiopathogenesis, involving a highly complex and diverse combination of genetic, epigenetic and environmental factors. One such environmental factor implicated as a potential cause in both syndromes is aluminium, as an element or as part of a salt, received, for example, in oral form or as an adjuvant. Such administration has the potential to induce pathology via several routes such as provoking dysfunction and/or activation of glial cells which play an indispensable role in the regulation of central nervous system homeostasis and neurodevelopment. Other routes include the generation of oxidative stress, depletion of reduced glutathione, direct and indirect reductions in mitochondrial performance and integrity, and increasing the production of proinflammatory cytokines in both the brain and peripherally. The mechanisms whereby environmental aluminium could contribute to the development of the highly specific pattern of neuropathology seen in Alzheimer's disease are described. Also detailed are several mechanisms whereby significant quantities of aluminium introduced via immunisation could produce chronic neuropathology in genetically susceptible children. Accordingly, it is recommended that the use of aluminium salts in immunisations should be discontinued and that adults should take steps to minimise their exposure to environmental aluminium.
Collapse
Affiliation(s)
- Gerwyn Morris
- Tir Na Nog, Bryn Road seaside 87, Llanelli, Wales, SA15 2LW, UK
| | - Basant K Puri
- Department of Medicine, Imperial College London, Hammersmith Hospital, London, England, W12 0HS, UK.
| | - Richard E Frye
- College of Medicine, Department of Pediatrics, University of Arkansas for Medical Sciences, Arkansas Children's Hospital Research Institute, Little Rock, AR, 72202, USA
| |
Collapse
|
78
|
Dipasquale V, Cutrupi MC, Colavita L, Manti S, Cuppari C, Salpietro C. Neuroinflammation in Autism Spectrum Disorders: Role of High Mobility Group Box 1 Protein. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2017; 6:148-155. [PMID: 29682486 PMCID: PMC5898638 DOI: 10.22088/acadpub.bums.6.3.148] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 09/06/2017] [Indexed: 12/13/2022]
Abstract
The pathogenesis of autism spectrum disorder (ASD) likely involves genetic and environmental factors, impacting the complex neurodevelopmental and behavioral abnormalities of the disorder. Scientific research studies emerging within the past two decades suggest that immune dysfunction and inflammation have pathogenic influences through different mechanisms, all leading to both a chronic state of low grade inflammation, and alterations in the central nervous system and immune response, respectively. The high mobility group box-1 protein (HMGB1) is an inflammatory marker which has been shown to play a role in inducing and influencing neuroinflammation. Current evidences suggest a possible role in the multiple pathogenic mechanisms of ASD. The aim of this manuscript is to review the major hypothesis for ASD pathogenesis, with specific regards to the immunological ones, and to provide a comprehensive review of the current data about the association between HMGB1 and ASD. A systematic search has been carried out through Medline via Pubmed to identify all original articles published in English, on the basis of the following keywords: “HMGB1”, “autism”, “autism spectrum disorder”, “neuroinflammation”, and “child”.
Collapse
Affiliation(s)
- Valeria Dipasquale
- Department of Adult and Childhood Human Pathology, Unit of Pediatrics, University Hospital of Messina, Messina, Italy
| | - Maria Concetta Cutrupi
- Department of Adult and Childhood Human Pathology, Unit of Pediatrics, University Hospital of Messina, Messina, Italy
| | - Laura Colavita
- Department of Pediatrics, Hospital Umberto I of Siracusa, Siracusa, Italy
| | - Sara Manti
- Department of Adult and Childhood Human Pathology, Unit of Pediatrics, University Hospital of Messina, Messina, Italy
| | - Caterina Cuppari
- Department of Adult and Childhood Human Pathology, Unit of Pediatrics, University Hospital of Messina, Messina, Italy
| | - Carmelo Salpietro
- Department of Adult and Childhood Human Pathology, Unit of Pediatrics, University Hospital of Messina, Messina, Italy
| |
Collapse
|
79
|
Saad K, Zahran AM, Elsayh KI, Abdel-Rahman AA, Al-Atram AA, Hussein A, El-Gendy YG. Frequency of Dendritic Cells and Their Expression of Costimulatory Molecules in Children with Autism Spectrum Disorders. J Autism Dev Disord 2017; 47:2671-2678. [PMID: 28589497 DOI: 10.1007/s10803-017-3190-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The aim of our study was to evaluate the frequencies of myeloid dendritic cells (mDCs) and plasmacytoid dendritic cells (pDCs) in children with ASD. Subjects were 32 children with ASD and 30 healthy children as controls. The numbers of mDCs and pDCs and the expression of CD86 and CD80 on the entire DCs were detected by flow cytometry. ASD children had significantly higher percentages of mDCs and pDCs when compared to controls. We found significant inverse relationships between serum 25-hydroxyvitamin D levels and the frequencies of mDCs and pDCs in autistic children. Our data suggested that DCs could play a role in the clinical course of ASD. The relationship of DCs to immune disorders in ASD remains to be determined.
Collapse
Affiliation(s)
- Khaled Saad
- Pediatric Department, Faculty of Medicine, Assiut University, Assiut, 71516, Egypt.
| | - Asmaa M Zahran
- Clinical Pathology Department, South Egypt Cancer Institute, Assiut University, Assiut, Egypt
| | - Khalid I Elsayh
- Pediatric Department, Faculty of Medicine, Assiut University, Assiut, 71516, Egypt
| | - Ahmed A Abdel-Rahman
- Department of Neuropsychiatry, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Abdulrahman A Al-Atram
- Department of Psychiatry, College of Medicine, Almajmaah University, Almajmaah, Kingdom of Saudi Arabia
| | - Almontaser Hussein
- Pediatric Department, Faculty of Medicine, Assiut University, Assiut, 71516, Egypt
| | - Yasmin G El-Gendy
- Pediatric Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
80
|
The Putative Role of Environmental Mercury in the Pathogenesis and Pathophysiology of Autism Spectrum Disorders and Subtypes. Mol Neurobiol 2017; 55:4834-4856. [DOI: 10.1007/s12035-017-0692-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Accepted: 07/13/2017] [Indexed: 01/28/2023]
|
81
|
Arcuri C, Mecca C, Bianchi R, Giambanco I, Donato R. The Pathophysiological Role of Microglia in Dynamic Surveillance, Phagocytosis and Structural Remodeling of the Developing CNS. Front Mol Neurosci 2017; 10:191. [PMID: 28674485 PMCID: PMC5474494 DOI: 10.3389/fnmol.2017.00191] [Citation(s) in RCA: 176] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 05/30/2017] [Indexed: 12/13/2022] Open
Abstract
In vertebrates, during an early wave of hematopoiesis in the yolk sac between embryonic day E7.0 and E9.0, cells of mesodermal leaflet addressed to macrophage lineage enter in developing central nervous system (CNS) and originate the developing native microglial cells. Depending on the species, microglial cells represent 5–20% of glial cells resident in adult brain. Here, we briefly discuss some canonical functions of the microglia, i.e., cytokine secretion and functional transition from M1 to M2 phenotype. In addition, we review studies on the non-canonical functions of microglia such as regulation of phagocytosis, synaptic pruning, and sculpting postnatal neural circuits. In this latter context the contribution of microglia to some neurodevelopmental disorders is now well established. Nasu-Hakola (NHD) disease is considered a primary microgliopathy with alterations of the DNAX activation protein 12 (DAP12)-Triggering receptor expressed on myeloid cells 2 (TREM-2) signaling and removal of macromolecules and apoptotic cells followed by secondary microglia activation. In Rett syndrome Mecp2-/- microglia shows a substantial impairment of phagocytic ability, although the role of microglia is not yet clear. In a mouse model of Tourette syndrome (TS), microglia abnormalities have also been described, and deficient microglia-mediated neuroprotection is obvious. Here we review the role of microglial cells in neurodevelopmental disorders without inflammation and on the complex role of microglia in developing CNS.
Collapse
Affiliation(s)
- Cataldo Arcuri
- Department of Experimental Medicine, Centro Universitario per la Ricerca sulla Genomica Funzionale, Perugia Medical School, University of PerugiaPerugia, Italy
| | - Carmen Mecca
- Department of Experimental Medicine, Centro Universitario per la Ricerca sulla Genomica Funzionale, Perugia Medical School, University of PerugiaPerugia, Italy
| | - Roberta Bianchi
- Department of Experimental Medicine, Centro Universitario per la Ricerca sulla Genomica Funzionale, Perugia Medical School, University of PerugiaPerugia, Italy
| | - Ileana Giambanco
- Department of Experimental Medicine, Centro Universitario per la Ricerca sulla Genomica Funzionale, Perugia Medical School, University of PerugiaPerugia, Italy
| | - Rosario Donato
- Department of Experimental Medicine, Centro Universitario per la Ricerca sulla Genomica Funzionale, Perugia Medical School, University of PerugiaPerugia, Italy
| |
Collapse
|
82
|
Habib SS, Al-Regaiey K, Bashir S, Iqbal M. Role of Endocannabinoids on Neuroinflammation in Autism Spectrum Disorder Prevention. J Clin Diagn Res 2017; 11:CE01-CE03. [PMID: 28764155 DOI: 10.7860/jcdr/2017/23862.9969] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 11/19/2016] [Indexed: 12/20/2022]
Abstract
Autism Spectrum Disorder (ASD) disease has become a mounting socio-economical alarm around the world. Neuroinflammtion had been shown in postmortem brain specimens from ASD patients. The Endocannabinoids System (ES) consists of a family of locally produced, short-lived, endogenous, phospholipid-derived agonists (endocannabinoids) that control energy balance and body composition. The growing number of medical benefits of ES, such as their ability to regulate processes like neuroinflammation, neurogenesis and memory, raise the question of their potential role as a preventive treatment of ASD. To test this hypothesis, basic and clinical studies allow us a thorough investigation of the role of ES in the pathogenesis of ASD. This hypothesis will help to understand the mechanism of ES and its role in ASD.
Collapse
Affiliation(s)
- Syed Shahid Habib
- Professor, Department of Physiology, King Saud University, Riyadh, Saudi Arabia
| | - Khalid Al-Regaiey
- Assistant Professor, Department of Physiology, King Saud University, Riyadh, Saudi Arabia
| | - Shahid Bashir
- Assistant Professor, Department of Physiology, King Saud University, Riyadh, Saudi Arabia
| | - Muhammad Iqbal
- Researcher, Department of Physiology, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
83
|
Szoko N, McShane AJ, Natowicz MR. Proteomic explorations of autism spectrum disorder. Autism Res 2017; 10:1460-1469. [DOI: 10.1002/aur.1803] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 03/30/2017] [Accepted: 04/01/2017] [Indexed: 02/06/2023]
Affiliation(s)
- Nicholas Szoko
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland Clinic; Cleveland OH
| | - Adam J. McShane
- Pathology & Laboratory Medicine Institute, Cleveland Clinic; Cleveland OH
| | - Marvin R. Natowicz
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland Clinic; Cleveland OH
- Pathology & Laboratory Medicine Institute, Cleveland Clinic; Cleveland OH
- Genomic Medicine, Neurology and Pediatrics Institutes, Cleveland Clinic; Cleveland OH
| |
Collapse
|
84
|
Qin Y, Chen Y, Yang J, Wu F, Zhao L, Yang F, Xue P, Shi Z, Song T, Huang C. Serum glycopattern and Maackia amurensis lectin-II binding glycoproteins in autism spectrum disorder. Sci Rep 2017; 7:46041. [PMID: 28485374 PMCID: PMC5423032 DOI: 10.1038/srep46041] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 03/07/2017] [Indexed: 12/12/2022] Open
Abstract
The pathophysiology of autistic spectrum disorder (ASD) is not fully understood and there are no diagnostic or predictive biomarkers. Glycosylation modified as many as 70% of all human proteins can sensitively reflect various pathological changes. However, little is known about the alterations of glycosylation and glycoproteins in ASD. In this study, serum glycopattern and the maackia amurensis lectin-II binding glycoproteins (MBGs) in 65 children with ASD and 65 age-matched typically developing (TD) children were compared by using lectin microarrays and lectin-magnetic particle conjugate-assisted LC-MS/MS analyses. Expression of Siaα2-3 Gal/GalNAc was significantly increased in pooled (fold change = 3.33, p < 0.001) and individual (p = 0.009) serum samples from ASD versus TD children. A total of 194 and 217 MGBs were identified from TD and ASD sera respectively, of which 74 proteins were specially identified or up-regulated in ASD. Bioinformatic analysis revealed abnormal complement cascade and aberrant regulation of response-to-stimulus that might be novel makers or markers for ASD. Moreover, increase of APOD α2-3 sialoglycosylation could sensitively and specifically distinguish ASD samples from TD samples (AUC is 0.88). In conclusion, alteration of MBGs expression and their sialoglycosylation may serve as potential biomarkers for diagnosis of ASD, and provide useful information for investigations into the pathogenesis of ASD.
Collapse
Affiliation(s)
- Yannan Qin
- Department of Cell Biology and Genetics, Environment and Genes Related to Diseases Key Laboratory of Education Ministry, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, P. R. China
| | - Yanni Chen
- Xi'an Child's Hospital of Medical College of Xi'an Jiaotong University, Xi'an Child's Hospital, Xi'an 710002, P. R. China
| | - Juan Yang
- Department of Cell Biology and Genetics, Environment and Genes Related to Diseases Key Laboratory of Education Ministry, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, P. R. China
| | - Fei Wu
- Department of Cell Biology and Genetics, Environment and Genes Related to Diseases Key Laboratory of Education Ministry, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, P. R. China
| | - Lingyu Zhao
- Department of Cell Biology and Genetics, Environment and Genes Related to Diseases Key Laboratory of Education Ministry, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, P. R. China
| | - Fuquan Yang
- Laboratory of Proteomics, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, P. R. China
| | - Peng Xue
- Laboratory of Proteomics, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, P. R. China
| | - Zhuoyue Shi
- The Department of Biology, College of Liberal Arts and Science, The University of Iowa, Iowa 430015, USA
| | - Tusheng Song
- Department of Cell Biology and Genetics, Environment and Genes Related to Diseases Key Laboratory of Education Ministry, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, P. R. China
| | - Chen Huang
- Department of Cell Biology and Genetics, Environment and Genes Related to Diseases Key Laboratory of Education Ministry, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, P. R. China
| |
Collapse
|
85
|
|
86
|
Li Q, Han Y, Dy ABC, Hagerman RJ. The Gut Microbiota and Autism Spectrum Disorders. Front Cell Neurosci 2017; 11:120. [PMID: 28503135 PMCID: PMC5408485 DOI: 10.3389/fncel.2017.00120] [Citation(s) in RCA: 239] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 04/10/2017] [Indexed: 12/19/2022] Open
Abstract
Gastrointestinal (GI) symptoms are a common comorbidity in patients with autism spectrum disorder (ASD), but the underlying mechanisms are unknown. Many studies have shown alterations in the composition of the fecal flora and metabolic products of the gut microbiome in patients with ASD. The gut microbiota influences brain development and behaviors through the neuroendocrine, neuroimmune and autonomic nervous systems. In addition, an abnormal gut microbiota is associated with several diseases, such as inflammatory bowel disease (IBD), ASD and mood disorders. Here, we review the bidirectional interactions between the central nervous system and the gastrointestinal tract (brain-gut axis) and the role of the gut microbiota in the central nervous system (CNS) and ASD. Microbiome-mediated therapies might be a safe and effective treatment for ASD.
Collapse
Affiliation(s)
- Qinrui Li
- Department of Pediatrics, Peking University First HospitalBeijing, China
| | - Ying Han
- Department of Pediatrics, Peking University First HospitalBeijing, China
| | - Angel Belle C Dy
- School of Medicine and Public Health, Ateneo de Manila UniversityQuezon City, Philippines
| | - Randi J Hagerman
- MIND Institute, University of California Davis Medical CenterSacramento, CA, USA.,Department of Pediatrics, University of California Davis Medical CenterSacramento, CA, USA
| |
Collapse
|
87
|
Differential Gender Effects in the Relationship between Perceived Immune Functioning and Autistic Traits. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2017; 14:ijerph14040409. [PMID: 28417950 PMCID: PMC5409610 DOI: 10.3390/ijerph14040409] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Revised: 03/27/2017] [Accepted: 04/07/2017] [Indexed: 12/20/2022]
Abstract
Altered immune functioning has been demonstrated in individuals with autism spectrum disorder (ASD). The current study explores the relationship between perceived immune functioning and experiencing ASD traits in healthy young adults. N = 410 students from Utrecht University completed a survey on immune functioning and autistic traits. In addition to a 1-item perceived immune functioning rating, the Immune Function Questionnaire (IFQ) was completed to assess perceived immune functioning. The Dutch translation of the Autism-Spectrum Quotient (AQ) was completed to examine variation in autistic traits, including the domains “social insights and behavior”, “difficulties with change”, “communication”, “phantasy and imagination”, and “detail orientation”. The 1-item perceived immune functioning score did not significantly correlate with the total AQ score. However, a significant negative correlation was found between perceived immune functioning and the AQ subscale “difficulties with change” (r = −0.119, p = 0.019). In women, 1-item perceived immune functioning correlated significantly with the AQ subscales “difficulties with change” (r = −0.149, p = 0.029) and “communication” (r = −0.145, p = 0.032). In men, none of the AQ subscales significantly correlated with 1-item perceived immune functioning. In conclusion, a modest relationship between perceived immune functioning and several autistic traits was found.
Collapse
|
88
|
Independent Maternal and Fetal Genetic Effects on Midgestational Circulating Levels of Environmental Pollutants. G3-GENES GENOMES GENETICS 2017; 7:1287-1299. [PMID: 28235828 PMCID: PMC5386877 DOI: 10.1534/g3.117.039784] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Maternal exposure to environmental pollutants could affect fetal brain development and increase autism spectrum disorder (ASD) risk in conjunction with differential genetic susceptibility. Organohalogen congeners measured in maternal midpregnancy blood samples have recently shown significant, but negative associations with offspring ASD outcome. We report the first large-scale maternal and fetal genetic study of the midpregnancy serum levels of a set of 21 organohalogens in a subset of 790 genotyped women and 764 children collected in California by the Early Markers for Autism (EMA) Project. Levels of PCB (polychlorinated biphenyl) and PBDE (polybrominated diphenyl ether) congeners showed high maternal and fetal estimated SNP-based heritability (h2g) accounting for 39–99% of the total variance. Genome-wide association analyses identified significant maternal loci for p,p′-DDE (P = 7.8 × 10−11) in the CYP2B6 gene and for BDE-28 (P = 3.2 × 10−8) near the SH3GL2 gene, both involved in xenobiotic and lipid metabolism. Fetal genetic loci contributed to the levels of BDE-100 (P = 4.6 × 10−8) and PCB187 (P = 2.8 × 10−8), near the potential metabolic genes LOXHD1 and PTPRD, previously implicated in neurodevelopment. Negative associations were observed for BDE-100, BDE153, and the sum of PBDEs with ASD, partly explained by genome-wide additive genetic effects that predicted PBDE levels. Our results support genetic control of midgestational biomarkers for environmental exposures by nonoverlapping maternal and fetal genetic determinants, suggesting that future studies of environmental risk factors should take genetic variation into consideration. The independent influence of fetal genetics supports previous hypotheses that fetal genotypes expressed in placenta can influence maternal physiology and the transplacental transfer of organohalogens.
Collapse
|
89
|
Voltas N, Arija V, Hernández-Martínez C, Jiménez-Feijoo R, Ferré N, Canals J. Are there early inflammatory biomarkers that affect neurodevelopment in infancy? J Neuroimmunol 2017; 305:42-50. [DOI: 10.1016/j.jneuroim.2017.01.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 12/31/2016] [Accepted: 01/11/2017] [Indexed: 12/21/2022]
|
90
|
O'Connor RM, Stone EF, Wayne CR, Marcinkevicius EV, Ulgherait M, Delventhal R, Pantalia MM, Hill VM, Zhou CG, McAllister S, Chen A, Ziegenfuss JS, Grueber WB, Canman JC, Shirasu-Hiza MM. A Drosophila model of Fragile X syndrome exhibits defects in phagocytosis by innate immune cells. J Cell Biol 2017; 216:595-605. [PMID: 28223318 PMCID: PMC5350515 DOI: 10.1083/jcb.201607093] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Revised: 11/22/2016] [Accepted: 01/30/2017] [Indexed: 11/22/2022] Open
Abstract
Fragile X syndrome, the most common known monogenic cause of autism, results from the loss of FMR1, a conserved, ubiquitously expressed RNA-binding protein. Recent evidence suggests that Fragile X syndrome and other types of autism are associated with immune system defects. We found that Drosophila melanogaster Fmr1 mutants exhibit increased sensitivity to bacterial infection and decreased phagocytosis of bacteria by systemic immune cells. Using tissue-specific RNAi-mediated knockdown, we showed that Fmr1 plays a cell-autonomous role in the phagocytosis of bacteria. Fmr1 mutants also exhibit delays in two processes that require phagocytosis by glial cells, the immune cells in the brain: neuronal clearance after injury in adults and the development of the mushroom body, a brain structure required for learning and memory. Delayed neuronal clearance is associated with reduced recruitment of activated glia to the site of injury. These results suggest a previously unrecognized role for Fmr1 in regulating the activation of phagocytic immune cells both in the body and the brain.
Collapse
Affiliation(s)
- Reed M O'Connor
- Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032
| | - Elizabeth F Stone
- Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032
| | - Charlotte R Wayne
- Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032
| | - Emily V Marcinkevicius
- Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032
| | - Matt Ulgherait
- Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032
| | - Rebecca Delventhal
- Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032
| | - Meghan M Pantalia
- Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032
| | - Vanessa M Hill
- Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032
| | - Clarice G Zhou
- Department of Biological Sciences, Columbia University, New York, NY 10025
| | - Sophie McAllister
- Department of Biological Sciences, Columbia University, New York, NY 10025
| | - Anna Chen
- Department of Biological Sciences, Columbia University, New York, NY 10025
| | - Jennifer S Ziegenfuss
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY 10032
| | - Wesley B Grueber
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY 10032
| | - Julie C Canman
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032
| | - Mimi M Shirasu-Hiza
- Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032
| |
Collapse
|
91
|
Lyall K, Croen LA, Sjödin A, Yoshida CK, Zerbo O, Kharrazi M, Windham GC. Polychlorinated Biphenyl and Organochlorine Pesticide Concentrations in Maternal Mid-Pregnancy Serum Samples: Association with Autism Spectrum Disorder and Intellectual Disability. ENVIRONMENTAL HEALTH PERSPECTIVES 2017; 125:474-480. [PMID: 27548254 PMCID: PMC5332182 DOI: 10.1289/ehp277] [Citation(s) in RCA: 134] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 06/27/2016] [Accepted: 07/20/2016] [Indexed: 05/18/2023]
Abstract
BACKGROUND Polychlorinated biphenyls (PCBs) and organochlorine pesticides (OCPs) are neurodevelopmental toxicants, but few studies have examined associations with autism spectrum disorder (ASD). OBJECTIVES We aimed to determine whether prenatal exposure to PCBs and OCPs influences offspring risk of ASD and intellectual disability without autism (ID). METHODS We conducted a population-based case-control study among Southern California births, including children with ASD (n = 545) meeting Diagnostic and Statistical Manual of Mental Disorders, 4th Edition (DSM-IV-TR) criteria and ID (n = 181), as well as general population (GP) controls (n = 418). Concentrations of 11 PCB congeners and 2 OCPs measured in banked second-trimester serum samples were compared between the diagnostic groups. Logistic regression was used to calculate crude and adjusted odds ratios (AOR) for associations with ASD, and separately for ID, compared with GP controls, by quartiles of analyte concentrations in primary analyses. RESULTS Geometric mean levels of several PCB congeners were higher in the ASD group than in the ID and GP groups. ASD risk was elevated for a number of PCB congeners, particularly for the highest vs. lowest quartile of PCB138/158 (AOR = 1.79; 95% CI: 1.10, 2.71) and PCB153 (AOR = 1.82; 95% CI: 1.10, 3.02), and for highest deciles of other congeners in secondary analyses. PCB138/158 was also associated with increased ID (AOR = 2.41; 95% CI: 1.18, 4.91), though no trend was suggested. OCPs were not associated with increased risk of ASD in primary analyses, whereas nonmonotonic increases in risk of ID were found with p,p´-DDE. CONCLUSIONS Our results suggest higher levels of some organochlorine compounds during pregnancy are associated with ASD and ID. Citation: Lyall K, Croen LA, Sjödin A, Yoshida CK, Zerbo O, Kharrazi M, Windham GC. 2017. Polychlorinated biphenyl and organochlorine pesticide concentrations in maternal mid-pregnancy serum samples: association with autism spectrum disorder and intellectual disability. Environ Health Perspect 125:474-480; http://dx.doi.org/10.1289/EHP277.
Collapse
Affiliation(s)
- Kristen Lyall
- Environmental Health Investigations Branch, California Department of Public Health, Richmond, California, USA
- Address correspondence to K. Lyall, AJ Drexel Autism Institute, Suite 560, 3200 Market St., Philadelphia, PA 19104 USA. Telephone: (215) 571-3215. E-mail:
| | - Lisa. A. Croen
- Division of Research, Kaiser Permanente, Oakland, California, USA
| | - Andreas Sjödin
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | | | - Ousseny Zerbo
- Division of Research, Kaiser Permanente, Oakland, California, USA
| | - Martin Kharrazi
- Environmental Health Investigations Branch, California Department of Public Health, Richmond, California, USA
| | - Gayle C. Windham
- Environmental Health Investigations Branch, California Department of Public Health, Richmond, California, USA
| |
Collapse
|
92
|
Masi A, Glozier N, Dale R, Guastella AJ. The Immune System, Cytokines, and Biomarkers in Autism Spectrum Disorder. Neurosci Bull 2017; 33:194-204. [PMID: 28238116 PMCID: PMC5360854 DOI: 10.1007/s12264-017-0103-8] [Citation(s) in RCA: 151] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Accepted: 01/22/2017] [Indexed: 12/24/2022] Open
Abstract
Autism Spectrum Disorder (ASD) is a pervasive neurodevelopmental condition characterized by variable impairments in communication and social interaction as well as restricted interests and repetitive behaviors. Heterogeneity of presentation is a hallmark. Investigations of immune system problems in ASD, including aberrations in cytokine profiles and signaling, have been increasing in recent times and are the subject of ongoing interest. With the aim of establishing whether cytokines have utility as potential biomarkers that may define a subgroup of ASD, or function as an objective measure of response to treatment, this review summarizes the role of the immune system, discusses the relationship between the immune system, the brain, and behavior, and presents previously-identified immune system abnormalities in ASD, specifically addressing the role of cytokines in these aberrations. The roles and identification of biomarkers are also addressed, particularly with respect to cytokine profiles in ASD.
Collapse
Affiliation(s)
- Anne Masi
- Autism Clinic for Translational Research, Brain and Mind Centre, Central Clinical School, Sydney Medical School, University of Sydney, Sydney, New South Wales, 2050, Australia
| | - Nicholas Glozier
- Autism Clinic for Translational Research, Brain and Mind Centre, Central Clinical School, Sydney Medical School, University of Sydney, Sydney, New South Wales, 2050, Australia
| | - Russell Dale
- Childrens Hospital at Westmead Clinical School, University of Sydney, Sydney, New South Wales, 2145, Australia
| | - Adam J Guastella
- Autism Clinic for Translational Research, Brain and Mind Centre, Central Clinical School, Sydney Medical School, University of Sydney, Sydney, New South Wales, 2050, Australia.
| |
Collapse
|
93
|
Autism with intellectual disability is associated with increased levels of maternal cytokines and chemokines during gestation. Mol Psychiatry 2017; 22:273-279. [PMID: 27217154 PMCID: PMC5122473 DOI: 10.1038/mp.2016.77] [Citation(s) in RCA: 178] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 03/24/2016] [Accepted: 03/31/2016] [Indexed: 12/29/2022]
Abstract
Immune abnormalities have been described in some individuals with autism spectrum disorders (ASDs) as well as their family members. However, few studies have directly investigated the role of prenatal cytokine and chemokine profiles on neurodevelopmental outcomes in humans. In the current study, we characterized mid-gestational serum profiles of 22 cytokines and chemokines in mothers of children with ASD (N=415), developmental delay (DD) without ASD (N=188), and general population (GP) controls (N=428) using a bead-based multiplex technology. The ASD group was further divided into those with intellectual disabilities (developmental/cognitive and adaptive composite score<70) (ASD+ID, N=184) and those without (composite score⩾70) (ASD-noID, N=201). Levels of cytokines and chemokines were compared between groups using multivariate logistic regression analyses, adjusting for maternal age, ethnicity, birth country and weight, as well as infant gender, birth year and birth month. Mothers of children with ASD+ID had significantly elevated mid-gestational levels of numerous cytokines and chemokines, such as granulocyte macrophage colony-stimulating factor, interferon-γ, interleukin-1α (IL-1α) and IL-6, compared with mothers of children with either ASD-noID, those with DD, or GP controls. Conversely, mothers of children with either ASD-noID or with DD had significantly lower levels of the chemokines IL-8 and monocyte chemotactic protein-1 compared with mothers of GP controls. This observed immunologic distinction between mothers of children with ASD+ID from mothers of children with ASD-noID or DD suggests that the intellectual disability associated with ASD might be etiologically distinct from DD without ASD. These findings contribute to the ongoing efforts toward identification of early biological markers specific to subphenotypes of ASD.
Collapse
|
94
|
Pardo CA, Farmer CA, Thurm A, Shebl FM, Ilieva J, Kalra S, Swedo S. Serum and cerebrospinal fluid immune mediators in children with autistic disorder: a longitudinal study. Mol Autism 2017; 8:1. [PMID: 28070266 PMCID: PMC5217649 DOI: 10.1186/s13229-016-0115-7] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 12/14/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The causes of autism likely involve genetic and environmental factors that influence neurobiological changes and the neurological and behavioral features of the disorder. Immune factors and inflammation are hypothesized pathogenic influences, but have not been examined longitudinally. METHODS In a cohort of 104 participants with autism, we performed an assessment of immune mediators such as cytokines, chemokines, or growth factors in serum and cerebrospinal fluid (n = 67) to determine potential influences of such mediators in autism. RESULTS As compared with 54 typically developing controls, we found no evidence of differences in the blood profile of immune mediators supportive of active systemic inflammation mechanisms in participants with autism. Some modulators of immune function (e.g., EGF and soluble CD40 ligand) were increased in the autism group; however, no evidence of group differences in traditional markers of active inflammation (e.g., IL-6, TNFα, IL-1β) were observed in the serum. Further, within-subject stability (measured by estimated intraclass correlations) of most analytes was low, indicating that a single measurement is not a reliable prospective indicator of concentration for most analytes. Additionally, in participants with autism, there was little correspondence between the blood and CSF profiles of cytokines, chemokines, and growth factors, suggesting that peripheral markers may not optimally reflect the immune status of the central nervous system. Although the relatively high fraction of intrathecal production of selected chemokines involved in monocyte/microglia function may suggest a possible relationship with the homeostatic role of microglia, control data are needed for further interpretation of its relevance in autism. CONCLUSIONS These longitudinal observations fail to provide support for the hypothesized role of disturbances in the expression of circulating cytokines and chemokines as an indicator of systemic inflammation in autism. ClinicalTrials.gov, NCT00298246.
Collapse
Affiliation(s)
- Carlos A Pardo
- Johns Hopkins University School of Medicine, 627 Pathology Bld., 6000 North Wolfe Street, Baltimore, MD 21287 USA
| | - Cristan A Farmer
- Pediatrics and Developmental Neuroscience, National Institute of Mental Health, Bethesda, MD USA
| | - Audrey Thurm
- Pediatrics and Developmental Neuroscience, National Institute of Mental Health, Bethesda, MD USA
| | - Fatma M Shebl
- Yale School of Public Health, Yale University, New Haven, CT USA
| | - Jorjetta Ilieva
- Johns Hopkins University School of Medicine, 627 Pathology Bld., 6000 North Wolfe Street, Baltimore, MD 21287 USA
| | - Simran Kalra
- Pediatrics and Developmental Neuroscience, National Institute of Mental Health, Bethesda, MD USA
| | - Susan Swedo
- Pediatrics and Developmental Neuroscience, National Institute of Mental Health, Bethesda, MD USA
| |
Collapse
|
95
|
Neuroinflammation in Autism: Plausible Role of Maternal Inflammation, Dietary Omega 3, and Microbiota. Neural Plast 2016; 2016:3597209. [PMID: 27840741 PMCID: PMC5093279 DOI: 10.1155/2016/3597209] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 08/24/2016] [Accepted: 09/27/2016] [Indexed: 02/06/2023] Open
Abstract
Several genetic causes of autism spectrum disorder (ASD) have been identified. However, more recent work has highlighted that certain environmental exposures early in life may also account for some cases of autism. Environmental insults during pregnancy, such as infection or malnutrition, seem to dramatically impact brain development. Maternal viral or bacterial infections have been characterized as disruptors of brain shaping, even if their underlying mechanisms are not yet fully understood. Poor nutritional diversity, as well as nutrient deficiency, is strongly associated with neurodevelopmental disorders in children. For instance, imbalanced levels of essential fatty acids, and especially polyunsaturated fatty acids (PUFAs), are observed in patients with ASD and other neurodevelopmental disorders (e.g., attention deficit hyperactivity disorder (ADHD) and schizophrenia). Interestingly, PUFAs, and specifically n-3 PUFAs, are powerful immunomodulators that exert anti-inflammatory properties. These prenatal dietary and immunologic factors not only impact the fetal brain, but also affect the microbiota. Recent work suggests that the microbiota could be the missing link between environmental insults in prenatal life and future neurodevelopmental disorders. As both nutrition and inflammation can massively affect the microbiota, we discuss here how understanding the crosstalk between these three actors could provide a promising framework to better elucidate ASD etiology.
Collapse
|
96
|
Bryn V, Aass HCD, Skjeldal OH, Isaksen J, Saugstad OD, Ormstad H. Cytokine Profile in Autism Spectrum Disorders in Children. J Mol Neurosci 2016; 61:1-7. [PMID: 27730473 DOI: 10.1007/s12031-016-0847-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Accepted: 09/22/2016] [Indexed: 12/19/2022]
Abstract
The pathogenesis of autism spectrum disorders (ASD) is not completely understood, but there is evidence of associations with altered immune responses. The aim of this study was to determine the serum levels of various cytokines in children with ASD and in healthy controls, in order to determine their role in ASD and its diagnostic subgroups. Sixty-five ASD patients were enrolled from an epidemiological survey in Norway, of which 30 were diagnosed with childhood autism, 16 with Asperger syndrome, 12 with atypical autism, 1 with Rett syndrome, and 6 with another ASD diagnosis. The serum levels of 12 cytokines were measured in all of the patients and in 30 healthy children. The cytokine levels did not differ significantly between the ASD group and the healthy controls. However, the interleukin-8 (IL-8) level was significantly higher (6.82 vs 4.58 pg/ml, p = 0.017) while that of IL-10 was significantly lower (2.24 vs 6.49 pg/ml, p = 0.009) in patients with childhood autism than in controls. Furthermore, the IL-8 level was significantly higher in childhood autism than in Asperger syndrome (6.82 vs 4.05 pg/ml, p = 0.013). Our study shows that the cytokine profile of children diagnosed with ASD, regardless of the subdiagnosis, does not differ from healthy controls. However, differentiation into different diagnostic subgroups reveals significantly different levels of IL-8 and IL-10. This indicates that different mechanisms may underlie the different ASD subdiagnoses. Future research into the pathophysiological mechanisms of ASD should pay more attention to the different subdiagnoses of ASD.
Collapse
Affiliation(s)
- Vesna Bryn
- Department of Pediatrics, Innlandet Hospital Trust, Anders Sandvigs 17, 2629, Lillehammer, Norway.
| | | | - Ola H Skjeldal
- Gillberg Neuropsychiatry Centre, University of Gothenburg, Gothenburg, Sweden
| | - Jørn Isaksen
- Department of Habilitation, Innlandet Hospital Trust, Lillehammer, Norway
| | - Ola Didrik Saugstad
- Pediatric Research Institute, Rikshospitalet, Oslo University Hospital, Oslo, Norway
| | - Heidi Ormstad
- Faculty of Health Sciences, University College of Southeast Norway, Drammen, Norway
| |
Collapse
|
97
|
Heifert TA, Susi A, Hisle-Gorman E, Erdie-Lalena CR, Gorman G, Min SB, Nylund CM. Feeding Disorders in Children With Autism Spectrum Disorders Are Associated With Eosinophilic Esophagitis. J Pediatr Gastroenterol Nutr 2016; 63:e69-73. [PMID: 27276430 DOI: 10.1097/mpg.0000000000001282] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Eosinophilic esophagitis (EoE) can present as food selectivity or feeding disorders in children. Children with autism spectrum disorders (ASDs) commonly demonstrate behavioral food selectivity in type and texture, which often leads to the diagnosis of feeding disorder. We sought to evaluate the association of ASD with EoE. METHODS A retrospective matched case-cohort study was performed using the Military Health System database from October 2008 to September 2013. We performed a 1:5 case-control match by age, sex, and enrollment timeframe. Feeding disorders, EoE, and atopic disorders were defined using diagnostic and procedure codes. RESULTS There were 45,286 children with ASD and 226,430 matched controls. EoE was more common in children with ASD (0.4%) compared with controls (0.1%). Feeding disorders were associated with EoE in both children with ASD and controls. Feeding disorders also had a higher odds ratio for EoE compared with other atopic conditions, among both children with ASD (7.17, 95% confidence interval [CI] 4.87-10.5) and controls (11.5, 95% CI 7.57-17.5). Compared with controls with a feeding disorder, children with ASD and a feeding disorder had no difference in the rate of diagnosed EoE (0.85, 0.95% CI 0.39-1.88). CONCLUSIONS Children with ASD are more likely to be diagnosed with EoE compared with controls; however, among children with feeding disorders, there is no difference in the odds of EoE. A diagnosis of feeding disorder was strongly associated with EoE. Feeding disorders in children with ASD should not be assumed to be solely behavioral and an esophagogastroduodenoscopy should be performed to evaluate for EoE.
Collapse
Affiliation(s)
- Theresa A Heifert
- *Department of Pediatrics, Uniformed Services University, Bethesda, MD †Department of Pediatrics, Womack Army Medical Center, Fort Bragg, NC ‡Department of Pediatrics, Walter Reed National Military Medical Center, Bethesda, MD
| | | | | | | | | | | | | |
Collapse
|
98
|
Abstract
Animals share an intimate and life-long partnership with a myriad of resident microbial species, collectively referred to as the microbiota. Symbiotic microbes have been shown to regulate nutrition and metabolism and are critical for the development and function of the immune system. More recently, studies have suggested that gut bacteria can impact neurological outcomes--altering behavior and potentially affecting the onset and/or severity of nervous system disorders. In this review, we highlight emerging evidence that the microbiome extends its influence to the brain via various pathways connecting the gut to the central nervous system. While understanding and appreciation of a gut microbial impact on neurological function is nascent, unraveling gut-microbiome-brain connections holds the promise of transforming the neurosciences and revealing potentially novel etiologies for psychiatric and neurodegenerative disorders.
Collapse
Affiliation(s)
- Timothy R Sampson
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Sarkis K Mazmanian
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
99
|
Edmonson CA, Ziats MN, Rennert OM. A Non-inflammatory Role for Microglia in Autism Spectrum Disorders. Front Neurol 2016; 7:9. [PMID: 26869989 PMCID: PMC4734207 DOI: 10.3389/fneur.2016.00009] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 01/19/2016] [Indexed: 12/25/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by deficits in social interaction, difficulties with language, and repetitive/restricted behaviors. The etiology of ASD is still largely unclear, but immune dysfunction and abnormalities in synaptogenesis have repeatedly been implicated as contributing to the disease phenotype. However, an understanding of how and if these two processes are related has not firmly been established. As non-inflammatory roles of microglia become increasingly recognized as critical to normal neurodevelopment, it is important to consider how dysfunction in these processes might explain the seemingly disparate findings of immune dysfunction and aberrant synaptogenesis seen in ASD. In this review, we highlight research demonstrating the importance of microglia to the development of normal neural networks, review recent studies demonstrating abnormal microglia in autism, and discuss how the relationship between these processes may contribute to the development of autism and other neurodevelopmental disorders at the cellular level.
Collapse
Affiliation(s)
- Catherine A Edmonson
- University of Florida College of Medicine, Gainesville, FL, USA; National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Mark N Ziats
- National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA; Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA
| | - Owen M Rennert
- National Institute of Child Health and Human Development, National Institutes of Health , Bethesda, MD , USA
| |
Collapse
|
100
|
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: 56] [Impact Index Per Article: 7.0] [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
|