1
|
Jana A, Nath A, Sen P, Kundu S, Alghamdi BS, Abujamel TS, Saboor M, Woon-Khiong C, Alexiou A, Papadakis M, Alam MZ, Ashraf GM. Unraveling the Endocannabinoid System: Exploring Its Therapeutic Potential in Autism Spectrum Disorder. Neuromolecular Med 2024; 26:20. [PMID: 38744725 PMCID: PMC11093854 DOI: 10.1007/s12017-024-08781-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 03/04/2024] [Indexed: 05/16/2024]
Abstract
The salient features of autism spectrum disorder (ASD) encompass persistent difficulties in social communication, as well as the presence of restricted and repetitive facets of behavior, hobbies, or pursuits, which are often accompanied with cognitive limitations. Over the past few decades, a sizable number of studies have been conducted to enhance our understanding of the pathophysiology of ASD. Preclinical rat models have proven to be extremely valuable in simulating and analyzing the roles of a wide range of established environmental and genetic factors. Recent research has also demonstrated the significant involvement of the endocannabinoid system (ECS) in the pathogenesis of several neuropsychiatric diseases, including ASD. In fact, the ECS has the potential to regulate a multitude of metabolic and cellular pathways associated with autism, including the immune system. Moreover, the ECS has emerged as a promising target for intervention with high predictive validity. Particularly noteworthy are resent preclinical studies in rodents, which describe the onset of ASD-like symptoms after various genetic or pharmacological interventions targeting the ECS, providing encouraging evidence for further exploration in this area.
Collapse
Affiliation(s)
- Ankit Jana
- Department of Biological Sciences, National University of Singapore, Singapore, 117558, Singapore
| | - Arnab Nath
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore, Karnataka, 560012, India
| | - Palash Sen
- School of Biosciences, Indian Institute of Technology, Kharagpur, West Bengal, 721302, India
| | - Swikriti Kundu
- Siksha Bhavana, Visva-Bharati University, Bolpur, West Bengal, 731235, India
| | - Badrah S Alghamdi
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Physiology, Neuroscience Unit, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Turki S Abujamel
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Muhammad Saboor
- Department of Medical Laboratory Sciences, College of Health Sciences, and Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
| | - Chan Woon-Khiong
- Department of Biological Sciences, National University of Singapore, Singapore, 117558, Singapore
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Chandigarh-Ludhiana Highway, Mohali, Punjab, India
- Department of Research & Development, Funogen, Athens, Greece
- Department of Research & Development, AFNP Med, 1030, Vienna, Austria
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW, 2770, Australia
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany.
| | - Mohammad Zubair Alam
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ghulam Md Ashraf
- Department of Medical Laboratory Sciences, College of Health Sciences, and Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates.
| |
Collapse
|
2
|
Kim DHJ, Iosif AM, Ramirez-Celis A, Ashwood P, Ames JL, Lyall K, Berger K, Croen LA, Van de Water J. Neonatal immune signatures differ by sex regardless of neurodevelopmental disorder status: Macrophage migration inhibitory factor (MIF) alone reveals a sex by diagnosis interaction effect. Brain Behav Immun 2023; 111:328-333. [PMID: 37164311 PMCID: PMC10796272 DOI: 10.1016/j.bbi.2023.05.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/10/2023] [Accepted: 05/05/2023] [Indexed: 05/12/2023] Open
Abstract
Immune dysregulation, including aberrant peripheral cytokine/chemokine levels, is implicated in neurodevelopmental disorders (NDD) such as autism spectrum disorder (ASD). While the diagnosis of ASD is more common in males compared to females, sex effects in immune dysregulation related to neurodevelopment remain understudied. The aim of this exploratory study was to determine whether there are sex-specific effects in neonatal immune dysregulation with respect to an ASD or delayed development (DD) diagnosis. We utilized the data from the Early Markers for Autism study, a population based case-control study of prenatal and neonatal biomarkers of ASD. The immune profile of newborns later diagnosed with ASD (n = 482, 91 females), DD (n = 140, 61 females) and sex-matched general population controls (GP; n = 378, 67 females) were analyzed using neonatal bloodspots (NBS) via 42-plex multiplex assay. Multiple linear regression analysis was performed to identify whether sex was associated with differences in cytokine/chemokine levels of children with ASD, DD, and GP. A sex by diagnosis interaction effect was observed only for the chemokine macrophage migration inhibitory factor (MIF), with males displaying higher levels of NBS MIF than females in the GP control group (p = 0.02), but not in ASD (p = 0.52) or DD (p = 0.29) groups. We found that regardless of child diagnosis, newborn bloodspot eluates from females had a significantly higher concentration than males with the same diagnosis of the chemokines granulocyte chemotactic protein 2 (GCP-2; p < 0.0001), macrophage inflammatory protein 2-alpha (GROβ; p = 0.002), interferon-inducible t-cell alpha chemoattractant (I-TAC; p < 0.0001), stromal cell-derived factor 1 alpha and beta (SDF-1α-β; p = 0.03), innate inflammatory chemokines interferon-gamma induced protein 10 (IP-10; p = 0.02), macrophage inflammatory protein 1-alpha (MIP-1α; p = 0.02), and Th1-related pro-inflammatory cytokine interleukin-12 active heterodimer (IL-12p70; p = 0.002). In contrast, males had a higher concentration than females of secondary lymphoid-tissue chemokine (6CKINE; p = 0.02), monocyte chemotactic protein 1 (MCP-1; p = 0.005) and myeloid progenitor inhibitory factor 1 (MPIF-1; p = 0.03). Results were similar when analyses were restricted to NBS from DD and ASD further classified as ASD with intellectual disability (ID), ASD without ID, and DD (GCP-2, p = 0.007; I-TAC, p = 0.001; IP-10, p = 0.005; IL-12p70, p = 0.03 higher in females; MPIF-1, p = 0.03 higher in male). This study is the first to examine sex differences in neonatal cytokine/chemokine concentrations, and whether these differences are associated with neurodevelopmental outcomes. Results highlight the importance of considering sex as a critical factor in understanding the immune system as it relates to child development.
Collapse
Affiliation(s)
- Danielle H J Kim
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, University of California, Davis, CA, USA
| | - Ana-Maria Iosif
- Department of Public Health Sciences, University of California, Davis, CA, USA
| | - Alexandra Ramirez-Celis
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, University of California, Davis, CA, USA
| | - Paul Ashwood
- MIND Institute, University of California, Davis, CA, USA
| | | | - Kristen Lyall
- AJ Drexel Autism Institute, Drexel University, Philadelphia, PA, USA
| | | | - Lisa A Croen
- Kaiser Permanente Northern California-Oakland, USA
| | - Judy Van de Water
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, University of California, Davis, CA, USA; MIND Institute, University of California, Davis, CA, USA.
| |
Collapse
|
3
|
Lampiasi N, Bonaventura R, Deidda I, Zito F, Russo R. Inflammation and the Potential Implication of Macrophage-Microglia Polarization in Human ASD: An Overview. Int J Mol Sci 2023; 24:2703. [PMID: 36769026 PMCID: PMC9916462 DOI: 10.3390/ijms24032703] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/23/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
Autism spectrum disorder (ASD) is a heterogeneous collection of neurodevelopmental disorders, difficult to diagnose and currently lacking treatment options. The possibility of finding reliable biomarkers useful for early identification would offer the opportunity to intervene with treatment strategies to improve the life quality of ASD patients. To date, there are many recognized risk factors for the development of ASD, both genetic and non-genetic. Although genetic and epigenetic factors may play a critical role, the extent of their contribution to ASD risk is still under study. On the other hand, non-genetic risk factors include pollution, nutrition, infection, psychological states, and lifestyle, all together known as the exposome, which impacts the mother's and fetus's life, especially during pregnancy. Pathogenic and non-pathogenic maternal immune activation (MIA) and autoimmune diseases can cause various alterations in the fetal environment, also contributing to the etiology of ASD in offspring. Activation of monocytes, macrophages, mast cells and microglia and high production of pro-inflammatory cytokines are indeed the cause of neuroinflammation, and the latter is involved in ASD's onset and development. In this review, we focused on non-genetic risk factors, especially on the connection between inflammation, macrophage polarization and ASD syndrome, MIA, and the involvement of microglia.
Collapse
Affiliation(s)
- Nadia Lampiasi
- Istituto per la Ricerca e l’Innovazione Biomedica IRIB, Consiglio Nazionale delle Ricerche, Via Ugo La Malfa 153, 90146 Palermo, Italy
| | | | | | | | | |
Collapse
|
4
|
Nabi SU, Rehman MU, Arafah A, Taifa S, Khan IS, Khan A, Rashid S, Jan F, Wani HA, Ahmad SF. Treatment of Autism Spectrum Disorders by Mitochondrial-targeted Drug: Future of Neurological Diseases Therapeutics. Curr Neuropharmacol 2023; 21:1042-1064. [PMID: 36411568 PMCID: PMC10286588 DOI: 10.2174/1570159x21666221121095618] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/09/2022] [Accepted: 10/11/2022] [Indexed: 11/23/2022] Open
Abstract
Autism is a neurodevelopmental disorder with a complex etiology that might involve environmental and genetic variables. Recently, some epidemiological studies conducted in various parts of the world have estimated a significant increase in the prevalence of autism, with 1 in every 59 children having some degree of autism. Since autism has been associated with other clinical abnormalities, there is every possibility that a sub-cellular component may be involved in the progression of autism. The organelle remains a focus based on mitochondria's functionality and metabolic role in cells. Furthermore, the mitochondrial genome is inherited maternally and has its DNA and organelle that remain actively involved during embryonic development; these characteristics have linked mitochondrial dysfunction to autism. Although rapid stride has been made in autism research, there are limited studies that have made particular emphasis on mitochondrial dysfunction and autism. Accumulating evidence from studies conducted at cellular and sub-cellular levels has indicated that mitochondrial dysfunction's role in autism is more than expected. The present review has attempted to describe the risk factors of autism, the role of mitochondria in the progression of the disease, oxidative damage as a trigger point to initiate mitochondrial damage, genetic determinants of the disease, possible pathogenic pathways and therapeutic regimen in vogue and the developmental stage. Furthermore, in the present review, an attempt has been made to include the novel therapeutic regimens under investigation at different clinical trial stages and their potential possibility to emerge as promising drugs against ASD.
Collapse
Affiliation(s)
- Showkat Ul Nabi
- Large Animal Diagnostic Laboratory, Department of Clinical Veterinary Medicine, Ethics & Jurisprudence, Faculty of Veterinary Sciences and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology (SKUAST-K), Srinagar J&K, 190006, India
| | - Muneeb U. Rehman
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Azher Arafah
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Syed Taifa
- Large Animal Diagnostic Laboratory, Department of Clinical Veterinary Medicine, Ethics & Jurisprudence, Faculty of Veterinary Sciences and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology (SKUAST-K), Srinagar J&K, 190006, India
| | - Iqra Shafi Khan
- Large Animal Diagnostic Laboratory, Department of Clinical Veterinary Medicine, Ethics & Jurisprudence, Faculty of Veterinary Sciences and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology (SKUAST-K), Srinagar J&K, 190006, India
| | - Andleeb Khan
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan, 45142, Saudi Arabia
| | - Summya Rashid
- Department of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj, 11942, Saudi Arabia
| | - Fatimah Jan
- Department of Pharmaceutical Sciences, CT University, Ludhiana, Ferozepur Road, Punjab, 142024, India
| | - Hilal Ahmad Wani
- Department of Biochemistry, Government Degree College Sumbal, Bandipora, J&K, India
| | - Sheikh Fayaz Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| |
Collapse
|
5
|
The role of maternal immune activation in the immunological and neurological pathogenesis of autism. JOURNAL OF NEURORESTORATOLOGY 2022. [DOI: 10.1016/j.jnrt.2022.100030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
6
|
Tamouza R, Volt F, Richard JR, Wu CL, Bouassida J, Boukouaci W, Lansiaux P, Cappelli B, Scigliuolo GM, Rafii H, Kenzey C, Mezouad E, Naamoune S, Chami L, Lejuste F, Farge D, Gluckman E. Possible Effect of the use of Mesenchymal Stromal Cells in the Treatment of Autism Spectrum Disorders: A Review. Front Cell Dev Biol 2022; 10:809686. [PMID: 35865626 PMCID: PMC9294632 DOI: 10.3389/fcell.2022.809686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 06/13/2022] [Indexed: 11/23/2022] Open
Abstract
Autism spectrum disorder (ASD) represents a set of heterogeneous neurodevelopmental conditions defined by impaired social interactions and repetitive behaviors. The number of reported cases has increased over the past decades, and ASD is now a major public health burden. So far, only treatments to alleviate symptoms are available, with still unmet need for an effective disease treatment to reduce ASD core symptoms. Genetic predisposition alone can only explain a small fraction of the ASD cases. It has been reported that environmental factors interacting with specific inter-individual genetic background may induce immune dysfunctions and contribute to the incidence of ASD. Such dysfunctions can be observed at the central level, with increased microglial cells and activation in ASD brains or in the peripheral blood, as reflected by high circulating levels of pro-inflammatory cytokines, abnormal activation of T-cell subsets, presence of auto-antibodies and of dysregulated microbiota profiles. Altogether, the dysfunction of immune processes may result from immunogenetically-determined inefficient immune responses against a given challenge followed by chronic inflammation and autoimmunity. In this context, immunomodulatory therapies might offer a valid therapeutic option. Mesenchymal stromal cells (MSC) immunoregulatory and immunosuppressive properties constitute a strong rationale for their use to improve ASD clinical symptoms. In vitro studies and pre-clinical models have shown that MSC can induce synapse formation and enhance synaptic function with consequent improvement of ASD-like symptoms in mice. In addition, two preliminary human trials based on the infusion of cord blood-derived MSC showed the safety and tolerability of the procedure in children with ASD and reported promising clinical improvement of core symptoms. We review herein the immune dysfunctions associated with ASD provided, the rationale for using MSC to treat patients with ASD and summarize the current available studies addressing this subject.
Collapse
Affiliation(s)
- Ryad Tamouza
- Translational Neuropsychiatry, INSERM, IMRB, DMU, AP-HP, Univ Paris Est Créteil, Créteil, France
- *Correspondence: Ryad Tamouza,
| | - Fernanda Volt
- Institut de Recherche Saint Louis (IRSL), Eurocord, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
| | - Jean-Romain Richard
- Translational Neuropsychiatry, INSERM, IMRB, Univ Paris Est Créteil, Créteil, France
| | - Ching-Lien Wu
- Translational Neuropsychiatry, INSERM, IMRB, Univ Paris Est Créteil, Créteil, France
| | - Jihène Bouassida
- Translational Neuropsychiatry, INSERM, IMRB, Univ Paris Est Créteil, Créteil, France
| | - Wahid Boukouaci
- Translational Neuropsychiatry, INSERM, IMRB, Univ Paris Est Créteil, Créteil, France
| | - Pauline Lansiaux
- Unité de Médecine Interne (UF 04), CRMR MATHEC, Maladies Auto-immunes et Thérapie Cellulaire, Centre de Référence des Maladies Auto-immunes Systémiques Rares D’Ile-de-France MATHEC, AP-HP, Hôpital St-Louis, Paris, France
| | - Barbara Cappelli
- Institut de Recherche Saint Louis (IRSL), Eurocord, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
- Monacord, Centre Scientifique de Monaco, Monaco, Monaco
| | - Graziana Maria Scigliuolo
- Institut de Recherche Saint Louis (IRSL), Eurocord, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
- Monacord, Centre Scientifique de Monaco, Monaco, Monaco
| | - Hanadi Rafii
- Institut de Recherche Saint Louis (IRSL), Eurocord, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
| | - Chantal Kenzey
- Institut de Recherche Saint Louis (IRSL), Eurocord, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
| | - Esma Mezouad
- Translational Neuropsychiatry, INSERM, IMRB, DMU, AP-HP, Univ Paris Est Créteil, Créteil, France
| | - Soumia Naamoune
- Translational Neuropsychiatry, INSERM, IMRB, DMU, AP-HP, Univ Paris Est Créteil, Créteil, France
| | - Leila Chami
- Translational Neuropsychiatry, INSERM, IMRB, DMU, AP-HP, Univ Paris Est Créteil, Créteil, France
| | - Florian Lejuste
- Translational Neuropsychiatry, INSERM, IMRB, DMU, AP-HP, Univ Paris Est Créteil, Créteil, France
| | - Dominique Farge
- Unité de Médecine Interne (UF 04), CRMR MATHEC, Maladies Auto-immunes et Thérapie Cellulaire, Centre de Référence des Maladies Auto-immunes Systémiques Rares D’Ile-de-France MATHEC, AP-HP, Hôpital St-Louis, Paris, France
| | - Eliane Gluckman
- Institut de Recherche Saint Louis (IRSL), Eurocord, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
- Monacord, Centre Scientifique de Monaco, Monaco, Monaco
| |
Collapse
|
7
|
Han YMY, Yau SY, Chan MMY, Wong CK, Chan AS. Altered Cytokine and BDNF Levels in Individuals with Autism Spectrum Disorders. Brain Sci 2022; 12:brainsci12040460. [PMID: 35447993 PMCID: PMC9026457 DOI: 10.3390/brainsci12040460] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/20/2022] [Accepted: 03/25/2022] [Indexed: 12/10/2022] Open
Abstract
Previous studies have shown that immunological factors are involved in the pathogenesis of autism spectrum disorders (ASDs). The present study examined whether immunological abnormalities are associated with cognitive and behavioral deficits in children with ASD and whether children with ASD show different immunological biomarkers and brain-derived neurotrophic factor BDNF levels than typically developing (TD) children. Sixteen children with TD and 18 children with ASD, aged 6–18 years, voluntarily participated in the study. Participants’ executive functions were measured using neuropsychological tests, and behavioral measures were measured using parent ratings. Immunological measures were assessed by measuring the participants’ blood serum levels of chemokine ligand 2 (CCL2) and chemokine ligand 5 (CCL5). Children with ASD showed greater deficits in cognitive functions as well as altered levels of immunological measures when compared to TD children, and their cognitive functions and behavioral deficits were significantly associated with increased CCL5 levels and decreased BDNF levels. These results provide evidence to support the notion that altered immune functions and neurotrophin deficiency are involved in the pathogenesis of ASD.
Collapse
Affiliation(s)
- Yvonne M. Y. Han
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong SAR, China; (S.-Y.Y.); (M.M.Y.C.)
- Correspondence: ; Tel.: +852-2766-7578
| | - Suk-Yu Yau
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong SAR, China; (S.-Y.Y.); (M.M.Y.C.)
| | - Melody M. Y. Chan
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong SAR, China; (S.-Y.Y.); (M.M.Y.C.)
| | - Chun-Kwok Wong
- Department of Chemical Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China;
| | - Agnes S. Chan
- Department of Psychology, The Chinese University of Hong Kong, Hong Kong SAR, China;
| |
Collapse
|
8
|
Immune Dysregulation in Autism Spectrum Disorder: What Do We Know about It? Int J Mol Sci 2022; 23:ijms23063033. [PMID: 35328471 PMCID: PMC8955336 DOI: 10.3390/ijms23063033] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/03/2022] [Accepted: 03/09/2022] [Indexed: 02/06/2023] Open
Abstract
Autism spectrum disorder (ASD) is a group of complex multifactorial neurodevelopmental disorders characterized by a wide and variable set of neuropsychiatric symptoms, including deficits in social communication, narrow and restricted interests, and repetitive behavior. The immune hypothesis is considered to be a major factor contributing to autism pathogenesis, as well as a way to explain the differences of the clinical phenotypes and comorbidities influencing disease course and severity. Evidence highlights a link between immune dysfunction and behavioral traits in autism from several types of evidence found in both cerebrospinal fluid and peripheral blood and their utility to identify autistic subgroups with specific immunophenotypes; underlying behavioral symptoms are also shown. This review summarizes current insights into immune dysfunction in ASD, with particular reference to the impact of immunological factors related to the maternal influence of autism development; comorbidities influencing autism disease course and severity; and others factors with particular relevance, including obesity. Finally, we described main elements of similarities between immunopathology overlapping neurodevelopmental and neurodegenerative disorders, taking as examples autism and Parkinson Disease, respectively.
Collapse
|
9
|
Increased Monocyte Production of IL-6 after Toll-like Receptor Activation in Children with Autism Spectrum Disorder (ASD) Is Associated with Repetitive and Restricted Behaviors. Brain Sci 2022; 12:brainsci12020220. [PMID: 35203983 PMCID: PMC8870658 DOI: 10.3390/brainsci12020220] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/31/2022] [Accepted: 02/02/2022] [Indexed: 01/27/2023] Open
Abstract
The prevalence of autism spectrum disorder (ASD) has starkly increased, instigating research into risk factors for ASD. This research has identified immune risk factors for ASD, along with evidence of immune dysfunction and excess inflammation frequently experienced by autistic individuals. Increased innate inflammatory cytokines, including interleukin (IL)-6, are seen repeatedly in ASD; however, the origin of excess IL-6 in ASD has not been identified. Here we explore specific responses of circulating monocytes from autistic children. We isolated CD14+ monocytes from whole blood and stimulated them for 24 h under three conditions: media alone, lipoteichoic acid to activate TLR2, and lipopolysaccharide to activate TLR4. We then measured secreted cytokine concentrations in cellular supernatant using a human multiplex bead immunoassay. We found that after TLR4 activation, CD14+ monocytes from autistic children produce increased IL-6 compared to monocytes from children with typical development. IL-6 concentration also correlated with worsening restrictive and repetitive behaviors. These findings suggest dysfunctional activation of myeloid cells, and may indicate that other cells of this lineage, including macrophages, and microglia in the brain, might have a similar dysfunction. Further research on myeloid cells in ASD is warranted.
Collapse
|
10
|
Chen X, Chen Y, Qi D, Cui D. Multifaceted interconnections between macrophage migration inhibitory factor and psychiatric disorders. Prog Neuropsychopharmacol Biol Psychiatry 2022; 112:110422. [PMID: 34358623 DOI: 10.1016/j.pnpbp.2021.110422] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 07/30/2021] [Accepted: 07/31/2021] [Indexed: 01/02/2023]
Abstract
Inflammation is involved in the pathogenesis of psychiatric disorders. Many previous studies have defined the important roles of inflammatory factors in the pathogenesis, diagnosis, and treatment outcomes of psychiatric disorders. Macrophage migration inhibitory factor (MIF), a pro-inflammatory factor, has been gradually recognized to be involved in the development of neurological diseases in recent years. Our current review focuses on discussing the potential beneficial and detrimental roles of MIF in psychiatric disorders. We will provide new mechanistic insights for the development of potential diagnostic and therapeutic biomarkers based on MIF for psychiatric diseases.
Collapse
Affiliation(s)
- Xi Chen
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Psychotic Disorders, Shanghai, China.
| | - Yifan Chen
- Department of Psychology, Tufts University, Medford, MA, USA.
| | - Dake Qi
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.
| | - Donghong Cui
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Psychotic Disorders, Shanghai, China; Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
11
|
Liu W, Li L, Xia X, Zhou X, Du Y, Yin Z, Wang J. Integration of Urine Proteomic and Metabolomic Profiling Reveals Novel Insights Into Neuroinflammation in Autism Spectrum Disorder. Front Psychiatry 2022; 13:780747. [PMID: 35615451 PMCID: PMC9124902 DOI: 10.3389/fpsyt.2022.780747] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
Autism spectrum disorder (ASD) comprises a group of neurodevelopmental disorders whose etiology and pathogenesis are not fully understood. To gain insight into the molecular basis of ASD, we performed comparative integrated proteomic and metabolomic analyses of urine samples from children diagnosed with ASD and healthy children. All 160 samples underwent proteomics analysis and 60 were analyzed by liquid chromatography-mass spectrometry to obtain metabolite profiles. We identified 77 differentially expressed proteins (DEPs; 21 downregulated and 56 upregulated) and 277 differentially expressed metabolites; 31 of the DEPs including glutathione, leukocyte antigens, glycoproteins, neural adhesion factors, and immunoglobulins, have been implicated in neuroinflammation. The proteomic analysis also revealed 8 signaling pathways that were significantly dysregulated in ASD patients; 3 of these (transendothelial leukocyte migration, antigen processing and presentation, and graft vs. host disease) were associated with the neuroimmune response. The metabolism of tryptophan, which is also related to the neuroimmune response, has been found to play a potential role in ASD. Integrated proteome and metabolome analysis showed that 6 signaling pathways were significantly enriched in ASD patients, 3 of which were correlated with impaired neuroinflammation (glutathione metabolism, metabolism of xenobiotics by cytochrome P450 and transendothelial migration of leukocyte). We also found a correlation between prostaglandin (PG) E2 levels and the inflammatory response in ASD. These results underscore the prominent role of the neuroimmune response in ASD and provide potential biomarkers that can be used for diagnosis or as targets for early intervention.
Collapse
Affiliation(s)
- Wenlong Liu
- Department of Child Development and Behavior, School of Medicine, Women and Children's Hospital, Xiamen University, Xiamen, China
| | - Liming Li
- Department of Public Health and Medical Technology, Xiamen Medical College, Xiamen, China
| | - Xiaochun Xia
- Department of Public Health and Medical Technology, Xiamen Medical College, Xiamen, China
| | - Xulan Zhou
- Department of Public Health and Medical Technology, Xiamen Medical College, Xiamen, China
| | - Yukai Du
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhaoqing Yin
- Division of Neonatology, The People's Hospital of Dehong Autonomous Prefecture, Mangshi, China
| | - Juan Wang
- Department of Public Health and Medical Technology, Xiamen Medical College, Xiamen, China
| |
Collapse
|
12
|
Macrophage migration inhibitory factor in Nodding syndrome. PLoS Negl Trop Dis 2021; 15:e0009821. [PMID: 34662363 PMCID: PMC8553141 DOI: 10.1371/journal.pntd.0009821] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 10/28/2021] [Accepted: 09/18/2021] [Indexed: 11/19/2022] Open
Abstract
Nodding syndrome (NS) is a catastrophic and enigmatic childhood epilepsy, accompanied by multiple neurological impairments and neuroinflammation. Of all the infectious, environmental and psychological factors associated with NS, the major culprit is Onchocerca Volvulus (Ov)-a parasitic worm transmitted to human by blackflies. NS seems to be an 'Autoimmune Epilepsy' in light of the recent findings of deleterious autoimmune antibodies to Glutamate receptors and to Leiomodin-I in NS patients. Moreover, we recently found immunogenetic fingerprints in HLA peptide-binding grooves associate with protection or susceptibility to NS. Macrophage migration inhibitory factor (MIF) is an immune-regulatory cytokine playing a central role in modulating innate and adaptive immunity. MIF is also involved in various pathologies: infectious, autoimmune and neurodegenerative diseases, epilepsy and others. Herein, two functional polymorphisms in the MIF gene, a -794 CATT5-8 microsatellite repeat and a -173 G/C single-nucleotide polymorphism, were assessed in 49 NS patients and 51 healthy controls from South Sudan. We also measured MIF plasma levels in established NS patients and healthy controls. We discovered that the frequency of the high-expression MIF -173C containing genotype was significantly lower in NS patients compared to healthy controls. Interestingly however, MIF plasma levels were significantly elevated in NS patients than in healthy controls. We further demonstrated that the HLA protective and susceptibility associations are dominant over the MIF association with NS. Our findings suggest that MIF might have a dual role in NS. Genetically controlled high-expression MIF genotype is associated with disease protection. However, elevated MIF in the plasma may contribute to the detrimental autoimmunity, neuroinflammation and epilepsy.
Collapse
|
13
|
The role of microbiota-gut-brain axis in neuropsychiatric and neurological disorders. Pharmacol Res 2021; 172:105840. [PMID: 34450312 DOI: 10.1016/j.phrs.2021.105840] [Citation(s) in RCA: 353] [Impact Index Per Article: 88.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 08/14/2021] [Accepted: 08/17/2021] [Indexed: 12/12/2022]
Abstract
Emerging evidence indicates that the gut microbiota play a crucial role in the bidirectional communication between the gut and the brain suggesting that the gut microbes may shape neural development, modulate neurotransmission and affect behavior, and thereby contribute to the pathogenesis and/or progression of many neurodevelopmental, neuropsychiatric, and neurological conditions. This review summarizes recent data on the role of microbiota-gut-brain axis in the pathophysiology of neuropsychiatric and neurological disorders including depression, anxiety, schizophrenia, autism spectrum disorders, Parkinson's disease, migraine, and epilepsy. Also, the involvement of microbiota in gut disorders co-existing with neuropsychiatric conditions is highlighted. We discuss data from both in vivo preclinical experiments and clinical reports including: (1) studies in germ-free animals, (2) studies exploring the gut microbiota composition in animal models of diseases or in humans, (3) studies evaluating the effects of probiotic, prebiotic or antibiotic treatment as well as (4) the effects of fecal microbiota transplantation.
Collapse
|
14
|
Zhao H, Zhang H, Liu S, Luo W, Jiang Y, Gao J. Association of Peripheral Blood Levels of Cytokines With Autism Spectrum Disorder: A Meta-Analysis. Front Psychiatry 2021; 12:670200. [PMID: 34276441 PMCID: PMC8283413 DOI: 10.3389/fpsyt.2021.670200] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 05/27/2021] [Indexed: 01/03/2023] Open
Abstract
Background: Although increasing evidence suggests an association between alterations in peripheral cytokines and autism spectrum disorder (ASD), a consensus is lacking. To determine whether abnormal cytokine profiles in peripheral blood were associated with ASD, we performed this systemic review and meta-analysis. Methods: A systematic literature search was conducted through the Embase, PubMed, Web of Knowledge, PsycINFO, and Cochrane databases up to 4 June 2020. Clinical studies exploring the aberration of peripheral cytokines of autistic patients and controls were included in our meta-analysis. We pooled extracted data using fixed- or random-effects models based on heterogeneity tests with Comprehensive Meta-analysis software. We converted standardized mean differences to Hedges' g statistic to obtain the effect sizes adjusted for sample size. Subgroup analyses, sensitivity analyses, meta-regression, and publication bias tests were also carried out. Results: Sixty-one articles (326 studies) were included to assess the association between 76 cytokines and ASD. We conducted our meta-analysis based on 37 cytokines with 289 studies. Since there were fewer than three studies on any of the other 39 cytokines, we only provided basic information for them. The levels of peripheral IL-6, IL-1β, IL-12p70, macrophage migration inhibitory factor (MIF), eotaxin-1, monocyte chemotactic protein-1 (MCP-1), IL-8, IL-7, IL-2, IL-12, tumor necrosis factor-α (TNF-α), IL-17, and IL-4 were defined as abnormal cytokines in the peripheral blood of ASD patients compared with controls. The other 24 cytokines did not obviously change in ASD patients compared with the controls. Conclusions: The findings of our meta-analysis strengthen the evidence for an abnormal cytokine profile in ASD. These abnormal cytokines may be potential biomarkers for the diagnosis and treatment of ASD in the future.
Collapse
Affiliation(s)
- Huaying Zhao
- Department of Rehabilitation Medicine, Fuling Central Hospital of Chongqing City, Chongqing, China
| | - Hongqi Zhang
- Department of Pulmonary and Critical Care Medicine, Fuling Central Hospital of Chongqing City, Chongqing, China
| | - Shijie Liu
- The 947th Hospital of Army, Kashi, China
| | - Wulin Luo
- Department of Medical Psychology and Neurology, The 947th Hospital of Army, Kashi, China
| | - Yongfeng Jiang
- Department of Rehabilitation Medicine, The 947th Hospital of Army, Kashi, China
| | - Junwei Gao
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
15
|
Cyclooxygenase Inhibition Safety and Efficacy in Inflammation-Based Psychiatric Disorders. Molecules 2020; 25:molecules25225388. [PMID: 33217958 PMCID: PMC7698629 DOI: 10.3390/molecules25225388] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 12/21/2022] Open
Abstract
According to the World Health Organization, the major psychiatric and neurodevelopmental disorders include major depression, bipolar disorder, schizophrenia, and autism spectrum disorder. The potential role of inflammation in the onset and progression of these disorders is increasingly being studied. The use of non-steroidal anti-inflammatory drugs (NSAIDs), well-known cyclooxygenase (COX) inhibitors, combined with first-choice specific drugs have been long investigated. The adjunctive administration of COX inhibitors to classic clinical treatments seems to improve the prognosis of people who suffer from psychiatric disorders. In this review, a broad overview of the use of COX inhibitors in the treatment of inflammation-based psychiatric disorders is provided. For this purpose, a critical analysis of the use of COX inhibitors in the last ten years of clinical trials of the major psychiatric disorders was carried out.
Collapse
|
16
|
Chai X, Zhang W, Li L, Wu Y, Zhu X, Zhao S. Profile of MIF in Developing Hippocampus: Association With Cell Proliferation and Neurite Outgrowth. Front Mol Neurosci 2020; 13:147. [PMID: 32903462 PMCID: PMC7434973 DOI: 10.3389/fnmol.2020.00147] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 07/15/2020] [Indexed: 11/13/2022] Open
Abstract
Proinflammatory cytokine macrophage migration inhibitory factor (MIF) is a multifunctional cytokine and has been found involved in many neurological diseases such as Alzheimer disease (AD), epilepsy, and multiple sclerosis. Previous studies have shown that MIF is expressed in neocortex, hippocampus, hypothalamus, cerebellum, and spinal cord in adult mice. It is expressed by astrocytes and activates microglias in neuroinflammation. Further studies have shown that MIF is detected in moss fibers of dentate granule cells and in apical dendrites of pyramidal neurons in adult hippocampus. Only NeuroD-positive immature granule neurons but not NeuN-positive mature neurons express MIF. These findings led us eager to know the exact role of MIF in the development of hippocampus. Therefore, we systematically checked the spatial and temporal expression pattern of MIF and characterized MIF-positive cells in hippocampus from mice aged from postnatal day 0 (P0) to 3 months. Our results showed that the lowest level of MIF protein occurred at P7 and mif mRNA increased from P0, reached a peak at P7, and stably expressed until P30 before declining dramatically at 3 months. MIF was localized in fibers of GFAP- and BLBP-positive radial glial precursor cells in dentate gyrus (DG). DCX-expressing newly generated neurons were MIF-negative. Inhibition of MIF by MIF antagonist S, R-3-(4-hydroxyphenyl)-4, 5-dihydro-5-isoxazole acetic acid methyl ester (ISO-1) reduced BrdU-positive cells. Interestingly, MIF was expressed by NeuN-positive GABAergic interneurons including parvalbumin-and Reelin-expressing cells in the DG. Neither NeuN-positive granule cells nor NeuN-positive pyramidal neurons expressed MIF. In transgenic mice, POMC-EGFP–positive immature dentate granule cells and Thy1-EGFP–positive mature granule cells were MIF-negative. Treatment of neuronal cultures with ISO-1 inhibited neurite outgrowth. Therefore, we conclude that MIF might be important for feature maintenance of neural stem cells and neurite outgrowth during hippocampal development.
Collapse
Affiliation(s)
- Xuejun Chai
- College of Basic Medicine, Xi'an Medical University, Xi'an, China
| | - Wei Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Lingling Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Yongji Wu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Xiaoyan Zhu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Shanting Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| |
Collapse
|
17
|
Oh D, Cheon KA. Alteration of Gut Microbiota in Autism Spectrum Disorder: An Overview. Soa Chongsonyon Chongsin Uihak 2020; 31:131-145. [PMID: 32665757 PMCID: PMC7350540 DOI: 10.5765/jkacap.190039] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/25/2020] [Accepted: 04/16/2020] [Indexed: 02/06/2023] Open
Abstract
The microbiota-gut-brain axis, which refers to the bidirectional communication pathway between gut bacteria and the central nervous system, has a profound effect on important brain processes, from the synthesis of neurotransmitters to the modulation of complex behaviors such as sociability and anxiety. Previous studies have revealed that the gut microbiota is potentially related to not only gastrointestinal disturbances, but also social impairment and repetitive behavior-core symptoms of autism spectrum disorder (ASD). Although studies have been conducted to characterize the microbial composition in patients with ASD, the results are heterogeneous. Nevertheless, it is clear that there is a difference in the composition of the gut microbiota between ASD and typically developed individuals, and animal studies have repeatedly suggested that the gut microbiota plays an important role in ASD pathophysiology. This possibility is supported by abnormalities in metabolites produced by the gut microbiota and the association between altered immune responses and the gut microbiota observed in ASD patients. Based on these findings, various attempts have been made to use the microbiota in ASD treatment. The results reported to date suggest that microbiota-based therapies may be effective for ASD, but largescale, well-designed studies are needed to confirm this.
Collapse
Affiliation(s)
- Donghun Oh
- Department of Psychiatry, Yonsei University College of Medicine, Seoul, Korea.,Division of Child and Adolescent Psychiatry, Severance Children's Hospital, Seoul, Korea.,Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Keun-Ah Cheon
- Department of Psychiatry, Yonsei University College of Medicine, Seoul, Korea.,Division of Child and Adolescent Psychiatry, Severance Children's Hospital, Seoul, Korea.,Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
18
|
Kim M, Chung SK, Yang JC, Park JI, Nam SH, Park TW. Development of the Korean Form of the Premonitory Urge for Tics Scale: A Reliability and Validity Study. Soa Chongsonyon Chongsin Uihak 2020; 31:146-153. [PMID: 32665758 PMCID: PMC7350545 DOI: 10.5765/jkacap.200013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 03/30/2020] [Accepted: 04/16/2020] [Indexed: 12/26/2022] Open
Abstract
Objectives This study aimed to evaluate the reliability and validity of the Korean Form of the Premonitory Urge for Tics Scale (K-PUTS). Methods Thirty-eight patients with Tourette's disorder who visited Jeonbuk National University Hospital were assessed with the K-PUTS. Together with the PUTS, the Yale Global Tic Severity Scale (YGTSS), the Children's Yale-Brown Obsessive Compulsive Scale (CY-BOCS), the attention-deficit/hyperactivity disorder (ADHD) rating scale (ARS), and the Adult ADHD Self-Report Scale (ASRS) were implemented to evaluate concurrent and discriminant validity. Results The internal consistency of items on the PUTS was high, with a Cronbach's α of 0.79. The test-retest reliability of the PUTS, which was administered at 2 weeks to 2 months intervals, showed high reliability with a Pearson correlation coefficient of 0.60. There was a significant positive correlation between the overall PUTS score and the YGTSS score, showing concurrent validity. There was no correlation between the PUTS, CY-BOCS, and ASRS scores, demonstrating the discriminant validity of the PUTS. Factor analysis for construct validity revealed three factors: "presumed functional relationship between the tic and the urge to tic," "the quality of the premonitory urge," and "just right phenomena." Conclusion The results of this study indicate that the K-PUTS is a reliable and valid scale for rating premonitory urge of tics.
Collapse
Affiliation(s)
- Mira Kim
- Department of Psychiatry, Jeonbuk National University Hospital, Jeonju, Korea
| | - Sang-Keun Chung
- Department of Psychiatry, Jeonbuk National University Hospital, Jeonju, Korea.,Department of Psychiatry, Jeonbuk National University Medical School, Jeonju, Korea
| | - Jong-Chul Yang
- Department of Psychiatry, Jeonbuk National University Hospital, Jeonju, Korea.,Department of Psychiatry, Jeonbuk National University Medical School, Jeonju, Korea
| | - Jong-Il Park
- Department of Psychiatry, Jeonbuk National University Hospital, Jeonju, Korea.,Department of Psychiatry, Jeonbuk National University Medical School, Jeonju, Korea
| | - Seok Hyun Nam
- Department of Psychiatry, Jeonbuk National University Hospital, Jeonju, Korea
| | - Tae Won Park
- Department of Psychiatry, Jeonbuk National University Hospital, Jeonju, Korea.,Department of Psychiatry, Jeonbuk National University Medical School, Jeonju, Korea
| |
Collapse
|
19
|
Sala R, Amet L, Blagojevic-Stokic N, Shattock P, Whiteley P. Bridging the Gap Between Physical Health and Autism Spectrum Disorder. Neuropsychiatr Dis Treat 2020; 16:1605-1618. [PMID: 32636630 PMCID: PMC7335278 DOI: 10.2147/ndt.s251394] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/05/2020] [Indexed: 12/12/2022] Open
Abstract
Autism spectrum disorder (ASD) is a highly complex and heterogeneous developmental disorder that affects how individuals communicate with other people and relate to the world around them. Research and clinical focus on the behavioural and cognitive manifestations of ASD, whilst important, have obscured the recognition that ASD is also commonly associated with a range of physical and mental health conditions. Many physical conditions appear with greater frequency in individuals with ASD compared to non-ASD populations. These can contribute to a worsening of social communication and behaviour, lower quality of life, higher morbidity and premature mortality. We highlight some of the key physical comorbidities affecting the immune and the gastrointestinal systems, metabolism and brain function in ASD. We discuss how healthcare professionals working with individuals with ASD and parents/carers have a duty to recognise their needs in order to improve their overall health and wellbeing, deliver equality in their healthcare experiences and reduce the likelihood of morbidity and early mortality associated with the condition.
Collapse
Affiliation(s)
- Regina Sala
- Centre for Psychiatry, Wolfson Institute, Barts & The London School of Medicine & Dentistry Queen Mary University of London, London, UK
| | | | | | - Paul Shattock
- Education & Services for People with Autism, Sunderland, UK
| | - Paul Whiteley
- Education & Services for People with Autism Research, Sunderland, UK
| |
Collapse
|
20
|
Leng L, Siu E, Bucala R. Genotyping Two Promoter Polymorphisms in the MIF Gene: A -794 CATT 5-8 Microsatellite Repeat and a -173 G/C SNP. Methods Mol Biol 2020; 2080:67-84. [PMID: 31745872 DOI: 10.1007/978-1-4939-9936-1_7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Macrophage migration inhibitory factor (MIF) is an upstream proinflammatory cytokine encoded by a functionally polymorphic locus. The promoter region of the human MIF gene contains two polymorphisms. A variable nucleotide tandem repeat at position -794 comprises five to eight CATT repeats (referred to henceforth by numbers from 5 to 8, rs5844572). Gene reporter assays show a proportional increase in transcription with CATT repeat number; the 5-repeat allele leads to low expression, and the 6-, 7-, and 8-repeat alleles lead to correspondingly higher expression of MIF. A second MIF promoter polymorphism comprises a G-to-C single nucleotide polymorphism (SNP) at position -173 (rs755622), which is in strong linkage disequilibrium with -794 7-CATT and is associated with arthritis clinical severity and higher serum and synovial fluid MIF levels. This allele also has been reported to confer improved survival in patients with outpatient pneumonia. In this chapter, we will introduce the methods of genotyping CATT5-8 repeats and the MIF -173 G/C from human samples.
Collapse
Affiliation(s)
- Lin Leng
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Edwin Siu
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Richard Bucala
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA.
| |
Collapse
|
21
|
Hodges SL, Nolan SO, Tomac LA, Muhammad IDA, Binder MS, Taube JH, Lugo JN. Lipopolysaccharide-induced inflammation leads to acute elevations in pro-inflammatory cytokine expression in a mouse model of Fragile X syndrome. Physiol Behav 2019; 215:112776. [PMID: 31838149 DOI: 10.1016/j.physbeh.2019.112776] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/11/2019] [Accepted: 12/11/2019] [Indexed: 02/07/2023]
Abstract
Fragile X syndrome (FXS) is a neurodevelopmental disorder caused by a single genetic mutation in the Fmr1 gene, serving as the largest genetic cause of intellectual disability. Trinucleotide expansion mutations in Fmr1 result in silencing and hypermethylation of the gene, preventing synthesis of the RNA binding protein Fragile X mental retardation protein which functions as a translational repressor. Abnormal immune responses have been demonstrated to play a role in FXS pathophysiology, however, whether these alterations impact how those with FXS respond to an immune insult behaviorally is not entirely known. In the current study, we examine how Fmr1 knockout (KO) and wild type (WT) mice respond to the innate immune stimulus lipopolysaccharide (LPS), both on a molecular and behavioral level, to determine if Fmr1 mutations impact the normal physiological response to an immune insult. In response to LPS, Fmr1 KO mice had elevated hippocampal IL-1β and IL-6 mRNA levels 4 h post-treatment compared to WT mice, with no differences detected in any cytokines at baseline or between genotypes 24 h post-LPS administration. Fmr1 KO mice also had upregulated hippocampal BDNF gene expression 4 h post-treatment compared to WT mice, which was not dependent on LPS administration. There were no differences in hippocampal protein expression between genotypes in microglia (Iba1) or astrocyte (GFAP) reactivity. Further, both genotypes displayed the typical sickness response following LPS stimulation, demonstrated by a significant reduction in food burrowed by LPS-treated mice in a burrowing task. Additional investigation is critical to determine if the transient increases in cytokine expression could lead to long-term changes in downstream molecular signaling in FXS.
Collapse
Affiliation(s)
- Samantha L Hodges
- Institute of Biomedical Studies, Baylor University, Waco, TX 76798, USA
| | - Suzanne O Nolan
- Department of Psychology and Neuroscience, Baylor University, One Bear Place # 97334, Waco, TX 76798, USA
| | - Lindsay A Tomac
- Department of Psychology and Neuroscience, Baylor University, One Bear Place # 97334, Waco, TX 76798, USA
| | - Ilyasah D A Muhammad
- Department of Psychology and Neuroscience, Baylor University, One Bear Place # 97334, Waco, TX 76798, USA
| | - Matthew S Binder
- Department of Psychology and Neuroscience, Baylor University, One Bear Place # 97334, Waco, TX 76798, USA
| | - Joseph H Taube
- Institute of Biomedical Studies, Baylor University, Waco, TX 76798, USA; Department of Biology, Baylor University, Waco, TX 76798, USA
| | - Joaquin N Lugo
- Institute of Biomedical Studies, Baylor University, Waco, TX 76798, USA; Department of Psychology and Neuroscience, Baylor University, One Bear Place # 97334, Waco, TX 76798, USA; Department of Biology, Baylor University, Waco, TX 76798, USA.
| |
Collapse
|
22
|
Nudel R, Benros ME, Krebs MD, Allesøe RL, Lemvigh CK, Bybjerg-Grauholm J, Børglum AD, Daly MJ, Nordentoft M, Mors O, Hougaard DM, Mortensen PB, Buil A, Werge T, Rasmussen S, Thompson WK. Immunity and mental illness: findings from a Danish population-based immunogenetic study of seven psychiatric and neurodevelopmental disorders. Eur J Hum Genet 2019; 27:1445-1455. [PMID: 30976114 PMCID: PMC6777475 DOI: 10.1038/s41431-019-0402-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 03/11/2019] [Accepted: 03/19/2019] [Indexed: 01/11/2023] Open
Abstract
Human leukocyte antigen (HLA) genes encode proteins with important roles in the regulation of the immune system. Many studies have also implicated HLA genes in psychiatric and neurodevelopmental disorders. However, these studies usually focus on one disorder and/or on one HLA candidate gene, often with small samples. Here, we access a large dataset of 65,534 genotyped individuals consisting of controls (N = 19,645) and cases having one or more of autism spectrum disorder (N = 12,331), attention deficit hyperactivity disorder (N = 14,397), schizophrenia (N = 2401), bipolar disorder (N = 1391), depression (N = 18,511), anorexia (N = 2551) or intellectual disability (N = 3175). We imputed participants' HLA alleles to investigate the involvement of HLA genes in these disorders using regression models. We found a pronounced protective effect of DPB1*1501 on susceptibility to autism (p = 0.0094, OR = 0.72) and intellectual disability (p = 0.00099, OR = 0.41), with an increased protective effect on a comorbid diagnosis of both disorders (p = 0.003, OR = 0.29). We also identified a risk allele for intellectual disability, B*5701 (p = 0.00016, OR = 1.33). Associations with both alleles survived FDR correction and a permutation procedure. We did not find significant evidence for replication of previously-reported associations for autism or schizophrenia. Our results support an implication of HLA genes in autism and intellectual disability, which requires replication by other studies. Our study also highlights the importance of large sample sizes in HLA association studies.
Collapse
Affiliation(s)
- Ron Nudel
- Institute of Biological Psychiatry, Mental Health Centre Sct. Hans, Mental Health Services Copenhagen, Roskilde, Denmark
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen, Denmark
| | - Michael E Benros
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen, Denmark
- Mental Health Centre Copenhagen, University of Copenhagen Hospital, Copenhagen, Denmark
| | - Morten Dybdahl Krebs
- Institute of Biological Psychiatry, Mental Health Centre Sct. Hans, Mental Health Services Copenhagen, Roskilde, Denmark
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen, Denmark
| | - Rosa Lundbye Allesøe
- Department of Bio and Health Informatics, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Camilla Koldbæk Lemvigh
- Department of Bio and Health Informatics, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Jonas Bybjerg-Grauholm
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen, Denmark
- Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Anders D Børglum
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen, Denmark
- Department of Biomedicine, Aarhus University and Centre for Integrative Sequencing, iSEQ, Aarhus, Denmark
- Aarhus Genome Center, Aarhus, Denmark
| | - Mark J Daly
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Merete Nordentoft
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen, Denmark
- Mental Health Centre Copenhagen, University of Copenhagen Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ole Mors
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen, Denmark
- Psychosis Research Unit, Aarhus University Hospital, Risskov, Denmark
| | - David M Hougaard
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen, Denmark
- Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Preben Bo Mortensen
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen, Denmark
- National Center for Register-Based Research, Aarhus University, Aarhus, Denmark
| | - Alfonso Buil
- Institute of Biological Psychiatry, Mental Health Centre Sct. Hans, Mental Health Services Copenhagen, Roskilde, Denmark
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen, Denmark
| | - Thomas Werge
- Institute of Biological Psychiatry, Mental Health Centre Sct. Hans, Mental Health Services Copenhagen, Roskilde, Denmark
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Simon Rasmussen
- Department of Bio and Health Informatics, Technical University of Denmark, Kongens Lyngby, Denmark.
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Wesley K Thompson
- Institute of Biological Psychiatry, Mental Health Centre Sct. Hans, Mental Health Services Copenhagen, Roskilde, Denmark.
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen, Denmark.
- Department of Family Medicine and Public Health, Division of Biostatistics, University of California, San Diego, CA, USA.
| |
Collapse
|
23
|
Arroyo-López C. Helminth therapy for autism under gut-brain axis- hypothesis. Med Hypotheses 2019; 125:110-118. [PMID: 30902137 DOI: 10.1016/j.mehy.2019.02.042] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 02/18/2019] [Indexed: 12/20/2022]
Abstract
Autism is a neurodevelopmental disease included within Autism Syndrome Disorder (ASD) spectrum. ASD has been linked to a series of genes that play a role in immune response function and patients with autism, commonly suffer from immune-related comorbidities. Despite the complex pathophysiology of autism, Gut-brain axis is gaining strength in the understanding of several neurological disorders. In addition, recent publications have shown the correlation between immune dysfunctions, gut microbiota and brain with the behavioral alterations and comorbid symptoms found in autism. Gut-brain axis acts as the "second brain", in a communication network established between neural, endocrine and the immunological systems. On the other hand, Hygiene Hypothesis suggests that the increase in the incidence of autoimmune diseases in the modern world can be attributed to the decrease of exposure to infectious agents, as parasitic nematodes. Helminths induce modulatory and protective effects against several inflammatory disorders, maintaining gastrointestinal homeostasis and modulating brain functions. Helminthic therapy has been previously performed in diseases such as ulcerative colitis, Crohn's disease, diabetes, multiple sclerosis, asthma, rheumatoid arthritis, and food allergies. Considering gut-brain axis, Hygiene Hypothesis, and the modulatory effects of helminths I hypothesized that a treatment with Trichuris suis soluble products represents a feasible holistic treatment for autism, and the key for the development of novel treatments. Preclinical studies are required to test this hypothesis.
Collapse
Affiliation(s)
- Celia Arroyo-López
- Department of Pathology and Laboratory Medicine, UC Davis School of Medicine; Institute for Pediatric Regenerative Medicine and Shriners Hospitals for Children of Northern California, United States.
| |
Collapse
|
24
|
Sethi R, Gómez-Coronado N, Walker AJ, Robertson OD, Agustini B, Berk M, Dodd S. Neurobiology and Therapeutic Potential of Cyclooxygenase-2 (COX-2) Inhibitors for Inflammation in Neuropsychiatric Disorders. Front Psychiatry 2019; 10:605. [PMID: 31551825 PMCID: PMC6738329 DOI: 10.3389/fpsyt.2019.00605] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 07/30/2019] [Indexed: 12/15/2022] Open
Abstract
Neuropsychiatric disorders, such as depression, bipolar disorder, schizophrenia, obsessive-compulsive disorder, and neurodevelopmental disorders such as autism spectrum disorder, are associated with significant illness burden. Accumulating evidence supports an association between these disorders and inflammation. Consequently, anti-inflammatory agents, such as the cyclooxygenase-2 inhibitors, represent a novel avenue to prevent and treat neuropsychiatric illness. In this paper, we first review the role of inflammation in psychiatric pathophysiology including inflammatory cytokines' influence on neurotransmitters, the hypothalamic-pituitary-adrenal axis, and microglial mechanisms. We then discuss how cyclooxygenase-2-inhibitors influence these pathways with potential therapeutic benefit, with a focus on celecoxib, due to its superior safety profile. A search was conducted in PubMed, Embase, and PsychINFO databases, in addition to Clinicaltrials.gov and the Stanley Medical Research Institute trial registries. The results were presented as a narrative review. Currently available outcomes for randomized controlled trials up to November 2017 are also discussed. The evidence reviewed here suggests cyclooxygenase-2 inhibitors, and in particular celecoxib, may indeed assist in treating the symptoms of neuropsychiatric disorders; however, further studies are required to assess appropriate illness stage-related indication.
Collapse
Affiliation(s)
- Rickinder Sethi
- Department of Psychiatry, Western University, London, ON, Canada
| | - Nieves Gómez-Coronado
- Unidad de Gestión Clinica Salud Mental, Hospital Universitario Virgen del Rocio, Sevilla, Spain
| | - Adam J Walker
- IMPACT Strategic Research Centre, Deakin University, Geelong, VIC, Australia
| | - Oliver D'Arcy Robertson
- IMPACT Strategic Research Centre, Deakin University, Geelong, VIC, Australia.,University Hospital Geelong, Barwon Health, Geelong, VIC, Australia
| | - Bruno Agustini
- IMPACT Strategic Research Centre, Deakin University, Geelong, VIC, Australia
| | - Michael Berk
- IMPACT Strategic Research Centre, Deakin University, Geelong, VIC, Australia.,University Hospital Geelong, Barwon Health, Geelong, VIC, Australia.,Department of Psychiatry, The University of Melbourne, Parkville, VIC, Australia.,Orygen, The National Centre of Excellence in Youth Mental Health, Parkville, VIC, Australia.,Department of Psychiatry, Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Seetal Dodd
- IMPACT Strategic Research Centre, Deakin University, Geelong, VIC, Australia.,University Hospital Geelong, Barwon Health, Geelong, VIC, Australia.,Department of Psychiatry, The University of Melbourne, Parkville, VIC, Australia.,Orygen, The National Centre of Excellence in Youth Mental Health, Parkville, VIC, Australia
| |
Collapse
|
25
|
Shin MS, Kang Y, Wahl ER, Park HJ, Lazova R, Leng L, Mamula M, Krishnaswamy S, Bucala R, Kang I. Macrophage Migration Inhibitory Factor Regulates U1 Small Nuclear RNP Immune Complex-Mediated Activation of the NLRP3 Inflammasome. Arthritis Rheumatol 2018; 71:109-120. [PMID: 30009530 DOI: 10.1002/art.40672] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 07/12/2018] [Indexed: 12/15/2022]
Abstract
OBJECTIVE High-expression alleles of macrophage migration inhibitory factor (MIF) are linked genetically to the severity of systemic lupus erythematosus (SLE). The U1 small nuclear RNP (snRNP) immune complex containing U1 snRNP and anti-U1 snRNP antibodies, which are found in patients with SLE, activates the NLRP3 inflammasome, comprising NLRP3, ASC, and procaspase 1, in human monocytes, leading to the production of interleukin-1β (IL-1β). This study was undertaken to investigate the role of the snRNP immune complex in up-regulating the expression of MIF and its interface with the NLRP3 inflammasome. METHODS MIF, IL-1β, NLRP3, caspase 1, ASC, and MIF receptors were analyzed by enzyme-linked immunosorbent assay, Western blotting, quantitative polymerase chain reaction, and cytometry by time-of-flight mass spectrometry (CytoF) in human monocytes incubated with or without the snRNP immune complex. MIF pathway responses were probed with the novel small molecule antagonist MIF098. RESULTS The snRNP immune complex induced the production of MIF and IL-1β from human monocytes. High-dimensional, single-cell CytoF analysis established that MIF regulates activation of the NLRP3 inflammasome, including findings of a quantitative relationship between MIF and its receptors and IL-1β levels in the monocytes. MIF098, which blocks MIF binding to its cognate receptor, suppressed the production of IL-1β, the up-regulation of NLRP3, which is a rate-limiting step in NLRP3 inflammasome activation, and the activation of caspase 1 in snRNP immune complex-stimulated human monocytes. CONCLUSION The U1 snRNP immune complex is a specific stimulus of MIF production in human monocytes, with MIF having an upstream role in defining the inflammatory characteristics of activated monocytes by regulating NLRP3 inflammasome activation and downstream IL-1β production. These findings provide mechanistic insight and a therapeutic rationale for targeting MIF in subgroups of lupus patients, such as those classified as high genotypic MIF expressers or those with anti-snRNP antibodies.
Collapse
Affiliation(s)
- Min Sun Shin
- Yale University School of Medicine, New Haven, Connecticut
| | - Youna Kang
- Yale University School of Medicine, New Haven, Connecticut
| | - Elizabeth R Wahl
- Yale University School of Medicine, New Haven, Connecticut, and University of Washington, Seattle
| | - Hong-Jai Park
- Yale University School of Medicine, New Haven, Connecticut
| | - Rossitza Lazova
- Yale University School of Medicine, New Haven, Connecticut, and California Skin Institute, San Jose
| | - Lin Leng
- Yale University School of Medicine, New Haven, Connecticut
| | - Mark Mamula
- Yale University School of Medicine, New Haven, Connecticut
| | | | - Richard Bucala
- Yale University School of Medicine, New Haven, Connecticut
| | - Insoo Kang
- Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
26
|
Ning J, Xu L, Shen CQ, Zhang YY, Zhao Q. Increased serum levels of macrophage migration inhibitory factor in autism spectrum disorders. Neurotoxicology 2018; 71:1-5. [PMID: 30503813 DOI: 10.1016/j.neuro.2018.11.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 08/30/2018] [Accepted: 11/28/2018] [Indexed: 01/02/2023]
Abstract
OBJECTIVE Macrophage migration inhibitory factor (MIF) has been suggested as a pivotal regulator of innate immunity and inflammatory. The aim of this study was to measure serum circulating levels of MIF in relation to the degree of the severity of autism spectrum disorders (ASD). METHODS One hundred and two Chinese children with ASD and same their age-sex matched typical development children were included. Concentrations of MIF were tested by Quantikine Human MIF Immunoassay. Serum levels of homocysteine (HCY), C-reactive protein (CRP) and serum Interleukin 6 (IL-6) were also tested. The influence of serum levels of MIF on ASD risk and ASD severity were performed by binary logistic regression analysis. RESULTS The serum levels of MIF in the children with ASD (24.7 ± 08.9 ng/ml) were significantly higher than those of control subjects (18.3 ± 5.5 ng/ml) (t = 6.134, P < 0.001). Levels of MIF increased with increasing severity of ASD as defined by the CARS score (P < 0.001). In multivariate model, MIF was associated with an increased risk of ASD (OR 1.11, 95% CI: 1.05-1.17; P < 0.001). MIF improved the combined model (HCY/CRP/IL-6) to predict ASD (P < 0.001). At admission, 68 children (66.7%) had a severe autism. In these children, the mean serum level of MIF was higher than in those children with mild to moderate autism (28.1 ± 8.5 ng/ml VS. 17.9 ± 4.7 ng/ml; t = 6.482, P < 0.001). In multivariate model, MIF was still associated with an increased risk of severe ASD (OR: 1.15, 95% CI: 1.04-1.19; P < 0.001). MIF improved the combined model (HCY/CRP/IL-6) to predict severe ASD (P < 0.001). CONCLUSIONS These results identify high serum MIF levels are associated with severity of ASD. Further study is warranted on the precise involvement of MIF in ASD, and the mechanism by which MIF contributes to ASD pathogenesis.
Collapse
Affiliation(s)
- Jun Ning
- Department of pediatrics, Affiliated Hospital of Jining Medical University, Jining, China
| | - Li Xu
- Department of pediatrics, Affiliated Hospital of Jining Medical University, Jining, China
| | - Chang-Qing Shen
- Department of pediatrics, Affiliated Hospital of Jining Medical University, Jining, China.
| | - Yu-Yan Zhang
- Department of pediatrics, Affiliated Hospital of Jining Medical University, Jining, China
| | - Qing Zhao
- Department of pediatrics, Affiliated Hospital of Jining Medical University, Jining, China
| |
Collapse
|
27
|
Sreih AG, Ezzedine R, Leng L, Fan J, Yao J, Reid D, Piecychna M, Carette S, Cuthbertson D, Dellaripa P, Hoffman GS, Khalidi NA, Koening CL, Langford CA, Mahr A, McAlear CA, Maksimowicz-Mckinnon K, Monach PA, Seo P, Specks U, St Clair EW, Stone JH, Ytterberg SR, Edberg J, Merkel PA, Bucala R. Role of Macrophage Migration Inhibitory Factor in Granulomatosis With Polyangiitis. Arthritis Rheumatol 2018; 70:2077-2086. [PMID: 29953750 DOI: 10.1002/art.40655] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 06/26/2018] [Indexed: 12/17/2022]
Abstract
OBJECTIVE To examine the association between macrophage migration inhibitory factor (MIF) promoter polymorphisms and granulomatosis with polyangiitis (GPA) in human subjects, and to assess the role of MIF in a murine model of granulomatous vasculitis. METHODS The human study involved 1,077 patients with GPA and healthy controls whose serum was genotyped by capillary electrophoresis for the MIF -794 CATT5-8 promoter microsatellite (rs5844572). MIF promoter, CATT-length-dependent gene expression in response to β-glucan was assessed by gene reporter assays. In mouse studies, granulomatous disease was induced by injection of Candida albicans β-glucan into wild-type (WT) or Mif-knockout (Mif-KO) C57BL/6 mice and C57BL/6 mice transgenically overexpressing Mif in lung epithelium (Mif lung-Tg2.1). Mice were treated with a neutralizing anti-MIF antibody and analyzed for the density of pulmonary granulomas, expression of inflammatory chemokines, and frequency of mortality. RESULTS The percentage of human subjects carrying >5 CATT repeats in each MIF allele (high genotypic MIF expressers) was 60.2% among patients with GPA and 53.9% among healthy controls (adjusted P = 0.049). In response to granulomatous stimulation, human MIF gene expression increased proportionally with CATT length. Mif lung-Tg2.1 mice exhibited more pulmonary granulomas than WT mice, which in turn showed more granulomas than Mif-KO mice. A significantly higher percentage of Mif lung-Tg2.1 mice, compared to Mif-KO or WT mice, died when injected with Candida albicans β-glucan, and treatment of these mice with an anti-MIF monoclonal antibody protected against a lethal outcome. Levels of MIF-dependent neutrophil/macrophage chemokines were elevated in the bronchoalveolar lavage fluid or plasma of Mif lung-Tg2.1 mice. CONCLUSION Patients with GPA have an increased frequency of high MIF expression CATT alleles. Higher Mif expression increases the incidence of mortality and pulmonary granulomas in Mif lung-Tg2.1 mice, while anti-MIF treatment protects these mice against death. Blockade of MIF in high genotypic MIF expressers may therefore offer a selective pharmacologic therapy for GPA.
Collapse
Affiliation(s)
| | | | - Lin Leng
- Yale School of Medicine, New Haven, Connecticut
| | - Juan Fan
- Yale School of Medicine, New Haven, Connecticut
| | - Jie Yao
- Yale School of Medicine, New Haven, Connecticut
| | - Duncan Reid
- Yale School of Medicine, New Haven, Connecticut
| | | | - Simon Carette
- Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | | | - Paul Dellaripa
- Brigham and Women's Hospital, and Harvard University, Boston, Massachusetts
| | | | - Nader A Khalidi
- St. Joseph's Healthcare, McMaster University, Hamilton, Ontario, Canada
| | | | | | | | | | | | | | - Philip Seo
- Johns Hopkins University, Baltimore, Maryland
| | - Ulrich Specks
- Mayo Clinic College of Medicine, Rochester, Minnesota
| | | | - John H Stone
- Massachusetts General Hospital and Harvard University, Boston, Massachusetts
| | | | | | | | | |
Collapse
|
28
|
Gładysz D, Krzywdzińska A, Hozyasz KK. Immune Abnormalities in Autism Spectrum Disorder-Could They Hold Promise for Causative Treatment? Mol Neurobiol 2018; 55:6387-6435. [PMID: 29307081 PMCID: PMC6061181 DOI: 10.1007/s12035-017-0822-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 11/05/2017] [Indexed: 12/15/2022]
Abstract
Autism spectrum disorders (ASD) are characterized by impairments in language and communication development, social behavior, and the occurrence of stereotypic patterns of behavior and interests. Despite substantial speculation about causes of ASD, its exact etiology remains unknown. Recent studies highlight a link between immune dysfunction and behavioral traits. Various immune anomalies, including humoral and cellular immunity along with abnormalities at the molecular level, have been reported. There is evidence of altered immune function both in cerebrospinal fluid and peripheral blood. Several studies hypothesize a role for neuroinflammation in ASD and are supported by brain tissue and cerebrospinal fluid analysis, as well as evidence of microglial activation. It has been shown that immune abnormalities occur in a substantial number of individuals with ASD. Identifying subgroups with immune system dysregulation and linking specific cellular immunophenotypes to different symptoms would be key to defining a group of patients with immune abnormalities as a major etiology underlying behavioral symptoms. These determinations would provide the opportunity to investigate causative treatments for a defined patient group that may specifically benefit from such an approach. This review summarizes recent insights into immune system dysfunction in individuals with ASD and discusses the potential implications for future therapies.
Collapse
Affiliation(s)
- Dominika Gładysz
- Department of Pediatrics, Institute of Mother and Child, Warsaw, Poland
| | | | - Kamil K Hozyasz
- Department of Pediatrics, Institute of Mother and Child, Warsaw, Poland.
| |
Collapse
|
29
|
Bennabi M, Gaman A, Delorme R, Boukouaci W, Manier C, Scheid I, Si Mohammed N, Bengoufa D, Charron D, Krishnamoorthy R, Leboyer M, Tamouza R. HLA-class II haplotypes and Autism Spectrum Disorders. Sci Rep 2018; 8:7639. [PMID: 29769579 PMCID: PMC5955937 DOI: 10.1038/s41598-018-25974-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 04/26/2018] [Indexed: 02/06/2023] Open
Abstract
Infections and autoimmunity are associated with autism spectrum disorders (ASD), with both strongly influenced by the genetic regulation of the human leukocyte antigen (HLA) system. The relationship between ASD and the HLA genetic diversity requires further investigation. Using a case control design, the distribution of HLA class II-DRB1 and DQB1 alleles, genotypes and haplotypes were investigated in ASD patients, versus healthy controls (HC). ASD patients meeting DSM-IV TR criteria and HC (474 and 350 respectively) were genotyped at medium resolution using a Luminex-based SSO technology. Comparisons of genotypes, allele frequencies associated with a haplotype analysis were performed. Results indicate: (i) the HLA-DRB1 *11-DQB1*07 haplotype was more prevalent in ASD patients, versus HC (Pc = 0.001), partially replicating previous data and possibly linking to gastro-intestinal (GI)-related pro-inflammatory processes, given that this haplotype associates with pediatric celiac disorders; (ii) the HLA-DRB1 *17-DQB1*02 haplotype was higher in HC, versus ASD patients (Pc = 0.002), indicating that this is a protective haplotype. Using the Autism Diagnostic Interview to assess clinical dimensions, higher scores on social (Pc = 0.006) and non-verbal functioning (Pc = 0.004) associated with the DRB1 *11 DQB1*07 haplotype. Our results support HLA involvement in ASD, with possible relevance to GI and gut-brain axis dysregulation.
Collapse
Affiliation(s)
- Meriem Bennabi
- INSERM, U1160, Hôpital Saint Louis, Paris, France.,INSERM, U955, Psychiatrie Translationnelle, Créteil, France.,Fondation FondaMental, Créteil, France
| | - Alexandru Gaman
- INSERM, U955, Psychiatrie Translationnelle, Créteil, France.,Fondation FondaMental, Créteil, France
| | - Richard Delorme
- Fondation FondaMental, Créteil, France.,DHU Protect, AP-HP, Service de psychiatrie de l'enfant et de l'adolescent, Hôpital Robert Debré, Paris, France.,Département de génétique humaine et fonctions cognitives, Institut Pasteur, Paris, France.,Université Paris Diderot, Sorbonne Paris-Cité, Paris, France
| | | | | | - Isabelle Scheid
- INSERM, U955, Psychiatrie Translationnelle, Créteil, France.,Fondation FondaMental, Créteil, France
| | | | - Djaouida Bengoufa
- Laboratoire Jean Dausset and LabEx Transplantex, Hôpital Saint Louis, Paris, France
| | - Dominique Charron
- INSERM, U1160, Hôpital Saint Louis, Paris, France.,Université Paris Diderot, Sorbonne Paris-Cité, Paris, France.,Laboratoire Jean Dausset and LabEx Transplantex, Hôpital Saint Louis, Paris, France
| | - Rajagopal Krishnamoorthy
- INSERM, U955, Psychiatrie Translationnelle, Créteil, France.,Fondation FondaMental, Créteil, France
| | - Marion Leboyer
- INSERM, U955, Psychiatrie Translationnelle, Créteil, France.,Fondation FondaMental, Créteil, France.,DHU PePSY, AP-HP, Pôle de Psychiatrie, Hôpitaux Universitaires Henri Mondor, Créteil, France.,Université Paris-Est-Créteil, Faculté de médecine, Créteil, France
| | - Ryad Tamouza
- INSERM, U1160, Hôpital Saint Louis, Paris, France. .,INSERM, U955, Psychiatrie Translationnelle, Créteil, France. .,Fondation FondaMental, Créteil, France. .,Université Paris Diderot, Sorbonne Paris-Cité, Paris, France. .,Laboratoire Jean Dausset and LabEx Transplantex, Hôpital Saint Louis, Paris, France. .,DHU PePSY, AP-HP, Pôle de Psychiatrie, Hôpitaux Universitaires Henri Mondor, Créteil, France.
| |
Collapse
|
30
|
Increased serum levels and promoter polymorphisms of macrophage migration inhibitory factor in schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2018; 83:33-41. [PMID: 29305329 DOI: 10.1016/j.pnpbp.2018.01.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 01/01/2018] [Accepted: 01/01/2018] [Indexed: 01/29/2023]
Abstract
BACKGROUND Numerous studies have suggested that an immune system imbalance plays an important role in schizophrenia. Macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine. It plays multiple roles in various biological processes, including inflammation and neurogenesis. Furthermore, several exhaustive serum proteomic profiling studies have identified MIF as a potential biomarker of schizophrenia. Here, we investigate MIF protein levels in serum and postmortem prefrontal cortex in patients with schizophrenia and controls. Moreover, we investigate the association of two functional polymorphisms in the MIF gene promoter region (MIF-794CATT5-8 microsatellite and MIF-173G/C single-nucleotide polymorphism [SNP]) with schizophrenia. METHODS We measured serum MIF levels with an enzyme-linked immunosorbent assay (ELISA) (51 patients vs. 86 controls) and postmortem brain MIF levels with a western blotting assay (18 patients vs. 22 controls). Subsequently, we genotyped the MIF-794CATT5-8 microsatellite with a fluorescence-based fragment assay and the MIF-173G/C SNP with a TaqMan SNP genotyping assay (1483 patients vs. 1454 controls). RESULTS Serum MIF levels were significantly higher in patients with schizophrenia than in controls (p=0.00118), and were positively correlated with antipsychotic dose (Spearman's r=0.222, p=0.0402). In addition, an earlier age of onset was observed in patients with a high serum MIF level (≥40ng/mL) than those with a low serum MIF level (<40ng/mL) (p=0.0392). However, postmortem brain MIF levels did not differ between patients with schizophrenia and controls. The association study revealed that the CATT6-G haplotype was nominally significantly associated with schizophrenia (p=0.0338), and that the CATT6 allele and CATT6-G haplotype were significantly associated with female adolescent-onset schizophrenia (AsOS) (corrected p=0.0222 and p=0.0147, respectively). CONCLUSIONS These results suggest that serum MIF level is a potential pharmacodynamic and/or monitoring marker of schizophrenia, and is related to a novel antipsychotic effect beyond dopamine antagonism. Furthermore, the MIF gene polymorphisms are associated with the risk for schizophrenia especially in adolescent females, and are potential stratification markers of schizophrenia. Further studies of MIF are warranted to elucidate the pathophysiology of schizophrenia and the effects of antipsychotics.
Collapse
|
31
|
Cristiano C, Lama A, Lembo F, Mollica MP, Calignano A, Mattace Raso G. Interplay Between Peripheral and Central Inflammation in Autism Spectrum Disorders: Possible Nutritional and Therapeutic Strategies. Front Physiol 2018; 9:184. [PMID: 29563885 PMCID: PMC5845898 DOI: 10.3389/fphys.2018.00184] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 02/20/2018] [Indexed: 12/16/2022] Open
Abstract
Pre- and post-natal factors can affect brain development and function, impacting health outcomes with particular relevance to neurodevelopmental diseases, such as autism spectrum disorders (ASDs). Maternal obesity and its associated complications have been related to the increased risk of ASDs in offspring. Indeed, animals exposed to maternal obesity or high fat diets are prone to social communication impairment and repetitive behavior, the hallmarks of autism. During development, fatty acids and sugars, as well as satiety hormones, like insulin and leptin, and inflammatory factors related to obesity-induced low grade inflammation, could play a role in the impairment of neuroendocrine system and brain neuronal circuits regulating behavior in offspring. On the other side, post-natal factors, such as mode of delivery, stress, diet, or antibiotic treatment are associated to a modification of gut microbiota composition, perturbing microbiota-gut-brain axis. Indeed, the interplay between the gastrointestinal tract and the central nervous system not only occurs through neural, hormonal, and immune pathways, but also through microbe-derived metabolic products. The modification of unhealthy perinatal and postnatal environment, manipulation of gut microbiota, nutritional, and dietary interventions could represent possible strategies in preventing or limiting ASDs, through targeting inflammatory process and gut microbiota.
Collapse
Affiliation(s)
- Claudia Cristiano
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Adriano Lama
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Francesca Lembo
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Maria P Mollica
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Antonio Calignano
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | | |
Collapse
|
32
|
Macrophage migration inhibitory factor: A multifaceted cytokine implicated in multiple neurological diseases. Exp Neurol 2017; 301:83-91. [PMID: 28679106 DOI: 10.1016/j.expneurol.2017.06.021] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 06/06/2017] [Accepted: 06/21/2017] [Indexed: 12/12/2022]
Abstract
Macrophage migration inhibitory factor (MIF) is a conserved cytokine found as a homotrimer protein. It is found in a wide spectrum of cell types in the body including neuronal and non-neuronal cells. MIF is implicated in several biological processes; chemo-attraction, cytokine activity, and receptor binding, among other functions. More recently, a chaperone-like activity has been added to its repertoire. In this review, we focus on the implication of MIF in the central nervous system and peripheries, its role in neurological disorders, and the mechanisms by which MIF is regulated. Numerous studies have associated MIF with various disease settings. MIF plays an important role in advocating tumorigenic processes, Alzheimer's disease, and is also upregulated in autism-spectrum disorders and spinal cord injury where it contributes to the severity of the injured area. The protective effect of MIF has been reported in amyotrophic lateral sclerosis by its reduction of aggregated misfolded SOD1, subsequently reducing the severity of this disease. Interestingly, a protective as well as pathological role for MIF has been implicated in stroke and cerebral ischemia, as well as depression. Thus, the role of MIF in neurological disorders appears to be diverse with both beneficial and adversary effects. Furthermore, its modulation is rather complex and it is regulated by different proteins, either on a molecular or protein level. This complexity might be dependent on the pathophysiological context and/or cellular microenvironment. Hence, further clarification of its diverse roles in neurological pathologies is warranted to provide new mechanistic insights which may lead in the future to the development of therapeutic strategies based on MIF, to fight some of these neurological disorders.
Collapse
|
33
|
Onore C, Yang H, Van de Water J, Ashwood P. Dynamic Akt/mTOR Signaling in Children with Autism Spectrum Disorder. Front Pediatr 2017; 5:43. [PMID: 28361047 PMCID: PMC5350147 DOI: 10.3389/fped.2017.00043] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 02/15/2017] [Indexed: 12/15/2022] Open
Abstract
Autism spectrum disorder (ASD) is a behaviorally defined disorder affecting 1 in 68 children. Currently, there is no known cause for the majority of ASD cases nor are there physiological diagnostic tools or biomarkers to aid behavioral diagnosis. Whole-genome linkage studies, genome-wide association studies, copy number variation screening, and SNP analyses have identified several ASD candidate genes, but which vary greatly among individuals and family clusters, suggesting that a variety of genetic mutations may result in a common pathology or alter a common mechanistic pathway. The Akt/mammalian target of rapamycin (mTOR) pathway is involved in many cellular processes including synaptic plasticity and immune function that can alter neurodevelopment. In this study, we examined the activity of the Akt/mTOR pathway in cells isolated from children with ASD and typically developing controls. We observed higher activity of mTOR, extracellular receptor kinase, and p70S6 kinase and lower activity of glycogen synthase kinase 3 (GSK3)α and tuberin (TSC2) in cells from children with ASD. These data suggest a phosphorylation pattern indicative of higher activity in the Akt/mTOR pathway in children with general/idiopathic ASD and may suggest a common pathological pathway of interest for ASD.
Collapse
Affiliation(s)
- Charity Onore
- The M.I.N.D. Institute, University of California Davis, Davis, CA, USA
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
| | - Houa Yang
- The M.I.N.D. 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
- The M.I.N.D. Institute, University of California Davis, Davis, CA, USA
- Division of Rheumatology, Allergy and Clinical Immunology, University of California Davis, Davis, CA, USA
| | - Paul Ashwood
- The M.I.N.D. Institute, University of California Davis, Davis, CA, USA
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
| |
Collapse
|
34
|
Vuong HE, Hsiao EY. Emerging Roles for the Gut Microbiome in Autism Spectrum Disorder. Biol Psychiatry 2017; 81:411-423. [PMID: 27773355 PMCID: PMC5285286 DOI: 10.1016/j.biopsych.2016.08.024] [Citation(s) in RCA: 357] [Impact Index Per Article: 44.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 07/28/2016] [Accepted: 08/18/2016] [Indexed: 01/16/2023]
Abstract
Autism spectrum disorder (ASD) is a serious neurodevelopmental disorder that affects one in 45 children in the United States, with a similarly striking prevalence in countries around the world. However, mechanisms underlying its etiology and manifestations remain poorly understood. Although ASD is diagnosed based on the presence and severity of impaired social communication and repetitive behavior, immune dysregulation and gastrointestinal issues are common comorbidities. The microbiome is an integral part of human physiology; recent studies show that changes in the gut microbiota can modulate gastrointestinal physiology, immune function, and even behavior. Links between particular bacteria from the indigenous gut microbiota and phenotypes relevant to ASD raise the important question of whether microbial dysbiosis plays a role in the development or presentation of ASD symptoms. Here we review reports of microbial dysbiosis in ASD. We further discuss potential effects of the microbiota on ASD-associated symptoms, drawing on signaling mechanisms for reciprocal interactions among the microbiota, immunity, gut function, and behavior. In addition, we discuss recent findings supporting a role for the microbiome as an interface between environmental and genetic risk factors that are associated with ASD. These studies highlight the integration of pathways across multiple body systems that together can impact brain and behavior and suggest that changes in the microbiome may contribute to symptoms of neurodevelopmental disease.
Collapse
Affiliation(s)
- Helen E. Vuong
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Elaine Y. Hsiao
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA,Correspondence to: ; 610 Charles E. Young Drive MSB 3825A; Los Angeles CA 90095; 310-825-0228
| |
Collapse
|
35
|
Han YMY, Cheung WKY, Wong CK, Sze SL, Cheng TWS, Yeung MK, Chan AS. Distinct Cytokine and Chemokine Profiles in Autism Spectrum Disorders. Front Immunol 2017; 8:11. [PMID: 28167942 PMCID: PMC5253384 DOI: 10.3389/fimmu.2017.00011] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 01/04/2017] [Indexed: 12/17/2022] Open
Abstract
Previous studies have shown that immunological factors are involved in the pathogenesis of autism spectrum disorders (ASDs). However, this research has been conducted almost exclusively in Western contexts, and only a handful of studies on immune measures have been conducted in Asian populations, such as Chinese populations. The present study examined whether immunological abnormalities are associated with cognitive deficits and problem behaviors in Chinese children with ASD and whether these children show different immunological profiles. Thirteen typically developing (TD) children and 22 children with ASD, aged 6–17 years, participated voluntarily in the study. Executive functions and short-term memory were measured using neuropsychological tests, and behavioral measures were assessed using parent ratings. The children were also assessed on immunological measures, specifically, the levels of cytokines and chemokines in the blood serum. Children with ASD showed greater deficits in cognitive functions, as well as altered levels of immunological measures, including CCL2, CCL5, and CXCL9 levels, compared to TD children, and the cognitive functions and associated behavioral deficits of children with ASD were significantly associated with different immunological measures. The children were further sub-classified into ASD with only autistic features (ASD-only) or ASD comorbid with attention deficit hyperactivity disorder (ASD + ADHD). The comorbidity results showed that there were no differences between the two groups of ASD children in any of the cognitive or behavioral measures. However, the results pertaining to immunological measures showed that the children with ASD-only and ASD + ADHD exhibited distinct cytokine and chemokine profiles and that abnormal immunologic function was associated with cognitive functions and inattention/hyperactivity symptoms. These results support the notion that altered immune functions may play a role in the selective cognitive and behavioral symptoms of ASD.
Collapse
Affiliation(s)
- Yvonne M Y Han
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University , Hong Kong , China
| | - Winnie K Y Cheung
- Neuropsychology Laboratory, Department of Psychology, The Chinese University of Hong Kong , Hong Kong , China
| | - Chun Kwok Wong
- Department of Chemical Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong , Hong Kong , China
| | - Sophia L Sze
- Neuropsychology Laboratory, Department of Psychology, The Chinese University of Hong Kong, Hong Kong, China; Chanwuyi Research Center for Neuropsychological Well-Being, The Chinese University of Hong Kong, Hong Kong, China
| | - Timmy W S Cheng
- Department of Chemical Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong , Hong Kong , China
| | - Michael K Yeung
- Neuropsychology Laboratory, Department of Psychology, The Chinese University of Hong Kong , Hong Kong , China
| | - Agnes S Chan
- Neuropsychology Laboratory, Department of Psychology, The Chinese University of Hong Kong, Hong Kong, China; Chanwuyi Research Center for Neuropsychological Well-Being, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
36
|
Makinodan M, Iwata K, Ikawa D, Yamashita Y, Yamamuro K, Toritsuka M, Kimoto S, Okumura K, Yamauchi T, Yoshino H, Tsujii M, Sugiyama T, Tsuchiya K, Mori N, Matsuzaki H, Kishimoto T. Tumor necrosis factor-alpha expression in peripheral blood mononuclear cells correlates with early childhood social interaction in autism spectrum disorder. Neurochem Int 2016; 104:1-5. [PMID: 28007470 DOI: 10.1016/j.neuint.2016.12.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 12/12/2016] [Accepted: 12/15/2016] [Indexed: 12/14/2022]
Abstract
Autism spectrum disorder is a neurodevelopmental disorder characterized by impaired social interaction, poor communication skills, and repetitive/restrictive behaviors. Elevated blood levels of pro-inflammatory cytokines have been reported in subjects with autism spectrum disorder. On the other hand, early childhood adverse experience also increases blood levels of these cytokines. Since social experience of children with autism spectrum disorder is generally unlike to typically developing children, we hypothesized that social interaction during childhood contribute to pro-inflammatory cytokine expression in subjects with autism spectrum disorder. We compared revised Autism Diagnostic Interview scores and expression levels of pro-inflammatory cytokines in peripheral blood mononuclear cells of subjects with autism spectrum disorder (n = 30). The score of domain A on the revised Autism Diagnostic Interview, indicating social interaction impairment in early childhood, was negatively correlated with tumor necrosis factor-α mRNA expression level in peripheral blood mononuclear cells but not interleukin-1β or -6. Consistently, tumor necrosis factor-α mRNA expression was markedly low in subjects with autism spectrum disorder compared to typically developing children who presumably experienced the regular levels of social interaction. These findings suggest that the low blood levels of tumor necrosis factor-α mRNA in subjects with autism spectrum disorder might be due to impaired social interaction in early childhood.
Collapse
Affiliation(s)
- Manabu Makinodan
- Department of Psychiatry, Nara Medical University School of Medicine, Nara, 634-8522, Japan.
| | - Keiko Iwata
- Research Center for Child Mental Development, University of Fukui, Japan; Department of Development of Functional Brain Activities, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Fukui, Japan
| | - Daisuke Ikawa
- Department of Psychiatry, Nara Medical University School of Medicine, Nara, 634-8522, Japan
| | - Yasunori Yamashita
- Department of Psychiatry, Nara Medical University School of Medicine, Nara, 634-8522, Japan
| | - Kazuhiko Yamamuro
- Department of Psychiatry, Nara Medical University School of Medicine, Nara, 634-8522, Japan
| | - Michihiro Toritsuka
- Department of Psychiatry, Nara Medical University School of Medicine, Nara, 634-8522, Japan
| | - Sohei Kimoto
- Department of Psychiatry, Nara Medical University School of Medicine, Nara, 634-8522, Japan
| | - Kazuki Okumura
- Department of Psychiatry, Nara Medical University School of Medicine, Nara, 634-8522, Japan
| | - Takahira Yamauchi
- Department of Psychiatry, Nara Medical University School of Medicine, Nara, 634-8522, Japan
| | - Hiroki Yoshino
- Department of Psychiatry, Nara Medical University School of Medicine, Nara, 634-8522, Japan
| | - Masatsugu Tsujii
- Department of Faculty of Sociology, Chukyo University, Toyota, Japan
| | | | - Kenji Tsuchiya
- Department of Psychiatry and Neurology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Norio Mori
- Department of Psychiatry and Neurology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hideo Matsuzaki
- Research Center for Child Mental Development, University of Fukui, Japan; Department of Development of Functional Brain Activities, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Fukui, Japan
| | - Toshifumi Kishimoto
- Department of Psychiatry, Nara Medical University School of Medicine, Nara, 634-8522, Japan
| |
Collapse
|
37
|
Torres AR, Sweeten TL, Johnson RC, Odell D, Westover JB, Bray-Ward P, Ward DC, Davies CJ, Thomas AJ, Croen LA, Benson M. Common Genetic Variants Found in HLA and KIR Immune Genes in Autism Spectrum Disorder. Front Neurosci 2016; 10:463. [PMID: 27812316 PMCID: PMC5071356 DOI: 10.3389/fnins.2016.00463] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 09/26/2016] [Indexed: 11/13/2022] Open
Abstract
The "common variant-common disease" hypothesis was proposed to explain diseases with strong inheritance. This model suggests that a genetic disease is the result of the combination of several common genetic variants. Common genetic variants are described as a 5% frequency differential between diseased vs. matched control populations. This theory was recently supported by an epidemiology paper stating that about 50% of genetic risk for autism resides in common variants. However, rare variants, rather than common variants, have been found in numerous genome wide genetic studies and many have concluded that the "common variant-common disease" hypothesis is incorrect. One interpretation is that rare variants are major contributors to genetic diseases and autism involves the interaction of many rare variants, especially in the brain. It is obvious there is much yet to be learned about autism genetics. Evidence has been mounting over the years indicating immune involvement in autism, particularly the HLA genes on chromosome 6 and KIR genes on chromosome 19. These two large multigene complexes have important immune functions and have been shown to interact to eliminate unwanted virally infected and malignant cells. HLA proteins have important functions in antigen presentation in adaptive immunity and specific epitopes on HLA class I proteins act as cognate ligands for KIR receptors in innate immunity. Data suggests that HLA alleles and KIR activating genes/haplotypes are common variants in different autism populations. For example, class I allele (HLA-A2 and HLA-G 14 bp-indel) frequencies are significantly increased by more than 5% over control populations (Table 2). The HLA-DR4 Class II and shared epitope frequencies are significantly above the control populations (Table 2). Three activating KIR genes: 3DS1, 2DS1, and 2DS2 have increased frequencies of 15, 22, and 14% in autism populations, respectively. There is a 6% increase in total activating KIR genes in autism over control subjects. And, more importantly there is a 12% increase in activating KIR genes and their cognate HLA alleles over control populations (Torres et al., 2012a). These data suggest the interaction of HLA ligand/KIR receptor pairs encoded on two different chromosomes is more significant as a ligand/receptor complex than separately in autism.
Collapse
Affiliation(s)
- Anthony R. Torres
- Center for Persons with Disabilities, Utah State UniversityLogan, UT, USA
| | | | - Randall C. Johnson
- BSP CCR Genetics Core, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer ResearchFrederick, MD, USA
| | - Dennis Odell
- Center for Persons with Disabilities, Utah State UniversityLogan, UT, USA
| | - Jonna B. Westover
- Center for Persons with Disabilities, Utah State UniversityLogan, UT, USA
| | - Patricia Bray-Ward
- Center for Persons with Disabilities, Utah State UniversityLogan, UT, USA
| | - David C. Ward
- Center for Persons with Disabilities, Utah State UniversityLogan, UT, USA
| | | | - Aaron J. Thomas
- Division of Research, Kaiser Permanente of Northern CaliforniaOakland, CA, USA
| | - Lisa A. Croen
- Center for Integrated BioSystems, Utah State UniversityLogan, UT, USA
| | - Michael Benson
- Center for Persons with Disabilities, Utah State UniversityLogan, UT, USA
| |
Collapse
|
38
|
Yao J, Leng L, Sauler M, Fu W, Zheng J, Zhang Y, Du X, Yu X, Lee P, Bucala R. Transcription factor ICBP90 regulates the MIF promoter and immune susceptibility locus. J Clin Invest 2016; 126:732-44. [PMID: 26752645 DOI: 10.1172/jci81937] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 11/18/2015] [Indexed: 02/02/2023] Open
Abstract
The immunoregulatory cytokine macrophage migration inhibitory factor (MIF) is encoded in a functionally polymorphic locus that is linked to the susceptibility of autoimmune and infectious diseases. The MIF promoter contains a 4-nucleotide microsatellite polymorphism (-794 CATT) that repeats 5 to 8 times in the locus, with greater numbers of repeats associated with higher mRNA levels. Because there is no information about the transcriptional regulation of these common alleles, we used oligonucleotide affinity chromatography and liquid chromatography-mass spectrometry to identify nuclear proteins that interact with the -794 CATT5-8 site. An analysis of monocyte nuclear lysates revealed that the transcription factor ICBP90 (also known as UHRF1) is the major protein interacting with the MIF microsatellite. We found that ICBP90 is essential for MIF transcription from monocytes/macrophages, B and T lymphocytes, and synovial fibroblasts, and TLR-induced MIF transcription is regulated in an ICBP90- and -794 CATT5-8 length-dependent manner. Whole-genome transcription analysis of ICBP90 shRNA-treated rheumatoid synoviocytes uncovered a subset of proinflammatory and immune response genes that overlapped with those regulated by MIF shRNA. In addition, the expression levels of ICBP90 and MIF were correlated in joint synovia from patients with rheumatoid arthritis. These findings identify ICBP90 as a key regulator of MIF transcription and provide functional insight into the regulation of the polymorphic MIF locus.
Collapse
|
39
|
Reinards THCM, Albers HM, Brinkman DMC, Kamphuis SSM, van Rossum MAJ, Girschick HJ, Wouters C, Hoppenreijs EPAH, Saurenmann RK, Hinks A, Ellis JA, Bakker E, Verduijn W, Slagboom P, Huizinga TWJ, Toes REM, Houwing-Duistermaat JJ, ten Cate R, Schilham MW. CD226 (DNAM-1) is associated with susceptibility to juvenile idiopathic arthritis. Ann Rheum Dis 2015; 74:2193-8. [PMID: 25057181 DOI: 10.1136/annrheumdis-2013-205138] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 07/11/2014] [Indexed: 01/08/2023]
Abstract
OBJECTIVES Juvenile idiopathic arthritis (JIA) is considered a complex genetic autoimmune disease. We investigated the association of genetic variants previously implicated in JIA, autoimmunity and/or immunoregulation, with susceptibility to JIA. METHODS A genetic association study was performed in 639 JIA patients and 1613 healthy controls of northwest European descent. Ninety-three single nucleotide polymorphisms (SNP) were genotyped in a candidate gene approach. Results of the entire JIA patient group (all subtypes) were compared with results obtained, alternatively, with a clinically homogeneous patient group including only oligoarticular and rheumatoid factor (RF) negative polyarticular JIA patients (n=493). Meta-analyses were performed for all SNPs that have been typed in other Caucasian JIA cohorts before. RESULTS SNPs in or near PTPN22, VTCN1, the IL2-IL21 region, ANKRD55 and TNFA were confirmed to be associated with JIA (p<0.05), strengthening the evidence for involvement of these genes in JIA. In the majority of these replicated SNPs, effect sizes were larger when analysing a homogeneous patient cohort than when analysing all subtypes. We identified two novel associations with oligoarticular and RF-negative polyarticular JIA: CD226 rs763361 (OR 1.30, 95% CI 1.12 to 1.51, p=0.0006) and CD28 rs1980422 (OR 1.29, 95% CI 1.07 to 1.55, p=0.008). Meta-analyses including reported studies confirmed the association of both SNPs with susceptibility to JIA (OR 1.16, p=0.001 and OR 1.18, p=0.001, for rs763361 and rs1980422, respectively). CONCLUSIONS The CD226 gene has been identified as novel association with JIA, and a SNP near CD28 as a suggestive association. Both genes are probable candidate risk factors, since they are involved in costimulation of T cells.
Collapse
Affiliation(s)
- T H C M Reinards
- Department of Pediatrics/Pediatric Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - H M Albers
- Department of Pediatrics/Pediatric Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - D M C Brinkman
- Department of Pediatrics/Pediatric Rheumatology, Rijnland Hospital, Leiderdorp, The Netherlands
| | - S S M Kamphuis
- Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital, Rotterdam, The Netherlands
| | - M A J van Rossum
- Department of Pediatrics/Pediatric Rheumatology, Academic Medical Centre/Emma Children's Hospital and Reade (Jan van Breemen location), Amsterdam, The Netherlands
| | - H J Girschick
- Vivantes Children's Hospital, Berlin-Friedrichshain, Germany
| | - C Wouters
- University Hospital Gasthuisberg, Leuven, Belgium
| | - E P A H Hoppenreijs
- Department of Pediatrics/Pediatric Rheumatology, St Maartenskliniek and Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - R K Saurenmann
- Zürich University Children's Hospital, Zürich, Switzerland
| | - A Hinks
- Arthritis Research UK Epidemiology Unit, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - J A Ellis
- Department of Pediatrics, The University of Melbourne, Melbourne, Australia Murdoch Childrens Research Institute, Royal Children's Hospital, Parkville, Australia
| | - E Bakker
- Centre for Human and Clinical Genetics/Laboratory for Diagnostic Genome Analysis, Leiden University Medical Center, Leiden, The Netherlands
| | - W Verduijn
- Department of Immunohematology and Bloodtransfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - P Slagboom
- Department of Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - T W J Huizinga
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - R E M Toes
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - R ten Cate
- Department of Pediatrics/Pediatric Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - M W Schilham
- Department of Pediatrics/Laboratory for Immunology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
40
|
Bennabi M, Delorme R, Oliveira J, Fortier C, Lajnef M, Boukouaci W, Feugeas JP, Marzais F, Gaman A, Charron D, Ghaleh B, Krishnamoorthy R, Leboyer M, Tamouza R. Dectin-1 Polymorphism: A Genetic Disease Specifier in Autism Spectrum Disorders? PLoS One 2015; 10:e0137339. [PMID: 26352598 PMCID: PMC4564239 DOI: 10.1371/journal.pone.0137339] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 08/14/2015] [Indexed: 12/26/2022] Open
Abstract
Introduction In autism spectrum disorders (ASD), complex gene-environment interactions contribute to disease onset and progress. Given that gastro-intestinal dysfunctions are common in ASD, we postulated involvement of microbial dysbiosis in ASD and investigated, under a case-control design, the influence of DNA polymorphisms in the CLEC7A gene that encodes a pivotal fungal sensor, Dectin-1. Material and methods DNAs from 478 ASD patients and 351 healthy controls (HC) were analyzed for the CLEC7A rs16910631G/A and rs2078178 A/G single nucleotide polymorphisms (SNPs). Differences in the distribution of allele, genotype and haplotype by Chi-square testing and nonparametric analysis by Kruskal-Wallis/Mann–Whitney tests, where appropriate, were performed. The free statistical package R.2.13 software was used for the statistical analysis. Results We found that the CLEC7A rs2078178 G allele and GG genotype were more prevalent in HC as compared to ASD but failed to reach statistical significance for the latter (pc = 0.01, 0.06 respectively). However, after phenotype-based stratification, the CLEC7A rs2078178 G allele and GG genotype were found to be significantly more frequent in the Asperger group as compared to other ASD subsets (pc = 0.02, 0.01), a finding reinforced by haplotype analysis (rs2078178/rs16910631 G-G/G-G) (pc = 0.002). Further, intellectual quotient (IQ)-based stratification of ASD patients revealed that IQ values increase linearly along the CLEC7A rs2078178 AA, AG and GG genotypes (p = 0.05) and in a recessive manner (GG vs. AA+AG p = 0.02), further confirmed by haplotype distribution (CLEC7A rs2078178-16910631; A-G/A-G, A-G/G-G and G-G/G-G, p = 0.02, G-G/G-G vs. others, p = 0.01). Conclusion Our data suggest that the genetic diversity of CLEC7A gene influences the ASD phenotype by behaving as a disease specifier and imply that the genetic control of innate immune response could determine the ASD phenotype.
Collapse
Affiliation(s)
- Meriem Bennabi
- INSERM, U1160, Hôpital Saint Louis, Paris, France
- INSERM, U955, Psychiatrie Génétique, Créteil, France
- Fondation FondaMental, Créteil, France
| | - Richard Delorme
- DHU Protect, Service de Psychiatrie de l’Enfant et de l’Adolescent, Hôpital Robert Debré, Paris, France
- Département de Génétique Humaine et Fonctions Cognitives, Institut Pasteur, Paris, France
| | - José Oliveira
- INSERM, U1160, Hôpital Saint Louis, Paris, France
- INSERM, U955, Psychiatrie Génétique, Créteil, France
- Fondation FondaMental, Créteil, France
| | - Catherine Fortier
- Laboratoire Jean Dausset and LabEx Transplantex, Hôpital Saint Louis, Paris, France
| | - Mohamed Lajnef
- INSERM, U955, Psychiatrie Génétique, Créteil, France
- Fondation FondaMental, Créteil, France
| | | | - Jean-Paul Feugeas
- INSERM, U1137, Hôpital Bichat, Paris, France
- Université Paris Diderot, Sorbonne Paris-Cité, Paris, France
| | - François Marzais
- Laboratoire Jean Dausset and LabEx Transplantex, Hôpital Saint Louis, Paris, France
| | - Alexandru Gaman
- INSERM, U955, Psychiatrie Génétique, Créteil, France
- Fondation FondaMental, Créteil, France
| | - Dominique Charron
- Laboratoire Jean Dausset and LabEx Transplantex, Hôpital Saint Louis, Paris, France
- Université Paris Diderot, Sorbonne Paris-Cité, Paris, France
| | - Bijan Ghaleh
- Centre de Ressources Biologiques, Hôpitaux Universitaires Henri Mondor, Créteil, France
| | | | - Marion Leboyer
- INSERM, U955, Psychiatrie Génétique, Créteil, France
- Fondation FondaMental, Créteil, France
- AP-HP, Pôle de Psychiatrie, DHU PePSY, Hôpitaux Universitaires Henri Mondor, Créteil, France
- Université Paris-Est, Faculté de Médecine, Créteil, France
| | - Ryad Tamouza
- INSERM, U1160, Hôpital Saint Louis, Paris, France
- Fondation FondaMental, Créteil, France
- Laboratoire Jean Dausset and LabEx Transplantex, Hôpital Saint Louis, Paris, France
- Université Paris Diderot, Sorbonne Paris-Cité, Paris, France
- * E-mail:
| |
Collapse
|
41
|
Abstract
Autism spectrum disorders (ASD) are complex neurodevelopmental disorders characterized by impairments in three core behavioral areas. As prevalence rates for ASD continue to rise there is also increasing interest in finding biomarkers associated with ASD. The use of biomarkers could help identify those at risk for ASD or ASD-associated comorbid conditions and help to predict the developmental course of these children. Due to the heterogeneity of ASD, biomarkers may help to identify subpopulations within ASD that share similar traits or profiles. Such work could lead to specialized therapy and help to develop biomarkers whereby the benefits of treatments/therapies for individuals could be monitored over time and through clinical trials. Over the last 10 years, the evidence of immune involvement in ASD has been steadily growing and many investigators have begun to look at possible immune biomarkers, such as immune cytokine profiles, in children with ASD.
Collapse
Affiliation(s)
- Destanie Rose
- Department of Medical Microbiology & Immunology, University of California Davis, CA 95616, USA
| | | |
Collapse
|
42
|
Exploring the Potential Role of Inflammation as an Etiological Process in ASD. REVIEW JOURNAL OF AUTISM AND DEVELOPMENTAL DISORDERS 2015. [DOI: 10.1007/s40489-015-0051-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
43
|
Jaiswal P, Mohanakumar KP, Rajamma U. Serotonin mediated immunoregulation and neural functions: Complicity in the aetiology of autism spectrum disorders. Neurosci Biobehav Rev 2015; 55:413-31. [PMID: 26021727 DOI: 10.1016/j.neubiorev.2015.05.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 05/11/2015] [Accepted: 05/12/2015] [Indexed: 12/19/2022]
Abstract
Serotonergic system has long been implicated in the aetiology of autism spectrum disorders (ASD), since platelet hyperserotonemia is consistently observed in a subset of autistic patients, who respond well to selective serotonin reuptake inhibitors. Apart from being a neurotransmitter, serotonin functions as a neurotrophic factor directing brain development and as an immunoregulator modulating immune responses. Serotonin transporter (SERT) regulates serotonin level in lymphoid tissues to ensure its proper functioning in innate and adaptive responses. Immunological molecules such as cytokines in turn regulate the transcription and activity of SERT. Dysregulation of serotonergic system could trigger signalling cascades that affect normal neural-immune interactions culminating in neurodevelopmental and neural connectivity defects precipitating behavioural abnormalities, or the disease phenotypes. Therefore, we suggest that a better understanding of the cross talk between serotonergic genes, immune systems and serotonergic neurotransmission will open wider avenues to develop pharmacological leads for addressing the core ASD behavioural deficits.
Collapse
Affiliation(s)
- Preeti Jaiswal
- Manovikas Biomedical Research and Diagnostic Centre, Manovikas Kendra, 482 Madudah, Plot I-24, Sector-J, EM Bypass, Kolkata 700 107, India
| | - Kochupurackal P Mohanakumar
- Division of Cell Biology & Physiology, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata 700 032, India
| | - Usha Rajamma
- Manovikas Biomedical Research and Diagnostic Centre, Manovikas Kendra, 482 Madudah, Plot I-24, Sector-J, EM Bypass, Kolkata 700 107, India.
| |
Collapse
|
44
|
Altered mRNA Levels of Glucocorticoid Receptor, Mineralocorticoid Receptor, and Co-Chaperones (FKBP5 and PTGES3) in the Middle Frontal Gyrus of Autism Spectrum Disorder Subjects. Mol Neurobiol 2015; 53:2090-9. [DOI: 10.1007/s12035-015-9178-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 04/13/2015] [Indexed: 01/20/2023]
|
45
|
The many roads to mitochondrial dysfunction in neuroimmune and neuropsychiatric disorders. BMC Med 2015; 13:68. [PMID: 25889215 PMCID: PMC4382850 DOI: 10.1186/s12916-015-0310-y] [Citation(s) in RCA: 169] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 03/04/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Mitochondrial dysfunction and defects in oxidative metabolism are a characteristic feature of many chronic illnesses not currently classified as mitochondrial diseases. Examples of such illnesses include bipolar disorder, multiple sclerosis, Parkinson's disease, schizophrenia, depression, autism, and chronic fatigue syndrome. DISCUSSION While the majority of patients with multiple sclerosis appear to have widespread mitochondrial dysfunction and impaired ATP production, the findings in patients diagnosed with Parkinson's disease, autism, depression, bipolar disorder schizophrenia and chronic fatigue syndrome are less consistent, likely reflecting the fact that these diagnoses do not represent a disease with a unitary pathogenesis and pathophysiology. However, investigations have revealed the presence of chronic oxidative stress to be an almost invariant finding in study cohorts of patients afforded each diagnosis. This state is characterized by elevated reactive oxygen and nitrogen species and/or reduced levels of glutathione, and goes hand in hand with chronic systemic inflammation with elevated levels of pro-inflammatory cytokines. SUMMARY This paper details mechanisms by which elevated levels of reactive oxygen and nitrogen species together with elevated pro-inflammatory cytokines could conspire to pave a major road to the development of mitochondrial dysfunction and impaired oxidative metabolism seen in many patients diagnosed with these disorders.
Collapse
|
46
|
Bashir S, Al-Ayadhi LY. Effect of camel milk on thymus and activation-regulated chemokine in autistic children: double-blind study. Pediatr Res 2014; 75:559-63. [PMID: 24375082 DOI: 10.1038/pr.2013.248] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 09/14/2013] [Indexed: 11/09/2022]
Abstract
BACKGROUND This study aimed to investigate the role of the effectiveness of camel milk (CM) (raw and boiled) on thymus and activation-regulated chemokine (TARC) serum levels and childhood autism rating scale (CARS) score in subjects with autism and compared to placebo group (cow milk). METHODS Forty-five subjects diagnosed with autism were randomly assigned to receive boiled CM for group I (n = 15), raw CM for group II (n = 15), and placebo for group III (n = 15) for 2 wk. Measures included changes in professionally completed CARS score and blood samples for TARC serum level were taken before and after milk consumption of 500 ml per day in children's regular daily diet. RESULTS The serum levels of TARC decreased significantly (P = 0.004) in boiled CM and in raw CM group (P = 0.01) too, but no effect was observed (P = 0.68) in placebo group. Furthermore, significant improvements were observed in CARS score (P = 0.04) in raw CM group only. There were no significant relationships between the serum of TARC level and the CARS score, age, or gender for any group. CONCLUSION CM administered for 2 wk significantly improved clinical measurements of autism severity and decreased serum level of TARC in autistic children, but subsequent studies are recommended.
Collapse
Affiliation(s)
- Shahid Bashir
- 1] Department of Physiology, Autism Research and Treatment Center, Shaik AL-Amodi Autism Research Chair, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia [2] Division of Cognitive Neurology, Department of Neurology, Berenson-Allen Center for Noninvasive Brain Stimulation, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Laila Y Al-Ayadhi
- Department of Physiology, Autism Research and Treatment Center, Shaik AL-Amodi Autism Research Chair, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
47
|
Goyal DK, Miyan JA. Neuro-immune abnormalities in autism and their relationship with the environment: a variable insult model for autism. Front Endocrinol (Lausanne) 2014; 5:29. [PMID: 24639668 PMCID: PMC3945747 DOI: 10.3389/fendo.2014.00029] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 02/20/2014] [Indexed: 12/20/2022] Open
Abstract
Autism spectrum disorder (ASD) is a heterogeneous condition affecting an individual's ability to communicate and socialize and often presents with repetitive movements or behaviors. It tends to be severe with less than 10% achieving independent living with a marked variation in the progression of the condition. To date, the literature supports a multifactorial model with the largest, most detailed twin study demonstrating strong environmental contribution to the development of the condition. Here, we present a brief review of the neurological, immunological, and autonomic abnormalities in ASD focusing on the causative roles of environmental agents and abnormal gut microbiota. We present a working hypothesis attempting to bring together the influence of environment on the abnormal neurological, immunological, and neuroimmunological functions and we explain in brief how such pathophysiology can lead to, and/or exacerbate ASD symptomatology. At present, there is a lack of consistent findings relating to the neurobiology of autism. Whilst we postulate such variable findings may reflect the marked heterogeneity in clinical presentation and as such the variable findings may be of pathophysiological relevance, more research into the neurobiology of autism is necessary before establishing a working hypothesis. Both the literature review and hypothesis presented here explore possible neurobiological explanations with an emphasis of environmental etiologies and are presented with this bias.
Collapse
Affiliation(s)
- Daniel K. Goyal
- Faculty of Life Sciences, The University of Manchester, Manchester, UK
| | - Jaleel A. Miyan
- Faculty of Life Sciences, The University of Manchester, Manchester, UK
| |
Collapse
|
48
|
Noriega DB, Savelkoul HFJ. Immune dysregulation in autism spectrum disorder. Eur J Pediatr 2014; 173:33-43. [PMID: 24297668 DOI: 10.1007/s00431-013-2183-4] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 10/09/2013] [Indexed: 12/25/2022]
Abstract
UNLABELLED Autism spectrum disorder (ASD) is a common and severe neuro-developmental disorder in early childhood which is defined by social and communication deficits and repetitive and stereotypic behaviours. The aetiology of ASD remains poorly understood. Susceptibility to development of ASD has significant environmental components, in addition to the profound genetic heritability. Few genes have been associated to the risk for ASD development. There is substantial evidence implicating chronic neurological inflammation and immune dysregulation leading to upregulation of inflammatory cytokines in the ASD brain, probably due to altered blood-brain barrier function. The immune system is characterized by excessive and skewed cytokine responses, modulated T cell reactivity, decreased regulation and production of immunosuppressive cytokines, modified NK function and increased autoantibody production. CONCLUSION The perinatal environment generates vulnerability to chronic neuro-inflammation in the brain associated with profound modulation and dysregulation in the immune system leading to the rapid development of ASD in genetically susceptible children.
Collapse
Affiliation(s)
- Daniela Briceno Noriega
- Cell Biology and Immunology Group, Wageningen University, P.O. Box 338, 6700 AH, Wageningen, The Netherlands
| | | |
Collapse
|
49
|
Current progress and challenges in the search for autism biomarkers. DISEASE MARKERS 2013; 35:55-65. [PMID: 24167349 PMCID: PMC3774962 DOI: 10.1155/2013/476276] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 04/06/2013] [Indexed: 11/17/2022]
Abstract
Autism spectrum disorders (ASD) encompass a range of neurodevelopmental conditions that are clinically and etiologically very heterogeneous. ASD is currently diagnosed entirely on behavioral criteria, but intensive research efforts are focused on identifying biological markers for disease risk and early diagnosis. Here, we discuss recent progress toward identifying biological markers for ASD and highlight specific challenges as well as ethical aspects of translating ASD biomarker research into the clinic.
Collapse
|
50
|
Napolioni V, Ober-Reynolds B, Szelinger S, Corneveaux JJ, Pawlowski T, Ober-Reynolds S, Kirwan J, Persico AM, Melmed RD, Craig DW, Smith CJ, Huentelman MJ. Plasma cytokine profiling in sibling pairs discordant for autism spectrum disorder. J Neuroinflammation 2013; 10:38. [PMID: 23497090 PMCID: PMC3616926 DOI: 10.1186/1742-2094-10-38] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Accepted: 02/19/2013] [Indexed: 02/03/2023] Open
Abstract
OBJECTIVE Converging lines of evidence point to the existence of immune dysfunction in autism spectrum disorder (ASD), which could directly affect several key neurodevelopmental processes. Previous studies have shown higher cytokine levels in patients with autism compared with matched controls or subjects with other developmental disorders. In the current study, we used plasma-cytokine profiling for 25 discordant sibling pairs to evaluate whether these alterations occur within families with ASD. METHODS Plasma-cytokine profiling was conducted using an array-based multiplex sandwich ELISA for simultaneous quantitative measurement of 40 unique targets. We also analyzed the correlations between cytokine levels and clinically relevant quantitative traits (Vineland Adaptive Behavior Scale in Autism (VABS) composite score, Social Responsiveness Scale (SRS) total T score, head circumference, and full intelligence quotient (IQ)). In addition, because of the high phenotypic heterogeneity of ASD, we defined four subgroups of subjects (those who were non-verbal, those with gastrointestinal issues, those with regressive autism, and those with a history of allergies), which encompass common and/or recurrent endophenotypes in ASD, and tested the cytokine levels in each group. RESULTS None of the measured parameters showed significant differences between children with ASD and their related typically developing siblings. However, specific target levels did correlate with quantitative clinical traits, and these were significantly different when the ASD subgroups were analyzed. It is notable that these differences seem to be attributable to a predisposing immunogenetic background, as no other significant differences were noticed between discordant sibling pairs. Interleukin-1β appears to be the cytokine most involved in quantitative traits and clinical subgroups of ASD. CONCLUSIONS In the present study, we found a lack of significant differences in plasma-cytokine levels between children with ASD and in their related non-autistic siblings. Thus, our results support the evidence that the immune profiles of children with autism do not differ from their typically developing siblings. However, the significant association of cytokine levels with the quantitative traits and the clinical subgroups analyzed suggests that altered immune responses may affect core feature of ASD.
Collapse
Affiliation(s)
- Valerio Napolioni
- Neurogenomics Division, The Translational Genomics Research Institute (TGen), 445 N Fifth Street, Phoenix, AZ 85004, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|