1
|
Ballal SA, Greenwell S, Liu E, Buie T, Silvester J, Leier M, Filippelli M, Bousvaros A, Hron B. Comparing Gastrointestinal Endoscopy Findings in Children with Autism, Developmental Delay, or Typical Development. J Pediatr 2024; 264:113737. [PMID: 37722553 PMCID: PMC10872435 DOI: 10.1016/j.jpeds.2023.113737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/11/2023] [Accepted: 09/13/2023] [Indexed: 09/20/2023]
Abstract
OBJECTIVE To compare endoscopic and histologic upper endoscopy (esophagogastroduodenoscopy [EGD]) findings in children with autism spectrum disorders (ASD) to age- and gender-matched controls with developmental delay (DD) or with typical development (TD). METHODS Retrospective, cross-sectional study of children undergoing EGD, identifying those diagnosed with ASD, and matching on age and gender to children with DD or TD in ratio of 1:1:2. Rates of EGD findings were compared between the 3 groups using χ² or Fisher exact test. Multivariable linear regression was performed to identify predictors of abnormal histology. RESULTS A total of 2104 patients were included (526 ASD; 526 DD; 1052 TD). Children with ASD had higher rates of abnormal esophageal histology (ASD 38.4%; DD 33.4%; TD 30.4%, P = .008), particularly esophagitis. In multivariable modeling, ASD diagnosis was an independent predictor of abnormal esophageal histology (OR [95% CI] 1.38 [1.09, 1.76]) compared with TD. Stomach findings did not differ among the groups. In the duodenum, histologic abnormalities were observed with lower frequency in ASD (ASD 17.0%; DD 20.1%; TD 24.2%, P = .005). In multivariable analysis, ASD diagnosis was not a significant predictor (OR 0.78 [0.56, 1.09]) of abnormal duodenal histology. CONCLUSIONS Children with ASD have higher rates of histologic esophagitis compared with age- and gender-matched DD and TD controls. ASD was a significant independent predictor of abnormal esophageal, but not, duodenal, histology. These results underscore the importance of EGD in children with ASD.
Collapse
Affiliation(s)
- Sonia A Ballal
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA.
| | - Saige Greenwell
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA
| | - Enju Liu
- Institutional Centers for Clinical and Translational Research, Boston Children's Hospital, Boston, MA
| | - Timothy Buie
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA
| | - Jocelyn Silvester
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA
| | - McKenzie Leier
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA
| | - Maura Filippelli
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA
| | - Athos Bousvaros
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA
| | - Bridget Hron
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA
| |
Collapse
|
2
|
Ashwood P. Preliminary Findings of Elevated Inflammatory Plasma Cytokines in Children with Autism Who Have Co-Morbid Gastrointestinal Symptoms. Biomedicines 2023; 11:436. [PMID: 36830973 PMCID: PMC9952966 DOI: 10.3390/biomedicines11020436] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 01/27/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Autism spectrum disorder (AU) is present in approximately 2% of the population and is often associated with co-morbidities that can impact quality of life. One of the most common co-morbidities in autism is the presence of gastrointestinal (GI) symptoms consisting of irregular bowel habits such as constipation, diarrhea, or alternating bowel habit. Evidence of immune infiltration and immune activation has been shown in the ileum and colon of children with AU with GI symptoms. Moreover, immune dysfunction is a contributing factor in many GI diseases, and we hypothesize that it would be more apparent in children with AU that exhibit GI symptoms than those who do not present with GI symptoms. The aim of this preliminary study was to determine whether there are altered cytokine levels in plasma in children with AU with GI symptoms compared with children with AU without GI symptoms, typically developing (TD) children with GI symptoms and TD children without GI symptoms, from the same population-based cohort. Plasma cytokine levels were assessed by multiplex assays. No differences in plasma cytokines were observed in TD controls with or without GI symptoms; however, many innate (IL-1α, TNFα, GM-CSF, IFNα) and adaptive cytokines (IL-4, IL-13, IL-12p70) were increased in AU children with GI symptoms compared with children with AU with no GI symptoms. The mucosal relevant cytokine IL-15 was increased in AU with GI symptoms compared with all groups. In contrast, the regulatory cytokine IL-10, was reduced in AU with GI symptoms and may suggest an imbalance in pro-inflammatory/regulatory signals. These data suggest that children with AU and GI symptoms have an imbalance in their immune response that is evident in their circulating plasma cytokine levels. A finding that could point to potential therapeutic and/or monitoring strategies for GI issues in AU.
Collapse
Affiliation(s)
- Paul Ashwood
- Department of Medical Microbiology and Immunology, School of Medicine, MIND Institute, University of California Davis, Davis, CA 95616, USA
| |
Collapse
|
3
|
Krigsman A, Walker SJ. Gastrointestinal disease in children with autism spectrum disorders: Etiology or consequence? World J Psychiatry 2021; 11:605-618. [PMID: 34631464 PMCID: PMC8474996 DOI: 10.5498/wjp.v11.i9.605] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 06/24/2021] [Accepted: 08/12/2021] [Indexed: 02/06/2023] Open
Abstract
Chronic gastrointestinal (GI) symptoms and disorders are common in children with autism spectrum disorder and have been shown to be significantly correlated with the degree of behavioral and cognitive impairment. In this unique population, GI symptoms often arise very early in development, during infancy or toddlerhood, and may be misdiagnosed - or not diagnosed at all – due in part to the challenges associated with recognition of symptoms in a minimally or non-communicative child. Evidence demonstrating that the gut-brain-axis can communicate gut dysbiosis and systemic immune dysregulation in a bidirectional manner raises the question as to whether an untreated gastrointestinal disorder can directly impact neurodevelopment or, conversely, whether having a neurodevelopmental disorder predisposes a child to chronic GI issues. From the data presented in this mini review, we conclude that the preponderance of available evidence would suggest the former scenario is more strongly supported.
Collapse
Affiliation(s)
- Arthur Krigsman
- Pediatric Gastroenterology Resources of New York and Texas, Georgetown, TX 78628, United States
| | - Stephen J Walker
- Institute for Regenerative Medicine, Wake Forest Baptist Medical Center, Winston Salem, NC 27157, United States
| |
Collapse
|
4
|
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
|
5
|
Rose D, Ashwood P. Rapid Communication: Plasma Interleukin-35 in Children with Autism. Brain Sci 2019; 9:E152. [PMID: 31252635 PMCID: PMC6680732 DOI: 10.3390/brainsci9070152] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 06/21/2019] [Accepted: 06/24/2019] [Indexed: 12/29/2022] Open
Abstract
In autism spectrum disorders (ASD) many individuals have co-morbid immune dysregulation that can lead to inflammation in the brain and periphery. The novel cytokine interleukin (IL)-35 has described anti-inflammatory properties; however, the plasma levels of IL-35 in children with ASD have never been investigated. The plasma levels of IL-35 were measured by an enzyme-linked immunosorbent assay in 30 children with ASD and 39 typically developing (TD) controls. In the current study, we found that plasma IL-35 levels were significantly decreased in children with ASD compared with TD children. Furthermore, lower IL-35 levels were associated with worse behaviors as assessed using the aberrant behavior checklist. These findings are in line with other observations of decreased regulatory cytokines such as transforming growth factor beta and IL-10 in ASD, and associations with severity of behaviors. In conclusion, regulating the expression of IL-35 may provide a new possible target for the treatment of immune issues in ASD to address an imbalance between pro- and anti-inflammatory signals that alter the behavioral phenotype.
Collapse
Affiliation(s)
- Destanie Rose
- Department of Medical Microbiology and Immunology, and The Medical Investigation of Neurodevelopmental Disorders Institute, University of California, Davis, CA 95817, USA
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, and The Medical Investigation of Neurodevelopmental Disorders Institute, University of California, Davis, CA 95817, USA.
| |
Collapse
|
6
|
Walker SJ, Langefeld CD, Zimmerman K, Schwartz MZ, Krigsman A. A molecular biomarker for prediction of clinical outcome in children with ASD, constipation, and intestinal inflammation. Sci Rep 2019; 9:5987. [PMID: 30979947 PMCID: PMC6461625 DOI: 10.1038/s41598-019-42568-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 04/03/2019] [Indexed: 12/22/2022] Open
Abstract
In children with autism spectrum disorder (ASD) who present to the gastroenterologist with chronic constipation on a background of colonic inflammation, we have identified two distinct clinical subtypes: (1) patients who experience a sustained state of GI symptomatic remission while on maintenance anti-inflammatory therapy (fast responders) and, (2) those with recurrent right-sided fecal loading requiring regular colon cleanouts during treatment for enterocolitis (slow responders). We hypothesized that a detailed molecular analysis of tissue from the affected region of the colon would provide mechanistic insights regarding the fast versus slow response to anti-inflammatory therapy. To test this, ascending colon biopsy tissues from 35 children with ASD (20 slow responders and 15 fast responders) were analyzed by RNAseq. Hierarchical cluster analysis was performed to assign samples to clusters and gene expression analysis was performed to identify differentially expressed transcripts (DETs) between samples within the clusters. Significant differences were found between the two clusters with fast responder-predominant cluster showing an upregulation of transcripts involved in the activation of immune and inflammatory response and the slow responder-predominant cluster showing significant over-representation of pathways impacting colonic motility (e.g. genes involved in tryptophan and serotonin degradation and mitochondrial dysfunction). Regression analysis identified a single long non-coding RNA that could predict cluster assignment with a high specificity (0.88), sensitivity (0.89) and accuracy (0.89). Comparison of gene expression profiles in the ascending colon from a subset of patients with ASD, chronic right-sided fecal loading constipation and a slow versus fast response to therapy has identified molecular mechanisms that likely contribute to this differential response following the primary therapeutic intervention (i.e. treatment for colonic inflammation with brief induction immunosuppression followed by maintenance non-steroidal anti-inflammatory therapy). Importantly, we have identified a transcript that, if validated, may provide a biomarker that can predict from the outset which patients will be slow responders who would benefit from an alternate therapeutic strategy in treating their constipation.
Collapse
Affiliation(s)
- Stephen J Walker
- Wake Forest University Health Sciences, Winston-Salem, North Carolina, USA.
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, North Carolina, USA.
| | - Carl D Langefeld
- Wake Forest University Health Sciences, Winston-Salem, North Carolina, USA
- Department of Biostatistical Sciences, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Kip Zimmerman
- Wake Forest University Health Sciences, Winston-Salem, North Carolina, USA
- Department of Biostatistical Sciences, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Marshall Z Schwartz
- Wake Forest University Health Sciences, Winston-Salem, North Carolina, USA
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, North Carolina, USA
| | - Arthur Krigsman
- Pediatric Gastroenterology Resources of New York and Texas, Austin, Texas, USA
| |
Collapse
|
7
|
Azhari A, Azizan F, Esposito G. A systematic review of gut-immune-brain mechanisms in Autism Spectrum Disorder. Dev Psychobiol 2018; 61:752-771. [PMID: 30523646 DOI: 10.1002/dev.21803] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 09/10/2018] [Accepted: 10/09/2018] [Indexed: 12/13/2022]
Abstract
Despite decades of research, the etiological origins of Autism Spectrum Disorder (ASD) remain elusive. Recently, the mechanisms of ASD have encompassed emerging theories involving the gastrointestinal, immune, and nervous systems. While each of these perspectives presents its own set of supporting evidence, the field requires an integration of these modular concepts and an overarching view of how these subsystems intersect. In this systematic review, we have synthesized relevant evidences from the existing literature, evaluating them in an interdependent manner and in doing so, outlining their possible connections. Specifically, we first discussed gastrointestinal and immuno-inflammation pathways in-depth, exploring the relationships between microbial composition, bacterial metabolites, gut mucosa, and immune system constituents. Accounting for temporal differences in the mechanisms involved in neurodevelopment, prenatal and postnatal phases were further elucidated, where the former focused on maternal immune activation (MIA) and fetal development, while the latter addressed the role of immune dysregulation in contributing to atypical neurodevelopment. As autism remains, foremost, a neurodevelopmental disorder, this review presents an integration of disparate modules into a "Gut-Immune-Brain" paradigm. Existing gaps in the literature have been highlighted, and possible avenues for future research with an integrated physiological perspective underlying ASD have also been suggested.
Collapse
Affiliation(s)
- Atiqah Azhari
- Psychology Program, School of Social Sciences, Nanyang Technological University, Singapore, Singapore
| | - Farouq Azizan
- Psychology Program, School of Social Sciences, Nanyang Technological University, Singapore, Singapore
| | - Gianluca Esposito
- Psychology Program, School of Social Sciences, Nanyang Technological University, Singapore, Singapore.,Department of Psychology and Cognitive Science, University of Trento, Rovereto, TN, Italy
| |
Collapse
|
8
|
Severance EG, Yolken RH. Deciphering microbiome and neuroactive immune gene interactions in schizophrenia. Neurobiol Dis 2018; 135:104331. [PMID: 30471416 DOI: 10.1016/j.nbd.2018.11.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 10/29/2018] [Accepted: 11/19/2018] [Indexed: 02/07/2023] Open
Abstract
The body's microbiome represents an actively regulated network of novel mechanisms that potentially underlie the etiology and pathophysiology of a wide range of diseases. For complex brain disorders such as schizophrenia, understanding the cellular and molecular pathways that intersect the bidirectional gut-brain axis is anticipated to lead to new methods of treatment. The means by which the microbiome might differ across neuropsychiatric and neurological disorders are not known. Brain disorders as diverse as schizophrenia, major depression, Parkinson's disease and multiple sclerosis appear to share a common pathology of an imbalanced community of commensal microbiota, often measured in terms of a leaky gut phenotype accompanied by low level systemic inflammation. While environmental factors associated with these disease states might contribute to intestinal pathologies, products from a perturbed microbiome may also directly promote specific signs, symptoms and etiologies of individual disorders. We hypothesize that in schizophrenia, it is the putatively unique susceptibility related to genes that modulate the immune system and the gut-brain pleiotropy of these genes which leads to a particularly neuropathological response when challenged by a microbiome in dysbiosis. Consequences from exposure to this dysbiosis may occur during pre- or post-natal time periods and thus may interfere with normal neurodevelopment in those who are genetically predisposed. Here, we review the evidence from the literature which supports the idea that the intersection of the microbiome and immune gene susceptibility in schizophrenia is relevant etiologically and for disease progression. Figuring prominently at both ends of the gut-brain axis and at points in between are proteins encoded by genes found in the major histocompatibility complex (MHC), including select MHC as well as non-MHC complement pathway genes.
Collapse
Affiliation(s)
- Emily G Severance
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| | - Robert H Yolken
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
9
|
Bağ Ö, Alşen Güney S, Cevher Binici N, Tuncel T, Şahin A, Berksoy E, Ecevit Ç. Infant colic or early symptom of autism spectrum disorder? Pediatr Int 2018; 60:517-522. [PMID: 29573066 DOI: 10.1111/ped.13565] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 03/05/2018] [Accepted: 03/09/2018] [Indexed: 12/27/2022]
Abstract
BACKGROUND Gastrointestinal (GI) disorders are common in autism spectrum disorder (ASD). Infant colic (IC), the functional GI disorder of infancy, has not been evaluated in this patient group. The aim of this study was therefore to determine the rate of IC in ASD and investigate a possible association between ASD and IC. METHODS The subjects consisted of 100 ASD patients (mean age, 6.6 ± 3.5 years) and 100 healthy controls (mean age, 5.3 ± 2.8 years). The parents were questioned using the diagnostic criteria for infant colic for clinical research purposes defined in Rome IV to diagnose IC, retrospectively. The sample size was estimated using a maximum type I error probability of 5% (alpha) and a type II error of 20%. RESULTS The rate of IC was 16% and 17% in the ASD group and control group, respectively (P ˃ 0.05). Excessive crying with late onset and long duration in infants was defined as persistent crying. The rate of persistent crying was significantly higher in the ASD group than in the control group (32% vs 9%, P < 0.001). The relative risk of persistent crying was 4.40 in ASD. The likelihood of being misdiagnosed with IC in this group was 78%. CONCLUSION The rate of IC is not increased in patients with ASD, but infants with excessive crying should be very thoroughly evaluated before being diagnosed with IC. In particular, persistent crying in infants (i.e. excessive crying with late onset and long duration) may be an early symptom of ASD.
Collapse
Affiliation(s)
- Özlem Bağ
- Department of Social Pediatrics, Dr Behçet Uz Children's Hospital, İzmir, Turkey
| | - Sevay Alşen Güney
- Department of Child and Adolescent Psychiatry, Dr Behçet Uz Children's Hospital, İzmir, Turkey
| | - Nagihan Cevher Binici
- Department of Child and Adolescent Psychiatry, Dr Behçet Uz Children's Hospital, İzmir, Turkey
| | - Tuba Tuncel
- Department of Pediatrics, İzmir Katip Çelebi University Medical Faculty, Tepecik Teaching and Research Hospital, İzmir, Turkey
| | - Aslıhan Şahin
- Department of General Pediatrics, Dr Behçet Uz Children's Hospital, İzmir, Turkey
| | - Emel Berksoy
- Department of Pediatrics, Tepecik Teaching and Research Hospital, İzmir, Turkey
| | - Çiğdem Ecevit
- Department of Pediatric Gastroenterology, Dr Behçet Uz Children's Hospital, İzmir, Turkey
| |
Collapse
|
10
|
Rose DR, Yang H, Serena G, Sturgeon C, Ma B, Careaga M, Hughes HK, Angkustsiri K, Rose M, Hertz-Picciotto I, Van de Water J, Hansen RL, Ravel J, Fasano A, Ashwood P. Differential immune responses and microbiota profiles in children with autism spectrum disorders and co-morbid gastrointestinal symptoms. Brain Behav Immun 2018; 70:354-368. [PMID: 29571898 PMCID: PMC5953830 DOI: 10.1016/j.bbi.2018.03.025] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 03/08/2018] [Accepted: 03/19/2018] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES Many studies have reported the increased presence of gastrointestinal (GI) symptoms in children with autism spectrum disorders (ASD). Altered microbiome profiles, pro-inflammatory responses and impaired intestinal permeability have been observed in children with ASD and co-morbid GI symptoms, yet few studies have compared these findings to ASD children without GI issues or similarly aged typical developing children. The aim of this study was to determine whether there are biological signatures in terms of immune dysfunction and microbiota composition in children with ASD with GI symptoms. METHODS Children were enrolled in one of four groups: ASD and GI symptoms of irregular bowel habits (ASDGI), children with ASD but without current or previous GI symptoms (ASDNoGI), typically developing children with GI symptoms (TDGI) and typically developing children without current or previous GI symptoms (TDNoGI). Peripheral blood mononuclear cells (PBMC) were isolated from the blood, stimulated and assessed for cytokine production, while stool samples were analyzed for microbial composition. RESULTS Following Toll-Like receptor (TLR)-4 stimulation, the ASDGI group produced increased levels of mucosa-relevant cytokines including IL-5, IL-15 and IL-17 compared to ASDNoGI. The production of the regulatory cytokine TGFβ1 was decreased in the ASDGI group compared with both the ASDNoGI and TDNoGI groups. Analysis of the microbiome at the family level revealed differences in microbiome composition between ASD and TD children with GI symptoms; furthermore, a predictive metagenome functional content analysis revealed that pathways were differentially represented between ASD and TD subjects, independently of the presence of GI symptoms. The ASDGI also showed an over-representation of the gene encoding zonulin, a molecule regulating gut permeability, compared to the other groups. CONCLUSIONS Overall our findings suggest that children with ASD who experience GI symptoms have an imbalance in their immune response, possibly influenced by or influencing metagenomic changes, and may have a propensity to impaired gut barrier function which may contribute to their symptoms and clinical outcome.
Collapse
Affiliation(s)
- Destanie R Rose
- Department of Medical Microbiology and Immunology, University of California Davis, USA; MIND Institute, University of California Davis, USA
| | - Houa Yang
- Department of Medical Microbiology and Immunology, University of California Davis, USA; MIND Institute, University of California Davis, USA
| | - Gloria Serena
- Division of Pediatric Gastroenterology and Nutrition, Center for Celiac Research, MassGeneral Hospital for Children, Boston, MA, USA; Graduate Program in Life Sciences University of Maryland School of Medicine, Baltimore, MD, USA
| | - Craig Sturgeon
- Division of Pediatric Gastroenterology and Nutrition, Center for Celiac Research, MassGeneral Hospital for Children, Boston, MA, USA; Graduate Program in Life Sciences University of Maryland School of Medicine, Baltimore, MD, USA
| | - Bing Ma
- Institute of Genomic Science, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Milo Careaga
- Department of Medical Microbiology and Immunology, University of California Davis, USA; MIND Institute, University of California Davis, USA
| | - Heather K Hughes
- Department of Medical Microbiology and Immunology, University of California Davis, USA; MIND Institute, University of California Davis, USA
| | - Kathy Angkustsiri
- MIND Institute, University of California Davis, USA; Department of Pediatrics, University of California Davis, USA; Children's Center for Environmental Health, University of California Davis, CA, USA
| | - Melissa Rose
- Children's Center for Environmental Health, University of California Davis, CA, USA; Public Health Sciences, University of California Davis, CA, USA
| | - Irva Hertz-Picciotto
- MIND Institute, University of California Davis, USA; Children's Center for Environmental Health, University of California Davis, CA, USA; Public Health Sciences, University of California Davis, CA, USA
| | - Judy Van de Water
- MIND Institute, University of California Davis, USA; Children's Center for Environmental Health, University of California Davis, CA, USA; Division of Rheumatology, Allergy and Clinical Immunology, University of California Davis, CA, USA
| | - Robin L Hansen
- MIND Institute, University of California Davis, USA; Department of Pediatrics, University of California Davis, USA; Children's Center for Environmental Health, University of California Davis, CA, USA
| | - Jacques Ravel
- Institute of Genomic Science, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alessio Fasano
- Division of Pediatric Gastroenterology and Nutrition, Center for Celiac Research, MassGeneral Hospital for Children, Boston, MA, USA
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, University of California Davis, USA; MIND Institute, University of California Davis, USA; Children's Center for Environmental Health, University of California Davis, CA, USA.
| |
Collapse
|
11
|
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
|
12
|
Extracerebral Dysfunction in Animal Models of Autism Spectrum Disorder. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2017; 224:159-187. [PMID: 28551756 DOI: 10.1007/978-3-319-52498-6_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Genetic factors might be largely responsible for the development of autism spectrum disorder (ASD) that alone or in combination with specific environmental risk factors trigger the pathology. Multiple mutations identified in ASD patients that impair synaptic function in the central nervous system are well studied in animal models. How these mutations might interact with other risk factors is not fully understood though. Additionally, how systems outside of the brain are altered in the context of ASD is an emerging area of research. Extracerebral influences on the physiology could begin in utero and contribute to changes in the brain and in the development of other body systems and further lead to epigenetic changes. Therefore, multiple recent studies have aimed at elucidating the role of gene-environment interactions in ASD. Here we provide an overview on the extracerebral systems that might play an important associative role in ASD and review evidence regarding the potential roles of inflammation, trace metals, metabolism, genetic susceptibility, enteric nervous system function and the microbiota of the gastrointestinal (GI) tract on the development of endophenotypes in animal models of ASD. By influencing environmental conditions, it might be possible to reduce or limit the severity of ASD pathology.
Collapse
|
13
|
Fiorentino M, Sapone A, Senger S, Camhi SS, Kadzielski SM, Buie TM, Kelly DL, Cascella N, Fasano A. Blood-brain barrier and intestinal epithelial barrier alterations in autism spectrum disorders. Mol Autism 2016; 7:49. [PMID: 27957319 PMCID: PMC5129651 DOI: 10.1186/s13229-016-0110-z] [Citation(s) in RCA: 305] [Impact Index Per Article: 33.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 11/12/2016] [Indexed: 12/11/2022] Open
Abstract
Background Autism spectrum disorders (ASD) are complex conditions whose pathogenesis may be attributed to gene–environment interactions. There are no definitive mechanisms explaining how environmental triggers can lead to ASD although the involvement of inflammation and immunity has been suggested. Inappropriate antigen trafficking through an impaired intestinal barrier, followed by passage of these antigens or immune-activated complexes through a permissive blood–brain barrier (BBB), can be part of the chain of events leading to these disorders. Our goal was to investigate whether an altered BBB and gut permeability is part of the pathophysiology of ASD. Methods Postmortem cerebral cortex and cerebellum tissues from ASD, schizophrenia (SCZ), and healthy subjects (HC) and duodenal biopsies from ASD and HC were analyzed for gene and protein expression profiles. Tight junctions and other key molecules associated with the neurovascular unit integrity and function and neuroinflammation were investigated. Results Claudin (CLDN)-5 and -12 were increased in the ASD cortex and cerebellum. CLDN-3, tricellulin, and MMP-9 were higher in the ASD cortex. IL-8, tPA, and IBA-1 were downregulated in SCZ cortex; IL-1b was increased in the SCZ cerebellum. Differences between SCZ and ASD were observed for most of the genes analyzed in both brain areas. CLDN-5 protein was increased in ASD cortex and cerebellum, while CLDN-12 appeared reduced in both ASD and SCZ cortexes. In the intestine, 75% of the ASD samples analyzed had reduced expression of barrier-forming TJ components (CLDN-1, OCLN, TRIC), whereas 66% had increased pore-forming CLDNs (CLDN-2, -10, -15) compared to controls. Conclusions In the ASD brain, there is an altered expression of genes associated with BBB integrity coupled with increased neuroinflammation and possibly impaired gut barrier integrity. While these findings seem to be specific for ASD, the possibility of more distinct SCZ subgroups should be explored with additional studies.
Collapse
Affiliation(s)
- Maria Fiorentino
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Boston, MA USA ; Department of Pediatrics, Harvard Medical School, Boston, MA USA
| | - Anna Sapone
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Boston, MA USA ; Department of Medicine, Celiac Center, Division of Gastroenterology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA USA
| | - Stefania Senger
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Boston, MA USA ; Department of Pediatrics, Harvard Medical School, Boston, MA USA
| | - Stephanie S Camhi
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Boston, MA USA ; Center for Celiac Research and Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital for Children, Boston, MA USA
| | | | - Timothy M Buie
- Department of Pediatrics, Harvard Medical School, Boston, MA USA
| | - Deanna L Kelly
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD USA
| | - Nicola Cascella
- Neuropsychiatry Program, Sheppard Pratt Health System, Baltimore, MD USA
| | - Alessio Fasano
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Boston, MA USA ; Center for Celiac Research and Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital for Children, Boston, MA USA ; Department of Pediatrics, Harvard Medical School, Boston, MA USA
| |
Collapse
|
14
|
Walker SJ, Beavers DP, Fortunato J, Krigsman A. A Putative Blood-Based Biomarker for Autism Spectrum Disorder-Associated Ileocolitis. Sci Rep 2016; 6:35820. [PMID: 27767057 PMCID: PMC5073317 DOI: 10.1038/srep35820] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 10/06/2016] [Indexed: 12/11/2022] Open
Abstract
Gastrointestinal symptoms are common in children with autism spectrum disorder (ASD). A significant proportion of children with ASD and gastrointestinal symptoms have histologic evidence of ileocolitis (inflammation of the terminal ileum and/or colon). We previously reported the molecular characterization of gastrointestinal biopsy tissue from ASD children with ileocolitis (ASDIC+) compared to anatomically similar inflamed tissue from typically developing children with inflammatory bowel disease (IBD; i.e. Crohn’s disease or ulcerative colitis) and typically developing children with gastrointestinal symptoms but no evidence of gastrointestinal mucosal inflammation (TDIC−). ASDIC+ children had a gene expression profile that, while primarily overlapping with known IBD, had distinctive differences. The present study confirms these findings and replicates this molecular characterization in a second cohort of cases (ASDIC+) and controls (TDIC−). In these two separate case/control mucosal-based cohorts, we have demonstrated overlap of 59 differentially expressed transcripts (DETs) unique to inflamed ileocolonic tissue from symptomatic ASDIC+ children. We now report that 9 of these 59 transcripts are also differentially expressed in the peripheral blood of the second cohort of ASDIC+ children. This set of transcripts represents a putative blood-based biomarker for ASD-associated ileocolonic inflammation.
Collapse
Affiliation(s)
- Stephen J Walker
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston Salem, NC, USA
| | - Daniel P Beavers
- Department of Biostatistical Sciences, Public Health Sciences, Wake Forest University Health Sciences, Winston Salem, NC, USA
| | - John Fortunato
- Pediatric Gastroenterology, Hepatology, and Nutrition, Ann &Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Arthur Krigsman
- Pediatric Gastroenterology Resources, 148 Beach 9th Street, Suite 2B, Far Rockaway, NY, USA
| |
Collapse
|
15
|
Severance EG, Yolken RH, Eaton WW. Autoimmune diseases, gastrointestinal disorders and the microbiome in schizophrenia: more than a gut feeling. Schizophr Res 2016; 176:23-35. [PMID: 25034760 PMCID: PMC4294997 DOI: 10.1016/j.schres.2014.06.027] [Citation(s) in RCA: 162] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 06/17/2014] [Accepted: 06/19/2014] [Indexed: 12/12/2022]
Abstract
Autoimmunity, gastrointestinal (GI) disorders and schizophrenia have been associated with one another for a long time. This paper reviews these connections and provides a context by which multiple risk factors for schizophrenia may be related. Epidemiological studies strongly link schizophrenia with autoimmune disorders including enteropathic celiac disease. Exposure to wheat gluten and bovine milk casein also contribute to non-celiac food sensitivities in susceptible individuals. Co-morbid GI inflammation accompanies humoral immunity to food antigens, occurs early during the course of schizophrenia and appears to be independent from antipsychotic-generated motility effects. This inflammation impacts endothelial barrier permeability and can precipitate translocation of gut bacteria into systemic circulation. Infection by the neurotropic gut pathogen, Toxoplasma gondii, will elicit an inflammatory GI environment. Such processes trigger innate immunity, including activation of complement C1q, which also functions at synapses in the brain. The emerging field of microbiome research lies at the center of these interactions with evidence that the abundance and diversity of resident gut microbiota contribute to digestion, inflammation, gut permeability and behavior. Dietary modifications of core bacterial compositions may explain inefficient gluten digestion and how immigrant status in certain situations is a risk factor for schizophrenia. Gut microbiome research in schizophrenia is in its infancy, but data in related fields suggest disease-associated altered phylogenetic compositions. In summary, this review surveys associative and experimental data linking autoimmunity, GI activity and schizophrenia, and proposes that understanding of disrupted biological pathways outside of the brain can lend valuable information regarding pathogeneses of complex, polygenic brain disorders.
Collapse
Affiliation(s)
- Emily G. Severance
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Blalock 1105, Baltimore, MD 21287-4933 U.S.A
| | - Robert H. Yolken
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Blalock 1105, Baltimore, MD 21287-4933 U.S.A
| | - William W. Eaton
- Department of Mental Health, Bloomberg School of Public Health, The Johns Hopkins University, Baltimore, MD, U.S.A
| |
Collapse
|
16
|
Fond G, Chevalier G, Eberl G, Leboyer M. [The potential role of microbiota in major psychiatric disorders: Mechanisms, preclinical data, gastro-intestinal comorbidities and therapeutic options]. Presse Med 2015; 45:7-19. [PMID: 26653939 DOI: 10.1016/j.lpm.2015.10.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 09/10/2015] [Accepted: 10/29/2015] [Indexed: 12/21/2022] Open
Abstract
While forecasts predict an increase in the prevalence of mental health disorders in the worldwide general population, the response rate to classical psychiatric treatment remains unsatisfactory. Resistance to psychotropic drugs can be due to clinical, pharmacological, pharmacokinetic, and pharmacodynamic factors. Among these factors, recent animal findings suggest that microbiota may have an underestimated influence on its host's behavior and on drug metabolism that may explain ineffectiveness or increased side effects of psychiatric medications such as weight gain. The following issues were identified in the present review: (i) microbiota dysbiosis and putative consequences on central nervous system functioning; (ii) chronic microbiota dysbiosis-associated illnesses in humans; (iii) microbiota-oriented treatments and their potential therapeutic applications in psychiatry.
Collapse
Affiliation(s)
- Guillaume Fond
- Inserm U955, équipe 15, université Paris-Est, fondation FondaMental, fondation de coopération scientifique, AP-HP, groupe hospitalo-universitaire Mondor, DHU Pe-Psy, hôpital A.-Chenevier, pôle de psychiatrie et d'addictologie, pavillon Hartmann, 40, rue de Mesly, 94000 Créteil, France.
| | - Grégoire Chevalier
- Institut Pasteur, unité de développement du tissu lymphoïde, 25, rue du Dr-Roux, 75724 Paris, France
| | - Gerard Eberl
- Institut Pasteur, unité de développement du tissu lymphoïde, 25, rue du Dr-Roux, 75724 Paris, France
| | - Marion Leboyer
- Inserm U955, équipe 15, université Paris-Est, fondation FondaMental, fondation de coopération scientifique, AP-HP, groupe hospitalo-universitaire Mondor, DHU Pe-Psy, hôpital A.-Chenevier, pôle de psychiatrie et d'addictologie, pavillon Hartmann, 40, rue de Mesly, 94000 Créteil, France
| |
Collapse
|
17
|
Abstract
BACKGROUND The objective of this study was to measure the prevalence of inflammatory bowel disease (IBD) among patients with autism spectrum disorders (ASD), which has not been well described previously. METHODS The rates of IBD among patients with and without ASD were measured in 4 study populations with distinct modes of ascertainment: a health care benefits company, 2 pediatric tertiary care centers, and a national ASD repository. The rates of IBD (established through International Classification of Diseases, Ninth Revision, Clinical Modification [ICD-9-CM] codes) were compared with respective controls and combined using a Stouffer meta-analysis. Clinical charts were also reviewed for IBD among patients with ICD-9-CM codes for both IBD and ASD at one of the pediatric tertiary care centers. This expert-verified rate was compared with the rate in the repository study population (where IBD diagnoses were established by expert review) and in nationally reported rates for pediatric IBD. RESULTS In all of case-control study populations, the rates of IBD-related ICD-9-CM codes for patients with ASD were significantly higher than that of their respective controls (Stouffer meta-analysis, P < 0.001). Expert-verified rates of IBD among patients with ASD were 7 of 2728 patients in one study population and 16 of 7201 in a second study population. The age-adjusted prevalence of IBD among patients with ASD was higher than their respective controls and nationally reported rates of pediatric IBD. CONCLUSIONS Across each population with different kinds of ascertainment, there was a consistent and statistically significant increased prevalance of IBD in patients with ASD than their respective controls and nationally reported rates for pediatric IBD.
Collapse
|
18
|
Bank I, Marcu-Malina V. Quantitative peripheral blood perturbations of γδ T cells in human disease and their clinical implications. Clin Rev Allergy Immunol 2015; 47:311-33. [PMID: 24126758 DOI: 10.1007/s12016-013-8391-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Human γδ T cells, which play innate and adaptive, protective as well as destructive, roles in the immune response, were discovered in 1986, but the clinical significance of alterations of the levels of these cells in the peripheral blood in human diseases has not been comprehensively reviewed. Here, we review patterns of easily measurable changes of this subset of T cells in peripheral blood from relevant publications in PubMed and their correlations with specific disease categories, specific diagnoses within disease categories, and prognostic outcomes. These collective data suggest that enumeration of γδ T cells and their subsets in the peripheral blood of patients could be a useful tool to evaluate diagnosis and prognosis in the clinical setting.
Collapse
Affiliation(s)
- Ilan Bank
- Department of Medicine F, Chaim Sheba Medical Center, Tel Hashomer, Ramat Gan, 52621, Israel,
| | | |
Collapse
|
19
|
Fond G, Boukouaci W, Chevalier G, Regnault A, Eberl G, Hamdani N, Dickerson F, Macgregor A, Boyer L, Dargel A, Oliveira J, Tamouza R, Leboyer M. The "psychomicrobiotic": Targeting microbiota in major psychiatric disorders: A systematic review. ACTA ACUST UNITED AC 2014; 63:35-42. [PMID: 25468489 DOI: 10.1016/j.patbio.2014.10.003] [Citation(s) in RCA: 140] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2014] [Accepted: 10/20/2014] [Indexed: 12/19/2022]
Abstract
The gut microbiota is increasingly considered as a symbiotic partner in the maintenance of good health. Metagenomic approaches could help to discover how the complex gut microbial ecosystem participates in the control of the host's brain development and function, and could be relevant for future therapeutic developments, such as probiotics, prebiotics and nutritional approaches for psychiatric disorders. Previous reviews focused on the effects of microbiota on the central nervous system in in vitro and animal studies. The aim of the present review is to synthetize the current data on the association between microbiota dysbiosis and onset and/or maintenance of major psychiatric disorders, and to explore potential therapeutic opportunities targeting microbiota dysbiosis in psychiatric patients.
Collapse
Affiliation(s)
- G Fond
- Inserm U955, FondaMental Foundation, Paris-Est university, Chenevier Hospital, AP-HP, GHU Mondor, DHU Pe-Psy, Pavillon Hartmann, 40, rue Mesly, 94000 Créteil, France.
| | - W Boukouaci
- Jean-Dausset Laboratory & Inserm, UMRS 940, Saint-Louis hospital, 1, avenue Claude-Vellefaux, 75010 Paris, France
| | - G Chevalier
- Unité de développement du tissu lymphoïde, Institut Pasteur, 25, rue du Dr. Roux, 75724 Paris, France
| | - A Regnault
- Inserm, Institut Pasteur, aviesan/institut multi-organismes immunologie, hématologie et pneumologie (ITMO IHP), bâtiment Biopark, 8, rue de la Croix Jarry 1(er) étage, 75013 Paris, France
| | - G Eberl
- Unité de développement du tissu lymphoïde, Institut Pasteur, 25, rue du Dr. Roux, 75724 Paris, France
| | - N Hamdani
- Inserm U955, FondaMental Foundation, Paris-Est university, Chenevier Hospital, AP-HP, GHU Mondor, DHU Pe-Psy, Pavillon Hartmann, 40, rue Mesly, 94000 Créteil, France
| | - F Dickerson
- Stanley Research Program, Sheppard Pratt Health System, 6501N, Charles Street, MD 21204 Baltimore, United States
| | - A Macgregor
- Inserm U1061, academic adult psychiatry department, Montpellier 1 university, La Colombière hospital, Montpellier CHRU, 191, avenue du doyen Gaston-Giraud, 34295 Montpellier cedex, France
| | - L Boyer
- EA 3279-Self-perceived Health Assessment Research Unit, School of Medicine, La Timone University, 27, boulevard Jean-Moulin, 13385 Marseille cedex 05, France
| | - A Dargel
- Inserm U955, FondaMental Foundation, Paris-Est university, Chenevier Hospital, AP-HP, GHU Mondor, DHU Pe-Psy, Pavillon Hartmann, 40, rue Mesly, 94000 Créteil, France
| | - J Oliveira
- Jean-Dausset Laboratory & Inserm, UMRS 940, Saint-Louis hospital, 1, avenue Claude-Vellefaux, 75010 Paris, France
| | - R Tamouza
- Jean-Dausset Laboratory & Inserm, UMRS 940, Saint-Louis hospital, 1, avenue Claude-Vellefaux, 75010 Paris, France
| | - M Leboyer
- Inserm U955, FondaMental Foundation, Paris-Est university, Chenevier Hospital, AP-HP, GHU Mondor, DHU Pe-Psy, Pavillon Hartmann, 40, rue Mesly, 94000 Créteil, France
| |
Collapse
|
20
|
Trent S, Fry JP, Ojarikre OA, Davies W. Altered brain gene expression but not steroid biochemistry in a genetic mouse model of neurodevelopmental disorder. Mol Autism 2014; 5:21. [PMID: 24602487 PMCID: PMC3946266 DOI: 10.1186/2040-2392-5-21] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 02/19/2014] [Indexed: 12/02/2022] Open
Abstract
Background The 39,XY*O mouse, which lacks the orthologues of the ADHD and autism candidate genes STS (steroid sulphatase) and ASMT (acetylserotonin O-methyltransferase), exhibits behavioural phenotypes relevant to developmental disorders. The neurobiology underlying these phenotypes is unclear, although there is evidence for serotonergic abnormalities in the striatum and hippocampus. Methods Using microarray and quantitative gene expression analyses, and gas chromatography–mass spectrometry, we compared brain gene expression and steroid biochemistry in wildtype (40,XY) and 39,XY*O adult mice to identify non-obvious genetic and endocrine candidates for between-group differences in behaviour and neurochemistry. We also tested whether acute STS inhibition by COUMATE in wildtype (40,XY) adult male mice recapitulated any significant gene expression or biochemical findings from the genetic comparison. Data were analysed by unpaired t-test or Mann Whitney U-test depending on normality, with a single factor of KARYOTYPE. Results Microarray analysis indicated seven robust gene expression differences between the two groups (Vmn2r86, Sfi1, Pisd-ps1, Tagap1, C1qc, Metap1d, Erdr1); Erdr1 and C1qc expression was significantly reduced in the 39,XY*O striatum and hippocampus, whilst the expression of Dhcr7 (encoding 7-dehydrocholesterol reductase, a modulator of serotonin system development), was only reduced in the 39,XY*O hippocampus. None of the confirmed gene expression changes could be recapitulated by COUMATE administration. We detected ten free, and two sulphated steroids in 40,XY and 39,XY*O brain; surprisingly, the concentrations of all of these were equivalent between groups. Conclusions Our data demonstrate that the mutation in 39,XY*O mice: i) directly disrupts expression of the adjacent Erdr1 gene, ii) induces a remarkably limited suite of downstream gene expression changes developmentally, with several of relevance to associated neurobehavioural phenotypes and iii) does not elicit large changes in brain steroid biochemistry. It is possible that individuals with STS/ASMT deficiency exhibit a similarly specific pattern of gene expression changes to the 39,XY*O mouse, and that these contribute towards their abnormal neurobiology. Future work may focus on whether complement pathway function, mitochondrial metabolism and cholesterol biosynthesis pathways are perturbed in such subjects.
Collapse
Affiliation(s)
| | | | | | - William Davies
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, UK.
| |
Collapse
|
21
|
Kim JW, Choi CS, Kim KC, Park JH, Seung H, Joo SH, Yang SM, Shin CY, Park SH. Gastrointestinal tract abnormalities induced by prenatal valproic Acid exposure in rat offspring. Toxicol Res 2014; 29:173-9. [PMID: 24386517 PMCID: PMC3877996 DOI: 10.5487/tr.2013.29.3.173] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 09/25/2013] [Accepted: 09/27/2013] [Indexed: 12/12/2022] Open
Abstract
In-utero exposure to valproic acid (VPA) has been known as a potent inducer of autism spectrum disorder (ASD), not only in humans, but also in animals. In addition to the defects in communication and social interaction as well as repetitive behaviors, ASD patients usually suffer from gastrointestinal (GI) problems. However, the exact mechanism underlying these disorders is not known. In this study, we examined the gross GI tract structure and GI motility in a VPA animal model of ASD. On embryonic day 12 (E12), 4 pregnant Sprague-Dawley (SD) rats were subcutaneously injected with VPA (400 mg/kg) in the treatment group, and with phosphate buffered saline (PBS) in the control group; the resulting male offspring were analyzed at 4 weeks of age. VPA exposure decreased the thickness of tunica mucosa and tunica muscularis in the stomach and ileum. Other regions such as duodenum, jejunum, and colon did not show a significant difference. In high-resolution microscopic observation, atrophy of the parietal and chief cells in the stomach and absorptive cells in the ileum was observed. In addition, decreased staining of the epithelial cells was observed in the hematoxylin and eosin (H&E)-stained ileum section. Furthermore, decreased motility in GI tract was also observed in rat offspring prenatally exposed to VPA. However, the mechanism underlying GI tract defects in VPA animal model as well as the association between abnormal GI structure and function with ASD is yet to be clearly understood. Nevertheless, the results from the present study suggest that this VPA ASD model undergoes abnormal changes in the GI structure and function, which in turn could provide beneficial clues pertaining to the pathophysiological relevance of GI complications and ASD phenotypes.
Collapse
Affiliation(s)
- Ji-Woon Kim
- Department of Neuroscience, School of Medicine and Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, Konkuk University, Korea
| | - Chang Soon Choi
- Department of Neuroscience, School of Medicine and Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, Konkuk University, Korea
| | - Ki Chan Kim
- Department of Neuroscience, School of Medicine and Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, Konkuk University, Korea
| | - Jin Hee Park
- Department of Neuroscience, School of Medicine and Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, Konkuk University, Korea
| | - Hana Seung
- Department of Neuroscience, School of Medicine and Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, Konkuk University, Korea
| | - So Hyun Joo
- Department of Neuroscience, School of Medicine and Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, Konkuk University, Korea
| | - Sung Min Yang
- Department of Neuroscience, School of Medicine and Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, Konkuk University, Korea
| | - Chan Young Shin
- Department of Neuroscience, School of Medicine and Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, Konkuk University, Korea ; Institute of Functional Genomics, Konkuk University, Korea
| | - Seung Hwa Park
- Department of Neuroscience, School of Medicine and Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, Konkuk University, Korea ; Institute of Functional Genomics, Konkuk University, Korea
| |
Collapse
|
22
|
Walker SJ, Fortunato J, Gonzalez LG, Krigsman A. Identification of unique gene expression profile in children with regressive autism spectrum disorder (ASD) and ileocolitis. PLoS One 2013; 8:e58058. [PMID: 23520485 PMCID: PMC3592909 DOI: 10.1371/journal.pone.0058058] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 01/31/2013] [Indexed: 12/11/2022] Open
Abstract
Gastrointestinal symptoms are common in children with autism spectrum disorder (ASD) and are often associated with mucosal inflammatory infiltrates of the small and large intestine. Although distinct histologic and immunohistochemical properties of this inflammatory infiltrate have been previously described in this ASD(GI) group, molecular characterization of these lesions has not been reported. In this study we utilize transcriptome profiling of gastrointestinal mucosal biopsy tissue from ASD(GI) children and three non-ASD control groups (Crohn's disease, ulcerative colitis, and histologically normal) in an effort to determine if there is a gene expression profile unique to the ASD(GI) group. Comparison of differentially expressed transcripts between the groups demonstrated that non-pathologic (normal) tissue segregated almost completely from inflamed tissue in all cases. Gene expression profiles in intestinal biopsy tissue from patients with Crohn's disease, ulcerative colitis, and ASD(GI), while having significant overlap with each other, also showed distinctive features for each group. Taken together, these results demonstrate that ASD(GI) children have a gastrointestinal mucosal molecular profile that overlaps significantly with known inflammatory bowel disease (IBD), yet has distinctive features that further supports the presence of an ASD-associated IBD variant, or, alternatively, a prodromal phase of typical inflammatory bowel disease. Although we report qPCR confirmation of representative differentially expressed transcripts determined initially by microarray, these findings may be considered preliminary to the extent that they require further confirmation in a validation cohort.
Collapse
Affiliation(s)
- Stephen J Walker
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston Salem, North Carolina, United States of America.
| | | | | | | |
Collapse
|
23
|
Coury DL, Ashwood P, Fasano A, Fuchs G, Geraghty M, Kaul A, Mawe G, Patterson P, Jones NE. Gastrointestinal conditions in children with autism spectrum disorder: developing a research agenda. Pediatrics 2012; 130 Suppl 2:S160-8. [PMID: 23118247 DOI: 10.1542/peds.2012-0900n] [Citation(s) in RCA: 160] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Daniel L Coury
- Developmental/Behavioral Pediatrics, Nationwide Children's Hospital, Columbus, OH 43205-2696, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Randolph-Gips M, Srinivasan P. Modeling autism: a systems biology approach. J Clin Bioinforma 2012; 2:17. [PMID: 23043674 PMCID: PMC3507704 DOI: 10.1186/2043-9113-2-17] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Accepted: 08/09/2012] [Indexed: 12/13/2022] Open
Abstract
Autism is the fastest growing developmental disorder in the world today. The prevalence of autism in the US has risen from 1 in 2500 in 1970 to 1 in 88 children today. People with autism present with repetitive movements and with social and communication impairments. These impairments can range from mild to profound. The estimated total lifetime societal cost of caring for one individual with autism is $3.2 million US dollars. With the rapid growth in this disorder and the great expense of caring for those with autism, it is imperative for both individuals and society that techniques be developed to model and understand autism. There is increasing evidence that those individuals diagnosed with autism present with highly diverse set of abnormalities affecting multiple systems of the body. To this date, little to no work has been done using a whole body systems biology approach to model the characteristics of this disorder. Identification and modelling of these systems might lead to new and improved treatment protocols, better diagnosis and treatment of the affected systems, which might lead to improved quality of life by themselves, and, in addition, might also help the core symptoms of autism due to the potential interconnections between the brain and nervous system with all these other systems being modeled. This paper first reviews research which shows that autism impacts many systems in the body, including the metabolic, mitochondrial, immunological, gastrointestinal and the neurological. These systems interact in complex and highly interdependent ways. Many of these disturbances have effects in most of the systems of the body. In particular, clinical evidence exists for increased oxidative stress, inflammation, and immune and mitochondrial dysfunction which can affect almost every cell in the body. Three promising research areas are discussed, hierarchical, subgroup analysis and modeling over time. This paper reviews some of the systems disturbed in autism and suggests several systems biology research areas. Autism poses a rich test bed for systems biology modeling techniques.
Collapse
Affiliation(s)
- Mary Randolph-Gips
- Systems Engineering and Computer Engineering, University of Houston - Clear Lake, 2700 Bay Area Bvd, Houston, TX, 77058, USA.
| | | |
Collapse
|
25
|
The role of immune dysfunction in the pathophysiology of autism. Brain Behav Immun 2012; 26:383-92. [PMID: 21906670 PMCID: PMC3418145 DOI: 10.1016/j.bbi.2011.08.007] [Citation(s) in RCA: 448] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Revised: 08/19/2011] [Accepted: 08/22/2011] [Indexed: 12/22/2022] Open
Abstract
Autism spectrum disorders (ASD) are a complex group of neurodevelopmental disorders encompassing impairments in communication, social interactions and restricted stereotypical behaviors. Although a link between altered immune responses and ASD was first recognized nearly 40 years ago, only recently has new evidence started to shed light on the complex multifaceted relationship between immune dysfunction and behavior in ASD. Neurobiological research in ASD has highlighted pathways involved in neural development, synapse plasticity, structural brain abnormalities, cognition and behavior. At the same time, several lines of evidence point to altered immune dysfunction in ASD that directly impacts some or all these neurological processes. Extensive alterations in immune function have now been described in both children and adults with ASD, including ongoing inflammation in brain specimens, elevated pro-inflammatory cytokine profiles in the CSF and blood, increased presence of brain-specific auto-antibodies and altered immune cell function. Furthermore, these dysfunctional immune responses are associated with increased impairments in behaviors characteristic of core features of ASD, in particular, deficits in social interactions and communication. This accumulating evidence suggests that immune processes play a key role in the pathophysiology of ASD. This review will discuss the current state of our knowledge of immune dysfunction in ASD, how these findings may impact on underlying neuro-immune mechanisms and implicate potential areas where the manipulation of the immune response could have an impact on behavior and immunity in ASD.
Collapse
|
26
|
Abstract
Autism spectrum disorders (ASD) are complex and heterogeneous with a spectrum of diverse symptoms. Mounting evidence from a number of disciplines suggests a link between immune function and ASD. Although the causes of ASD have yet to be identified, genetic studies have uncovered a host of candidate genes relating to immune regulation that are altered in ASD, while epidemiological studies have shown a relationship with maternal immune disturbances during pregnancy and ASD. Moreover, decades of research have identified numerous systemic and cellular immune abnormalities in individuals with ASD and their families. These include changes in immune cell number, differences in cytokine and chemokine production, and alterations of cellular function at rest and in response to immunological challenge. Many of these changes in immune responses are associated with increasing impairment in behaviors that are core features of ASD. Despite this evidence, much remains to be understood about the precise mechanism by which the immune system alters neurodevelopment and to what extent it is involved in the pathogenesis of ASD. With estimates of ASD as high as 1% of children, ASD is a major public health issue. Improvements in our understanding of the interactions between the nervous and immune system during early neurodevelopment and how this interaction is different in ASD will have important therapeutic implications with wide ranging benefits.
Collapse
Affiliation(s)
- Milo Careaga
- Department of Medical Microbiology and Immunology and the M.I.N.D. Institute, University of California at Davis, Davis, CA, USA
| | | |
Collapse
|
27
|
Adams JB, Audhya T, McDonough-Means S, Rubin RA, Quig D, Geis E, Gehn E, Loresto M, Mitchell J, Atwood S, Barnhouse S, Lee W. Effect of a vitamin/mineral supplement on children and adults with autism. BMC Pediatr 2011. [PMID: 22151477 DOI: 10.1186/1471–2431–11–111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Vitamin/mineral supplements are among the most commonly used treatments for autism, but the research on their use for treating autism has been limited. METHOD This study is a randomized, double-blind, placebo-controlled three month vitamin/mineral treatment study. The study involved 141 children and adults with autism, and pre and post symptoms of autism were assessed. None of the participants had taken a vitamin/mineral supplement in the two months prior to the start of the study. For a subset of the participants (53 children ages 5-16) pre and post measurements of nutritional and metabolic status were also conducted. RESULTS The vitamin/mineral supplement was generally well-tolerated, and individually titrated to optimum benefit. Levels of many vitamins, minerals, and biomarkers improved/increased showing good compliance and absorption. Statistically significant improvements in metabolic status were many including: total sulfate (+17%, p = 0.001), S-adenosylmethionine (SAM; +6%, p = 0.003), reduced glutathione (+17%, p = 0.0008), ratio of oxidized glutathione to reduced glutathione (GSSG:GSH; -27%, p = 0.002), nitrotyrosine (-29%, p = 0.004), ATP (+25%, p = 0.000001), NADH (+28%, p = 0.0002), and NADPH (+30%, p = 0.001). Most of these metabolic biomarkers improved to normal or near-normal levels.The supplement group had significantly greater improvements than the placebo group on the Parental Global Impressions-Revised (PGI-R, Average Change, p = 0.008), and on the subscores for Hyperactivity (p = 0.003), Tantrumming (p = 0.009), Overall (p = 0.02), and Receptive Language (p = 0.03). For the other three assessment tools the difference between treatment group and placebo group was not statistically significant.Regression analysis revealed that the degree of improvement on the Average Change of the PGI-R was strongly associated with several biomarkers (adj. R2 = 0.61, p < 0.0005) with the initial levels of biotin and vitamin K being the most significant (p < 0.05); both biotin and vitamin K are made by beneficial intestinal flora. CONCLUSIONS Oral vitamin/mineral supplementation is beneficial in improving the nutritional and metabolic status of children with autism, including improvements in methylation, glutathione, oxidative stress, sulfation, ATP, NADH, and NADPH. The supplement group had significantly greater improvements than did the placebo group on the PGI-R Average Change. This suggests that a vitamin/mineral supplement is a reasonable adjunct therapy to consider for most children and adults with autism. TRIAL REGISTRATION CLINICAL TRIAL REGISTRATION NUMBER NCT01225198.
Collapse
Affiliation(s)
- James B Adams
- Autism/Asperger's Research Program, Arizona State University, Tempe, AZ, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Adams JB, Audhya T, McDonough-Means S, Rubin RA, Quig D, Geis E, Gehn E, Loresto M, Mitchell J, Atwood S, Barnhouse S, Lee W. Effect of a vitamin/mineral supplement on children and adults with autism. BMC Pediatr 2011; 11:111. [PMID: 22151477 PMCID: PMC3266205 DOI: 10.1186/1471-2431-11-111] [Citation(s) in RCA: 150] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Accepted: 12/12/2011] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Vitamin/mineral supplements are among the most commonly used treatments for autism, but the research on their use for treating autism has been limited. METHOD This study is a randomized, double-blind, placebo-controlled three month vitamin/mineral treatment study. The study involved 141 children and adults with autism, and pre and post symptoms of autism were assessed. None of the participants had taken a vitamin/mineral supplement in the two months prior to the start of the study. For a subset of the participants (53 children ages 5-16) pre and post measurements of nutritional and metabolic status were also conducted. RESULTS The vitamin/mineral supplement was generally well-tolerated, and individually titrated to optimum benefit. Levels of many vitamins, minerals, and biomarkers improved/increased showing good compliance and absorption. Statistically significant improvements in metabolic status were many including: total sulfate (+17%, p = 0.001), S-adenosylmethionine (SAM; +6%, p = 0.003), reduced glutathione (+17%, p = 0.0008), ratio of oxidized glutathione to reduced glutathione (GSSG:GSH; -27%, p = 0.002), nitrotyrosine (-29%, p = 0.004), ATP (+25%, p = 0.000001), NADH (+28%, p = 0.0002), and NADPH (+30%, p = 0.001). Most of these metabolic biomarkers improved to normal or near-normal levels.The supplement group had significantly greater improvements than the placebo group on the Parental Global Impressions-Revised (PGI-R, Average Change, p = 0.008), and on the subscores for Hyperactivity (p = 0.003), Tantrumming (p = 0.009), Overall (p = 0.02), and Receptive Language (p = 0.03). For the other three assessment tools the difference between treatment group and placebo group was not statistically significant.Regression analysis revealed that the degree of improvement on the Average Change of the PGI-R was strongly associated with several biomarkers (adj. R2 = 0.61, p < 0.0005) with the initial levels of biotin and vitamin K being the most significant (p < 0.05); both biotin and vitamin K are made by beneficial intestinal flora. CONCLUSIONS Oral vitamin/mineral supplementation is beneficial in improving the nutritional and metabolic status of children with autism, including improvements in methylation, glutathione, oxidative stress, sulfation, ATP, NADH, and NADPH. The supplement group had significantly greater improvements than did the placebo group on the PGI-R Average Change. This suggests that a vitamin/mineral supplement is a reasonable adjunct therapy to consider for most children and adults with autism. TRIAL REGISTRATION CLINICAL TRIAL REGISTRATION NUMBER NCT01225198.
Collapse
Affiliation(s)
- James B Adams
- Autism/Asperger's Research Program, Arizona State University, Tempe, AZ, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
The potential role of probiotics in the management of childhood autism spectrum disorders. Gastroenterol Res Pract 2011; 2011:161358. [PMID: 22114588 PMCID: PMC3205659 DOI: 10.1155/2011/161358] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Accepted: 08/20/2011] [Indexed: 12/14/2022] Open
Abstract
Gastrointestinal (GI) dysfunction has been reported in a substantial number of children with autism spectrum disorders (ASD). Activation of the mucosal immune response and the presence of abnormal gut microbiota are repeatedly observed in these children. In children with ASD, the presence of GI dysfunction is often associated with increased irritability, tantrums, aggressive behaviour, and sleep disturbances. Moreover, modulating gut bacteria with short-term antibiotic treatment can lead to temporary improvement in behavioral symptoms in some individuals with ASD. Probiotics can influence microbiota composition and intestinal barrier function and alter mucosal immune responses. The administration of probiotic bacteria to address changes in the microbiota might, therefore, be a useful novel therapeutic tool with which to restore normal gut microbiota, reduce inflammation, restore epithelial barrier function, and potentially ameliorate behavioural symptoms associated with some children with ASD. In this review of the literature, support emerges for the clinical testing of probiotics in ASD, especially in the context of addressing GI symptoms.
Collapse
|
30
|
Brown AC, Mehl-Madrona L. Autoimmune and gastrointestinal dysfunctions: does a subset of children with autism reveal a broader connection? Expert Rev Gastroenterol Hepatol 2011; 5:465-77. [PMID: 21780894 DOI: 10.1586/egh.11.46] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
A large number of autoimmune disorders have a gastrointestinal (GI) dysfunction component that may interplay with genetic, hormonal, environmental and/or stress factors. This narrarive review investigates possible links between autism, immune system abnormalities and GI symptoms in a subgroup of children with autism. A literature search on Medline (1950 to September 2010) was conducted to identify relevant articles by using the keywords 'autism and gastrointestinal' (71 publications) and 'autism and immune' (237 publications), cross-referencing and general searching to evaluate the available literature on the immunological and GI aspects of autism. Sufficient evidence exists to support that a subgroup of children with autism may suffer from concomitant immune-related GI symptoms.
Collapse
Affiliation(s)
- Amy C Brown
- Department of Complementary & Alternative Medicine, John A Burns School of Medicine, University of Hawaii at Manoa, 651 Ilalo Street, Honolulu, HI 96813, USA.
| | | |
Collapse
|
31
|
The prevalence of gastrointestinal problems in children across the United States with autism spectrum disorders from families with multiple affected members. J Dev Behav Pediatr 2011; 32:351-60. [PMID: 21555957 DOI: 10.1097/dbp.0b013e31821bd06a] [Citation(s) in RCA: 159] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE : To perform a large registry-based study to determine the relative prevalence of gastrointestinal (GI) problems in children with an autism spectrum disorder (ASD) from families with multiple affected members compared with their unaffected sibling(s). METHODS : In-home structured retrospective medical history interviews by parent recall were conducted by a pediatric neurologist. Our analysis sample included information about GI health of 589 subjects with idiopathic, familial ASD and 163 of their unaffected sibling controls registered with Autism Genetic Resource Exchange. Individuals with ASD were subgrouped into 3 autism severity groups (Full Autism, Almost Autism, and Spectrum) based on their Autism Diagnostic Interview-Revised and Autism Diagnostic Observation Scale scores. RESULTS : Parents reported significantly more GI problems in children with ASD (249/589; 42%) compared with their unaffected siblings (20/163; 12%) (p < .001). The 2 most common Gl problems in children with ASD were constipation (116/589; 20%) and chronic diarrhea (111/589; 19%). Conditional logistic regression analysis showed that having Full Autism (adjusted odds ratio [AOR] = 14.28, 95% confidence interval [CI]: 6.22-32.77) or Almost Autism (AOR = 5.16, 95% CI 2.02-13.21) was most highly associated with experiencing GI problems. Increased autism symptom severity was associated with higher odds of GI problems (AOR for trend = 2.63, 95% CI: 1.56-4.45). CONCLUSIONS : Parents report significantly more GI problems in children with familial ASD, especially those with Full Autism, than in their unaffected children. Increased autism symptom severity is associated with increased odds of having GI problems.
Collapse
|
32
|
Lauwers GY, Fujita H, Nagata K, Shimizu M. Pathology of non-Helicobacter pylori gastritis: extending the histopathologic horizons. J Gastroenterol 2010; 45:131-45. [PMID: 19967418 DOI: 10.1007/s00535-009-0146-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2009] [Accepted: 09/29/2009] [Indexed: 02/04/2023]
Abstract
The development of modern endoscopic techniques, easier and greater access to healthcare, and interest in Helicobacter pylori infection and its implications have all led to a significant increase in upper endoscopies. In turn, gastroenterologists and pathologists have been recognizing an ever-increasing number of patterns of mucosal injury. Consequently, there is now an interest in a wider aspect of non-neoplastic gastric pathology, namely, non-HP (H. pylori) gastritis. In this review, we present major clinico-pathological entities, based on either the salient morphological features or the underlying etiologies.
Collapse
Affiliation(s)
- Gregory Y Lauwers
- Gastrointestinal Pathology Service, Department of Pathology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street WRN 2, Boston, MA 02114-2696, USA.
| | | | | | | |
Collapse
|
33
|
Buie T, Campbell DB, Fuchs GJ, Furuta GT, Levy J, Vandewater J, Whitaker AH, Atkins D, Bauman ML, Beaudet AL, Carr EG, Gershon MD, Hyman SL, Jirapinyo P, Jyonouchi H, Kooros K, Kushak R, Levitt P, Levy SE, Lewis JD, Murray KF, Natowicz MR, Sabra A, Wershil BK, Weston SC, Zeltzer L, Winter H. Evaluation, diagnosis, and treatment of gastrointestinal disorders in individuals with ASDs: a consensus report. Pediatrics 2010; 125 Suppl 1:S1-18. [PMID: 20048083 DOI: 10.1542/peds.2009-1878c] [Citation(s) in RCA: 470] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Autism spectrum disorders (ASDs) are common and clinically heterogeneous neurodevelopmental disorders. Gastrointestinal disorders and associated symptoms are commonly reported in individuals with ASDs, but key issues such as the prevalence and best treatment of these conditions are incompletely understood. A central difficulty in recognizing and characterizing gastrointestinal dysfunction with ASDs is the communication difficulties experienced by many affected individuals. A multidisciplinary panel reviewed the medical literature with the aim of generating evidence-based recommendations for diagnostic evaluation and management of gastrointestinal problems in this patient population. The panel concluded that evidence-based recommendations are not yet available. The consensus expert opinion of the panel was that individuals with ASDs deserve the same thoroughness and standard of care in the diagnostic workup and treatment of gastrointestinal concerns as should occur for patients without ASDs. Care providers should be aware that problem behavior in patients with ASDs may be the primary or sole symptom of the underlying medical condition, including some gastrointestinal disorders. For these patients, integration of behavioral and medical care may be most beneficial. Priorities for future research are identified to advance our understanding and management of gastrointestinal disorders in persons with ASDs.
Collapse
Affiliation(s)
- Timothy Buie
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Ibrahim SH, Voigt RG, Katusic SK, Weaver AL, Barbaresi WJ. Incidence of gastrointestinal symptoms in children with autism: a population-based study. Pediatrics 2009; 124:680-6. [PMID: 19651585 PMCID: PMC2747040 DOI: 10.1542/peds.2008-2933] [Citation(s) in RCA: 172] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE To determine whether children with autism have an increased incidence of gastrointestinal symptoms compared with matched control subjects in a population-based sample. DESIGN/METHODS In a previous study including all of the residents of Olmsted County, Minnesota, aged <21 years between 1976 and 1997, we identified 124 children who fulfilled criteria on the basis of Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition, for a research diagnosis of autism. Two matched control subjects were identified for each case subject. Through the Rochester Epidemiology Project, all medical diagnoses, are indexed for computerized retrieval. Gastrointestinal diagnoses before 21 years of age were grouped into 5 categories: (1) constipation; (2) diarrhea; (3) abdominal bloating, discomfort, or irritability; (4) gastroesophageal reflux or vomiting; and (5) feeding issues or food selectivity. The cumulative incidence of each category was calculated by using the Kaplan-Meier method. Cox proportional hazards models were fit to estimate the risk ratios (case subjects versus control subjects) and corresponding 95% confidence intervals. RESULTS Subjects were followed to median ages of 18.2 (case subjects) and 18.7 (control subjects) years. Significant differences between autism case and control subjects were identified in the cumulative incidence of constipation (33.9% vs 17.6%) and feeding issues/food selectivity (24.5% vs 16.1). No significant associations were found between autism case status and overall incidence of gastrointestinal symptoms or any other gastrointestinal symptom category. CONCLUSIONS As constipation and feeding issues/food selectivity often have a behavioral etiology, data suggest that a neurobehavioral rather than a primary organic gastrointestinal etiology may account for the higher incidence of these gastrointestinal symptoms in children with autism.
Collapse
Affiliation(s)
- Samar H. Ibrahim
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, United States
| | - Robert G. Voigt
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, United States
| | - Slavica K. Katusic
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, United States
| | - Amy L. Weaver
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, United States
| | - William J. Barbaresi
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
35
|
Autistic enterocolitis: fact or fiction? CANADIAN JOURNAL OF GASTROENTEROLOGY = JOURNAL CANADIEN DE GASTROENTEROLOGIE 2009; 23:95-8. [PMID: 19214283 DOI: 10.1155/2009/394317] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Autism spectrum disorder refers to syndromes of varying severity, typified by impaired social interactions, communicative delays and restricted, repetitive behaviours and interests. The prevalence of autism spectrum disorders has been on the rise, while the etiology remains unclear and most likely multifactorial. There have been several reports of a link between autism and chronic gastrointestinal symptoms. Endoscopy trials have demonstrated a higher prevalence of nonspecific colitis, lymphoid hyperplasia and focally enhanced gastritis compared with controls. Postulated mechanisms include aberrant immune responses to some dietary proteins, abnormal intestinal permeability and unfavourable gut microflora. Two autism spectrum disorder patients with chronic intestinal symptoms and abnormal endoscopic findings are described, followed by a review of this controversial topic.
Collapse
|
36
|
JOHNSON CHRISPLAUCHÉ, MYERS SCOTTM. Autism Spectrum Disorders. DEVELOPMENTAL-BEHAVIORAL PEDIATRICS 2008:519-577. [DOI: 10.1016/b978-0-323-04025-9.50018-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
37
|
Abstract
Pediatricians have an important role not only in early recognition and evaluation of autism spectrum disorders but also in chronic management of these disorders. The primary goals of treatment are to maximize the child's ultimate functional independence and quality of life by minimizing the core autism spectrum disorder features, facilitating development and learning, promoting socialization, reducing maladaptive behaviors, and educating and supporting families. To assist pediatricians in educating families and guiding them toward empirically supported interventions for their children, this report reviews the educational strategies and associated therapies that are the primary treatments for children with autism spectrum disorders. Optimization of health care is likely to have a positive effect on habilitative progress, functional outcome, and quality of life; therefore, important issues, such as management of associated medical problems, pharmacologic and nonpharmacologic intervention for challenging behaviors or coexisting mental health conditions, and use of complementary and alternative medical treatments, are also addressed.
Collapse
|
38
|
Macdonald TT, Domizio P. Autistic enterocolitis: is it a histopathological entity? Histopathology 2007; 51:552-3. [PMID: 17880534 DOI: 10.1111/j.1365-2559.2007.02805.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
39
|
Fagbemi AAO, Torrente F, Hilson AJW, Thomson MA, Heuschkel RB, Murch SH. Massive gastrointestinal haemorrhage in isolated intestinal Henoch-Schonlein purpura with response to intravenous immunoglobulin infusion. Eur J Pediatr 2007; 166:915-9. [PMID: 17120033 DOI: 10.1007/s00431-006-0337-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2006] [Accepted: 10/05/2006] [Indexed: 10/23/2022]
Abstract
There is increasing recognition that Henoch-Schonlein purpura may present in an atypical form in which gastrointestinal symptoms may predominate, and classic cutaneous changes may be delayed or absent. This may lead to significant diagnostic delay. We report the case of a 9-year-old girl who presented acutely with life-threatening gastrointestinal haemorrhage from multiple intestinal sites, with no skin rash and only mild evidence of renal involvement. Henoch-Schonlein purpura was confirmed by finding IgA deposition on vessels within gastric and duodenal mucosa, while immunohistochemistry also identified dense focal T cell infiltration in gastric mucosa and within duodenal epithelium. After initial stabilisation, the patient became shocked due to further gastrointestinal haemorrhage. Isotope bleeding scan identified multiple bleeding sites. Her endoscopically confirmed gastritis was sufficiently severe to preclude corticosteroids, and she was thus treated with intravenous immunoglobulin. This therapy induced prompt and sustained resolution of symptoms, and she has remained well since. Our patient's response concords with previous reports in corticosteroid-resistant cases to suggest that severe intestinal Henoch-Schonlein purpura may respond preferentially to intravenous immunoglobulin (IVIG) therapy. In severe cases where there is significant gastritis, IVIG provides an effective alternative to corticosteroids that may be employed as first-line therapy.
Collapse
Affiliation(s)
- Andrew A O Fagbemi
- Centre for Paediatric Gastroenterology, Royal Free and University College Medical School, London, UK
| | | | | | | | | | | |
Collapse
|
40
|
Schneider CK, Melmed RD, Barstow LE, Enriquez FJ, Ranger-Moore J, Ostrem JA. Oral human immunoglobulin for children with autism and gastrointestinal dysfunction: a prospective, open-label study. J Autism Dev Disord 2007; 36:1053-64. [PMID: 16845577 DOI: 10.1007/s10803-006-0141-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Immunoglobulin secretion onto mucosal surfaces is a major component of the mucosal immune system. We hypothesized that chronic gastrointestinal (GI) disturbances associated with autistic disorder (AD) may be due to an underlying deficiency in mucosal immunity, and that orally administered immunoglobulin would be effective in alleviating chronic GI dysfunction in these individuals. In this pilot study, twelve male subjects diagnosed with AD were evaluated using a GI severity index (GSI) while receiving daily dosing with encapsulated human immunoglobulin. Following eight weeks of treatment, 50% of the subjects met prespecified criteria for response in GI signs and symptoms and showed significant behavioral improvement as assessed by the Autism Behavior Checklist and parent and physician rated Clinical Global Impression of Improvement.
Collapse
|
41
|
|
42
|
Abstract
The discovery of Helicobacter pylori and its intimate role in the development of the most common form of chronic gastritis has elicited a much-needed interest in non-neoplastic gastric pathology. This has been paralleled by an increase in upper endoscopic examinations, which allow recognition of novel patterns and distribution of mucosal injury. Numerous attempts at classification have been made, most based on the acuteness or chronicity of gastric mucosal injury. In this review, we will not offer a new classification but present a detailed description of the major clinicopathological entities, based either on the salient morphological features or the underlying aetiologies, i.e. iatrogenic, autoimmune, vascular or idiopathic.
Collapse
Affiliation(s)
- A Srivastava
- Department of Pathology, Dartmouth Hitchcock Medical Center and Dartmouth Medical School, Lebanon, NH, USA
| | | |
Collapse
|
43
|
Abstract
H pylori is now accepted as the cause of gastritis and gastritis-associated diseases, such as duodenal ulcer, gastric ulcer, gastric carcinoma, and gastric MALT lymphoma. The natural history of H pylori gastritis includes inflammation progressing from the antrum into the adjacent corpus resulting in an atrophic front of advancing injury leading to a reduction in acid secretion and eventual loss of parietal cells and development of atrophy. Sub-typing intestinal metaplasia has no clinical value to the patient, the pathologist, or the endoscopist. The pattern, extent, and severity of atrophy, with or without intestinal metaplasia, is a far more important predictor than is intestinal metaplasia subtype. The challenge remains to identify a reliable marker that relates to pre-malignant potential.
Collapse
|
44
|
Ashwood P, Wakefield AJ. Immune activation of peripheral blood and mucosal CD3+ lymphocyte cytokine profiles in children with autism and gastrointestinal symptoms. J Neuroimmunol 2006; 173:126-34. [PMID: 16494951 DOI: 10.1016/j.jneuroim.2005.12.007] [Citation(s) in RCA: 144] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2005] [Revised: 12/19/2005] [Accepted: 12/19/2005] [Indexed: 12/13/2022]
Abstract
Gastrointestinal pathology, characterized by lymphoid nodular hyperplasia and entero-colitis, has been demonstrated in a cohort of children with autistic spectrum disorder (ASD). Systemic and intestinal mucosal immune dysregulation was assessed in ASD children with gastrointestinal (GI) symptoms (n = 18), and typically developing controls (n = 27), including non-inflamed controls (NIC) and inflamed GI control children with Crohn's disease (CD), by analysis of intracellular cytokines in CD3+ lymphocytes. In both peripheral blood and mucosa, CD3+ TNFalpha+ and CD3+ IFNgamma+ were increased in ASD children compared with NIC (p < 0.004) and reached levels similar to CD. In contrast, peripheral and mucosal CD3+ IL-10+ were markedly lower in ASD children with GI symptoms compared with both NIC and CD controls (p < 0.02). In addition, mucosal CD3+ IL-4+ cells were increased (p < 0.007) in ASD compared with NIC. There is a unique pattern of peripheral blood and mucosal CD3+ lymphocytes intracellular cytokines, which is consistent with significant immune dysregulation, in this ASD cohort.
Collapse
Affiliation(s)
- Paul Ashwood
- Department of Medical Microbiology and Immunology, University of California at Davis, M.I.N.D. Institute, Wet Lab building, 50th Street, Sacramento, CA 95817, USA.
| | | |
Collapse
|
45
|
Ricuarte O, Gutierrez O, Cardona H, Kim JG, Graham DY, El-Zimaity HMT. Atrophic gastritis in young children and adolescents. J Clin Pathol 2006; 58:1189-93. [PMID: 16254110 PMCID: PMC1770773 DOI: 10.1136/jcp.2005.026310] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Helicobacter pylori associated gastric cancer arises via a multistage process, with atrophic gastritis being the precursor lesion. Helicobacter pylori is typically acquired in childhood, yet little is known of the prevalence of atrophic gastritis in childhood. AIM To study atrophic gastritis among children from countries with high gastric cancer incidence. METHODS Sections from topographically mapped gastric biopsy specimens from children undergoing clinically indicated endoscopy in Korea and Colombia were evaluated using visual analogue scales. Atrophy was defined as loss of normal glandular components, including replacement with fibrosis, intestinal metaplasia (IM), and/or pseudopyloric metaplasia of the corpus (identified by the presence of pepsinogen I in mucosa that was topographically corpus but phenotypically antrum). RESULTS One hundred and seventy three children, 58 from Korea (median age, 14 years) and 115 from Colombia (median age, 13 years), were studied. Helicobacter pylori was present in 85% of Colombian children versus 17% of Korean children (p<0.01). Atrophic mucosa near the antrum-corpus border was present in 16% of children, primarily as pseudopyloric metaplasia (31%, IM; 63%, pseudopyloric metaplasia; 6%, both). The median age of children with corpus atrophy was 15 (range, 7-17) years. CONCLUSION Gastric atrophy occurs in H pylori infected children living in countries with high gastric cancer incidence. Identification and characterisation of the natural history of H pylori gastritis requires targeted biopsies to include the lesser and greater curve of the corpus, starting just proximal to the anatomical antrum-corpus junction, in addition to biopsies targeting the antrum and cardia.
Collapse
Affiliation(s)
- O Ricuarte
- Department of Gastroenterology, National University of Colombia, Bogotá, Colombia
| | | | | | | | | | | |
Collapse
|
46
|
|
47
|
Balzola F, Barbon V, Repici A, Rizzetto M, Clauser D, Gandione M, Sapino A. Panenteric IBD-like disease in a patient with regressive autism shown for the first time by the wireless capsule enteroscopy: another piece in the jigsaw of this gut-brain syndrome? Am J Gastroenterol 2005; 100:979-81. [PMID: 15784047 DOI: 10.1111/j.1572-0241.2005.41202_4.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
48
|
Gershon MD, Ratcliffe EM. Developmental biology of the enteric nervous system: pathogenesis of Hirschsprung's disease and other congenital dysmotilities. Semin Pediatr Surg 2004; 13:224-35. [PMID: 15660316 PMCID: PMC2835989 DOI: 10.1053/j.sempedsurg.2004.10.019] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Michael D Gershon
- Department of Anatomy and Cell Biology, Columbia University College of Physicians and Surgeons, 630 West 268th Street, New York, NY 10032, USA.
| | | |
Collapse
|
49
|
|