1
|
Kelly DL, Lee CM, Roche DJO, Talor MV, Clark S, Eaton WW. Randomized Double Blind Inpatient Study of a Gluten-Free Diet in Persons with Schizophrenia. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.24.25322813. [PMID: 40061327 PMCID: PMC11888518 DOI: 10.1101/2025.02.24.25322813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/18/2025]
Abstract
Background Schizophrenia and related disorders (SRD) are characterized by positive and negative symptoms, such as anhedonia and avolition. There are no current FDA approved treatments for negative symptoms, which is a critical gap in our treatment of people with SRDs, since they are a major determinant of functional impairment. An emerging literature suggests that SRDs have a relationship with immune function and inflammation. Recently an SRD subgroup with high inflammation and elevated levels of anti-gliadin antibodies (AGA) of immunoglobulin G type (IgG) has been characterized. Negative symptom improvement has been previously observed with gluten removal in this subgroup in two small clinical trials. Methods We conducted a 5-week confirmatory double-blind placebo-controlled trial of a gluten free diet (GFD) versus gluten-containing diet (GCD) for negative symptoms in people with SRD who have elevated AGA IgG (NCT03183609). Participants were between the ages of 18-64 years, had baseline negative symptoms and a diagnosis of schizophrenia or schizoaffective disorder. Those included were screened for an AGA IgG >20 U, no serologic evidence of celiac disease, and stable antipsychotic treatment and dose. All participants were inpatients, received a GFD and were randomized to 30 grams of gluten or rice flour daily delivered in protein shakes. The Clinical Assessment Interview for Negative Symptoms (CAINS) Motivation and Pleasure (MAP) scale was the primary outcome measure. We also examined the CAINS Expressivity (EXP) scale, the Scale for the Assessment of Negative Symptoms (SANS), the Brief Psychiatric Rating Scale (BPRS), the MATRICS Consensus Cognitive Battery (MCCB) and conducted regular side effect screening and laboratory measures for safety. Findings Between 2018 and 2024, we included 39 participants (N=21 GFD and N=18 GCD). There was a significant improvement over time in the CAINS MAP (treatment X time df=30.1, F=2.78, p=0.045) in the GFD compared to GCD, but no significant change in the CAINS EXP, the SANS, BPRS or MCCB. The diet was well tolerated; the most frequently occurring side effects were constipation (38.1% GFD, 33.3% GCD), sedation (33.3% GFD, 50% GCD), dry mouth (33.3% GFD, 33.3% GCD), headache (33.3% GFD, 27.8% GCD), and insomnia (33.3% GFD, 27.8% GCD). Interpretation This is the first large scale double-blind randomized clinical trial in SRD with AGA IgG+. This replication of smaller studies suggests that negative symptoms, particularly anhedonia and avolition may be improved. However, we did not replicate our previous finding of cognitive improvement and COVID-19 likely impacted the extent of improvement in negative symptoms due to quarantines and lockdowns. More work is needed to determine the mechanism of action of gluten removal in this subgroup with hopes of developing new treatment targets for motivational deficits of this illness. Funding This project was funded by NIMH R01 R01MH113617 (DL Kelly PI).
Collapse
Affiliation(s)
- Deanna L Kelly
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Christopher M Lee
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
- Sheppard E. Pratt Hospital, Towson, MD, USA
| | - Daniel J O Roche
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Monica V Talor
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sarah Clark
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - William W Eaton
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
2
|
Salem D, Clark SM, Roche DJO, Singh NJ, Talor MV, Buchanan RW, Harrington V, Ye Z, Chen S, Kelly DL. Pan T cells, Helper T cells, and Regulatory T cells are Associated with Negative Symptoms in Persons with Anti-Gliadin Antibody Positive Schizophrenia and Related Disorders. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.24.25322815. [PMID: 40061309 PMCID: PMC11888510 DOI: 10.1101/2025.02.24.25322815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/18/2025]
Abstract
Background About one in three persons with a schizophrenia related disorder (SRD) have elevated anti-gliadin IgG antibodies (AGA). This AGA positive (AGA+) subgroup of SRD clinically has a higher burden of negative symptoms and are symptoms associated with high functional impairments with a lack of effective therapeutics. Alterations in T cells have been demonstrated in SRD, and we have previously shown regulatory T cells (Tregs) are increased and correlate with fewer negative symptoms in persons with SRD compared with healthy controls. Methods To further elucidate the role of the immune system in AGA+ SRD pathology, we investigated the relationship of T cells and negative symptoms in 26 medicated and clinically stable persons with SRD. Participants had blood drawn; AGA-IgG measured by ELISA (AGA positive defined as ≥20 U); had flow cytometry performed to quantify proportions of pan T cells (CD3+), helper T cells (CD3+CD4+), Tregs (CD3+CD4+CD25+Foxp3+), and activated Tregs (aTregs) (CD3+CD4+CD25+Foxp3+RA-); had serum cytokines measured; and completed the Scale for the Assessment of Negative Symptoms (SANS) to measure negative symptoms. Results 46% of persons with SRD in this study were AGA+ and, in this group specifically, pan-T cells were correlated with worse SANS total, anhedonia, alogia, and avolition (p<0.05), while helper T cells and Tregs were correlated with less negative symptoms (respectively, SANS total and alogia; SANS total, anhedonia, alogia; P<0.05). AGA+ persons with SRD also had several elevated serum cytokines, corresponding with a broadly pro-inflammatory phenotype. Conclusions These hypothesis-generating findings highlight T cell dysfunction in AGA+ positive SRD, suggesting Tregs protecting against negative symptom severity but also an unidentified other T cell population to possibly be driving negative symptom severity.
Collapse
Affiliation(s)
- Deepak Salem
- Maryland Psychiatric Research Center, University of Maryland School of Medicine
- Department of Psychiatry, University of Maryland School of Medicine
| | - Sarah M Clark
- Department of Psychiatry, University of Maryland School of Medicine
| | - Daniel J O Roche
- Maryland Psychiatric Research Center, University of Maryland School of Medicine
- Department of Psychiatry, University of Maryland School of Medicine
| | - Nevil J Singh
- Department of Microbiology & Immunology, University of Maryland School of Medicine
| | - Monica V Talor
- Johns Hopkins University- School of Medicine, Department of Pathology, Baltimore MD
| | - Robert W Buchanan
- Maryland Psychiatric Research Center, University of Maryland School of Medicine
- Department of Psychiatry, University of Maryland School of Medicine
| | - Valerie Harrington
- Maryland Psychiatric Research Center, University of Maryland School of Medicine
- Department of Microbiology & Immunology, University of Maryland School of Medicine
| | - Zhenyao Ye
- Maryland Psychiatric Research Center, University of Maryland School of Medicine
| | - Shuo Chen
- Maryland Psychiatric Research Center, University of Maryland School of Medicine
- Department of Psychiatry, University of Maryland School of Medicine
| | - Deanna L Kelly
- Maryland Psychiatric Research Center, University of Maryland School of Medicine
- Department of Psychiatry, University of Maryland School of Medicine
| |
Collapse
|
3
|
Kim E, Redwood S, Liu F, Roche DJO, Chen S, Bentley WE, Eaton WW, Čiháková D, Talor MV, Kelly DL, Payne GF. Pilot study indicates that a gluten-free diet lowers oxidative stress for gluten-sensitive persons with schizophrenia. Schizophr Res 2024; 269:71-78. [PMID: 38749320 PMCID: PMC11215979 DOI: 10.1016/j.schres.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 04/19/2024] [Accepted: 05/02/2024] [Indexed: 05/22/2024]
Abstract
One-third of people with schizophrenia have elevated levels of anti-gliadin antibodies (AGA IgG). A 5-week randomized double-blind pilot study was performed in 2014-2017 in an inpatient setting to test the effect of a gluten-free diet (GFD) on participants with schizophrenia or schizoaffective disorder who also had elevated AGA IgG (≥ 20 U) but were negative for celiac disease. This earlier pilot study reported that the GFD-group showed improved gastrointestinal and psychiatric symptoms, and also improvements in TNF-α and the inflammatory cytokine IL-23. Here, we performed measurements of these banked plasma samples to detect levels of oxidative stress (OxSt) using a recently developed iridium (Ir)-reducing capacity assay. Triplicate measurements of these samples showed an Intraclass Correlation Coefficient of 0.84 which indicates good reproducibility. Further, a comparison of the OxSt measurements at the baseline and 5-week end-point for this small sample size shows that the GFD-group (N = 7) had lowered OxSt levels compared to the gluten-containing diet group (GCD; N = 9; p = 0.05). Finally, we showed that improvements in OxSt over these 5 weeks were correlated to improvements in gastrointestinal (r = +0.64, p = 0.0073) and psychiatric (r = +0.52, p = 0.039) symptoms. Also, we showed a possible association between the decrease in OxSt and the lowered levels of IL-23 (r = +0.44, p = 0.087), although without statistical significance. Thus, the Ir-reducing capacity assay provides a simple, objective measure of OxSt with the results providing further evidence that inflammation, redox dysregulation and OxSt may mediate interactions between the gut and brain.
Collapse
Affiliation(s)
- Eunkyoung Kim
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD 20742, United States; Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD 20742, United States
| | - Sidney Redwood
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, United States
| | - Fang Liu
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD 21228, United States
| | - Daniel J O Roche
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD 21228, United States
| | - Shuo Chen
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD 21228, United States
| | - William E Bentley
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD 20742, United States; Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD 20742, United States; Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, United States
| | - William W Eaton
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, United States
| | - Daniela Čiháková
- Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, United States; Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD 21205, United States
| | - Monica V Talor
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD 21205, United States
| | - Deanna L Kelly
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD 21228, United States.
| | - Gregory F Payne
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD 20742, United States; Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD 20742, United States.
| |
Collapse
|
4
|
Nguyen KD, Amerio A, Aguglia A, Magnani L, Parise A, Conio B, Serafini G, Amore M, Costanza A. Microglia and Other Cellular Mediators of Immunological Dysfunction in Schizophrenia: A Narrative Synthesis of Clinical Findings. Cells 2023; 12:2099. [PMID: 37626909 PMCID: PMC10453550 DOI: 10.3390/cells12162099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/13/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
Schizophrenia is a complex psychiatric condition that may involve immune system dysregulation. Since most putative disease mechanisms in schizophrenia have been derived from genetic association studies and fluid-based molecular analyses, this review aims to summarize the emerging evidence on clinical correlates to immune system dysfunction in this psychiatric disorder. We conclude this review by attempting to develop a unifying hypothesis regarding the relative contributions of microglia and various immune cell populations to the development of schizophrenia. This may provide important translational insights that can become useful for addressing the multifaceted clinical presentation of schizophrenia.
Collapse
Affiliation(s)
- Khoa D. Nguyen
- Department of Microbiology and Immunology, Stanford University, Palo Alto, CA 94305, USA;
- Tranquis Therapeutics, Palo Alto, CA 94065, USA
| | - Andrea Amerio
- Section of Psychiatry, Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16126 Genoa, Italy; (A.A.); (A.A.); (B.C.); (G.S.); (M.A.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Andrea Aguglia
- Section of Psychiatry, Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16126 Genoa, Italy; (A.A.); (A.A.); (B.C.); (G.S.); (M.A.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Luca Magnani
- Department of Psychiatry, San Maurizio Hospital of Bolzano, 39100 Bolzano, Italy;
| | - Alberto Parise
- Geriatric-Rehabilitation Department, University Hospital of Parma, 43126 Parma, Italy;
| | - Benedetta Conio
- Section of Psychiatry, Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16126 Genoa, Italy; (A.A.); (A.A.); (B.C.); (G.S.); (M.A.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Gianluca Serafini
- Section of Psychiatry, Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16126 Genoa, Italy; (A.A.); (A.A.); (B.C.); (G.S.); (M.A.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Mario Amore
- Section of Psychiatry, Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16126 Genoa, Italy; (A.A.); (A.A.); (B.C.); (G.S.); (M.A.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Alessandra Costanza
- Department of Psychiatry, Adult Psychiatry Service, University Hospitals of Geneva (HUG), 1207 Geneva, Switzerland
- Department of Psychiatry, Faculty of Biomedical Sciences, University of Italian Switzerland (USI), 6900 Lugano, Switzerland
- Department of Psychiatry, Faculty of Medicine, University of Geneva (UNIGE), 1211 Geneva, Switzerland
| |
Collapse
|
5
|
Kelly DL, Buchanan RW. Can the current schizophrenia construct endure? Schizophr Res 2022; 242:64-66. [PMID: 35067456 DOI: 10.1016/j.schres.2021.12.028] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/29/2022]
Affiliation(s)
- Deanna L Kelly
- University of Maryland, School of Medicine, Maryland Psychiatric Research Center, Box 21247, Baltimore, MD 21228, United States.
| | - Robert W Buchanan
- University of Maryland, School of Medicine, Maryland Psychiatric Research Center, Box 21247, Baltimore, MD 21228, United States.
| |
Collapse
|
6
|
Onaolapo OJ, Onaolapo AY. Nutrition, nutritional deficiencies, and schizophrenia: An association worthy of constant reassessment. World J Clin Cases 2021; 9:8295-8311. [PMID: 34754840 PMCID: PMC8554424 DOI: 10.12998/wjcc.v9.i28.8295] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 05/04/2021] [Accepted: 08/10/2021] [Indexed: 02/06/2023] Open
Abstract
Schizophrenia is a mental health disorder that occurs worldwide, cutting across cultures, socioeconomic groups, and geographical barriers. Understanding the details of the neurochemical basis of schizophrenia, factors that contribute to it and possible measures for intervention are areas of ongoing research. However, what has become more evident is the fact that in targeting the neurochemical imbalances that may underlie schizophrenia, the type of response seen with currently available phamacotherapeutic agents does not provide all the answers that are needed. Therefore, the possible contribution of non-pharmacological approaches to schizophrenia management is worthy of consideration. In recent times, research is beginning to show nutrition may play a possibly significant role in schizophrenia, affecting its development, progression and management; however, while attempts had been made to examine this possible relationship from different angles, articles addressing it from a holistic point of view are not common. In this review, we examine existing scientific literature dealing with the possible relationship between nutrition and schizophrenia, with a view to elucidating the impact of diet, nutritional deficiencies and excesses on the aetiology, progression, management and outcome of schizophrenia. Secondly, the effect of nutritional supplements in prevention, as sole therapy, or adjuncts in schizophrenia management are examined.
Collapse
Affiliation(s)
- Olakunle James Onaolapo
- Behavioural Neuroscience/Neuropharmacology Unit, Department of Pharmacology, Ladoke Akintola University of Technology, Osun State 234, Nigeria
| | | |
Collapse
|
7
|
Gluten and FODMAPs Relationship with Mental Disorders: Systematic Review. Nutrients 2021; 13:nu13061894. [PMID: 34072914 PMCID: PMC8228761 DOI: 10.3390/nu13061894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 05/28/2021] [Accepted: 05/28/2021] [Indexed: 11/16/2022] Open
Abstract
Nowadays, gluten and FODMAP food components (fermentable oligosaccharides, disaccharides, monosaccharides and polyols) are increasingly studied due to their possible relation with extraintestinal-associated conditions. In recent years, gluten-free diets (GFD) and low-FODMAP diets (LFD) are becoming more popular not only in order to avoid the food components that cause intolerances or allergies in some people, but also due to the direct influence of marketing movements or diet trends on feeding habits. Likewise, neurological and psychiatric diseases are currently of increasing importance in developed countries. For this reason, a bibliographic systematic review has been carried out to analyse whether there is a pathophysiological relationship between the dietary intake of gluten or FODMAPs with mental disorders. This review collects 13 clinical and randomized controlled trials, based on the PRISMA statement, which have been published in the last ten years. Based on these results, limiting or ruling out gluten or FODMAPs in the diet might be beneficial for symptoms such as depression, anxiety (7 out of 7 articles found any positive effect), or cognition deficiency (improvements in several cognition test measurements in one trial), and to a lesser extent for schizophrenia and the autism spectrum. Nevertheless, further studies are needed to obtain completely reliable conclusions.
Collapse
|
8
|
The Differences between Gluten Sensitivity, Intestinal Biomarkers and Immune Biomarkers in Patients with First-Episode and Chronic Schizophrenia. J Clin Med 2020; 9:jcm9113707. [PMID: 33218214 PMCID: PMC7699286 DOI: 10.3390/jcm9113707] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/13/2020] [Accepted: 11/16/2020] [Indexed: 12/15/2022] Open
Abstract
Schizophrenia is a heterogeneous disorder without a fully elucidated etiology and mechanisms. One likely explanation for the development of schizophrenia is low-grade inflammation, possibly caused by processes in the gastrointestinal tract related to gluten sensitivity. The aims of this study were to: (1) compare levels of markers of gluten sensitivity, inflammation and gut permeability, and (2) determine associations between gluten sensitivity, inflammation, and intestinal permeability in patients with first-episode/chronic (FS/CS) schizophrenia and healthy individuals (HC). The total sample comprised 162 individuals (52 FS; 50 CS, and 60 HC). The examination included clinical variables, nutritional assessment, and serum concentrations of: high-sensitivity C-reactive protein (hs-CRP), interleukin-6 (IL-6), soluble CD14 (sCD14), anti-Saccharomyces cerevisiae antibody (ASCA), antigliadin antibodies (AGA) IgA/IgG, antibodies against tissue transglutaminase 2 (anti-tTG) IgA, anti-deamidated gliadin peptides (anti-DGP) IgG. A significant difference between groups was found in sCD14, ASCA, hs-CRP, IL-6 and AGA IgA levels. AGA IgG/IgA levels were higher in the FS (11.54%; 30.77%) and CS (26%; 20%) groups compared to HC. The association between intestinal permeability and inflammation in the schizophrenic patients only was noted. The risk for developing schizophrenia was odds ratio (OR) = 4.35 (95% confidence interval (CI 1.23-15.39) for AGA IgA and 3.08 (95% CI 1.19-7.99) for positive AGA IgG. Inflammation and food hypersensitivity reactions initiated by increased intestinal permeability may contribute to the pathophysiology of schizophrenia. The immune response to gluten in FS differs from that found in CS.
Collapse
|
9
|
The Effects of a Gluten-Free Diet on Immune Markers and Kynurenic Acid Pathway Metabolites in Patients With Schizophrenia Positive for Antigliadin Antibodies Immunoglobulin G. J Clin Psychopharmacol 2020; 40:317-319. [PMID: 32332475 PMCID: PMC7185031 DOI: 10.1097/jcp.0000000000001197] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
10
|
Kelly DL, Demyanovich HK, Rodriguez KM, Ciháková D, Talor MV, McMahon RP, Richardson CM, Vyas G, Adams HA, August SM, Fasano A, Cascella NG, Feldman SM, Liu F, Sayer MA, Powell MM, Wehring HJ, Buchanan RW, Gold JM, Carpenter WT, Eaton WW. Randomized controlled trial of a gluten-free diet in patients with schizophrenia positive for antigliadin antibodies (AGA IgG): a pilot feasibility study. J Psychiatry Neurosci 2019; 44. [PMID: 30938127 PMCID: PMC6606425 DOI: 10.1503/jpn.180174] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Approximately one-third of people with schizophrenia have elevated levels of anti-gliadin antibodies of the immunoglobulin G type (AGA IgG) — a higher rate than seen in healthy controls. We performed the first double-blind clinical trial of gluten-free versus gluten-containing diets in a subset of patients with schizophrenia who were positive for AGA IgG. METHODS In this pilot feasibility study, 16 participants with schizophrenia or schizoaffective disorder who had elevated AGA IgG (≥ 20 U) but were negative for celiac disease were admitted to an inpatient unit for a 5-week trial. All participants received standardized gluten-free meals and were randomized in a double-blind fashion to receive a shake containing 10 g of gluten flour or 10 g of rice flour each day. Participants were rated for psychiatric, cognitive and gastrointestinal symptoms at baseline and endpoint. RESULTS Of the 16 participants, 14 completed the 5-week trial (2 discontinued early for administrative reasons). Compared with participants on the gluten-containing diet, participants on the gluten-free diet showed improvement on the Clinical Global Impressions scale (Cohen d = –0.75) and in negative symptoms (Cohen d = –0.53). We noted no improvement in positive or global cognitive symptoms, but did observe an improvement in attention favouring the gluten-free diet (Cohen d = 0.60). Robust improvements in gastrointestinal adverse effects occurred in the gluten-free group relative to the glutencontaining group. Adverse effects were similar between groups. LIMITATIONS This study was limited by its small sample size; larger studies are needed. CONCLUSION This feasibility study suggests that removal of gluten from the diet is associated with improvement in psychiatric and gastrointestinal symptoms in people with schizophrenia or schizoaffective disorder.
Collapse
Affiliation(s)
- Deanna L. Kelly
- From the Maryland Psychiatric Research Center (MPRC), School of Medicine, University of Maryland, College Park, MD (Kelly, McMahon, August, Feldman, Liu, Powell, Wehring, Buchanan, Gold, Carpenter); the Department of Orthopedics, School of Medicine, University of Maryland, College Park, MD (Demyanovich); the Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (Rodriguez, Eaton); the Department of Pathology, Division of Immunology, Immune Disorders Laboratory, Johns Hopkins University, Baltimore, MD (Cˇiháková, Talor); the Spring Grove Hospital Center, Baltimore, MD (Richardson, Vyas, Adams); the Center for Celiac Research and Treatment, Massachusetts General Hospital, Boston, MA (Fasano); the Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD (Cascella); the Department of Psychology, Kent State University, Kent, OH (Sayer)
| | - Haley K. Demyanovich
- From the Maryland Psychiatric Research Center (MPRC), School of Medicine, University of Maryland, College Park, MD (Kelly, McMahon, August, Feldman, Liu, Powell, Wehring, Buchanan, Gold, Carpenter); the Department of Orthopedics, School of Medicine, University of Maryland, College Park, MD (Demyanovich); the Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (Rodriguez, Eaton); the Department of Pathology, Division of Immunology, Immune Disorders Laboratory, Johns Hopkins University, Baltimore, MD (Cˇiháková, Talor); the Spring Grove Hospital Center, Baltimore, MD (Richardson, Vyas, Adams); the Center for Celiac Research and Treatment, Massachusetts General Hospital, Boston, MA (Fasano); the Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD (Cascella); the Department of Psychology, Kent State University, Kent, OH (Sayer)
| | - Katrina M. Rodriguez
- From the Maryland Psychiatric Research Center (MPRC), School of Medicine, University of Maryland, College Park, MD (Kelly, McMahon, August, Feldman, Liu, Powell, Wehring, Buchanan, Gold, Carpenter); the Department of Orthopedics, School of Medicine, University of Maryland, College Park, MD (Demyanovich); the Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (Rodriguez, Eaton); the Department of Pathology, Division of Immunology, Immune Disorders Laboratory, Johns Hopkins University, Baltimore, MD (Cˇiháková, Talor); the Spring Grove Hospital Center, Baltimore, MD (Richardson, Vyas, Adams); the Center for Celiac Research and Treatment, Massachusetts General Hospital, Boston, MA (Fasano); the Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD (Cascella); the Department of Psychology, Kent State University, Kent, OH (Sayer)
| | - Daniela Ciháková
- From the Maryland Psychiatric Research Center (MPRC), School of Medicine, University of Maryland, College Park, MD (Kelly, McMahon, August, Feldman, Liu, Powell, Wehring, Buchanan, Gold, Carpenter); the Department of Orthopedics, School of Medicine, University of Maryland, College Park, MD (Demyanovich); the Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (Rodriguez, Eaton); the Department of Pathology, Division of Immunology, Immune Disorders Laboratory, Johns Hopkins University, Baltimore, MD (Cˇiháková, Talor); the Spring Grove Hospital Center, Baltimore, MD (Richardson, Vyas, Adams); the Center for Celiac Research and Treatment, Massachusetts General Hospital, Boston, MA (Fasano); the Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD (Cascella); the Department of Psychology, Kent State University, Kent, OH (Sayer)
| | - Monica V. Talor
- From the Maryland Psychiatric Research Center (MPRC), School of Medicine, University of Maryland, College Park, MD (Kelly, McMahon, August, Feldman, Liu, Powell, Wehring, Buchanan, Gold, Carpenter); the Department of Orthopedics, School of Medicine, University of Maryland, College Park, MD (Demyanovich); the Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (Rodriguez, Eaton); the Department of Pathology, Division of Immunology, Immune Disorders Laboratory, Johns Hopkins University, Baltimore, MD (Cˇiháková, Talor); the Spring Grove Hospital Center, Baltimore, MD (Richardson, Vyas, Adams); the Center for Celiac Research and Treatment, Massachusetts General Hospital, Boston, MA (Fasano); the Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD (Cascella); the Department of Psychology, Kent State University, Kent, OH (Sayer)
| | - Robert P. McMahon
- From the Maryland Psychiatric Research Center (MPRC), School of Medicine, University of Maryland, College Park, MD (Kelly, McMahon, August, Feldman, Liu, Powell, Wehring, Buchanan, Gold, Carpenter); the Department of Orthopedics, School of Medicine, University of Maryland, College Park, MD (Demyanovich); the Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (Rodriguez, Eaton); the Department of Pathology, Division of Immunology, Immune Disorders Laboratory, Johns Hopkins University, Baltimore, MD (Cˇiháková, Talor); the Spring Grove Hospital Center, Baltimore, MD (Richardson, Vyas, Adams); the Center for Celiac Research and Treatment, Massachusetts General Hospital, Boston, MA (Fasano); the Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD (Cascella); the Department of Psychology, Kent State University, Kent, OH (Sayer)
| | - Charles M. Richardson
- From the Maryland Psychiatric Research Center (MPRC), School of Medicine, University of Maryland, College Park, MD (Kelly, McMahon, August, Feldman, Liu, Powell, Wehring, Buchanan, Gold, Carpenter); the Department of Orthopedics, School of Medicine, University of Maryland, College Park, MD (Demyanovich); the Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (Rodriguez, Eaton); the Department of Pathology, Division of Immunology, Immune Disorders Laboratory, Johns Hopkins University, Baltimore, MD (Cˇiháková, Talor); the Spring Grove Hospital Center, Baltimore, MD (Richardson, Vyas, Adams); the Center for Celiac Research and Treatment, Massachusetts General Hospital, Boston, MA (Fasano); the Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD (Cascella); the Department of Psychology, Kent State University, Kent, OH (Sayer)
| | - Gopal Vyas
- From the Maryland Psychiatric Research Center (MPRC), School of Medicine, University of Maryland, College Park, MD (Kelly, McMahon, August, Feldman, Liu, Powell, Wehring, Buchanan, Gold, Carpenter); the Department of Orthopedics, School of Medicine, University of Maryland, College Park, MD (Demyanovich); the Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (Rodriguez, Eaton); the Department of Pathology, Division of Immunology, Immune Disorders Laboratory, Johns Hopkins University, Baltimore, MD (Cˇiháková, Talor); the Spring Grove Hospital Center, Baltimore, MD (Richardson, Vyas, Adams); the Center for Celiac Research and Treatment, Massachusetts General Hospital, Boston, MA (Fasano); the Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD (Cascella); the Department of Psychology, Kent State University, Kent, OH (Sayer)
| | - Heather A. Adams
- From the Maryland Psychiatric Research Center (MPRC), School of Medicine, University of Maryland, College Park, MD (Kelly, McMahon, August, Feldman, Liu, Powell, Wehring, Buchanan, Gold, Carpenter); the Department of Orthopedics, School of Medicine, University of Maryland, College Park, MD (Demyanovich); the Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (Rodriguez, Eaton); the Department of Pathology, Division of Immunology, Immune Disorders Laboratory, Johns Hopkins University, Baltimore, MD (Cˇiháková, Talor); the Spring Grove Hospital Center, Baltimore, MD (Richardson, Vyas, Adams); the Center for Celiac Research and Treatment, Massachusetts General Hospital, Boston, MA (Fasano); the Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD (Cascella); the Department of Psychology, Kent State University, Kent, OH (Sayer)
| | - Sharon M. August
- From the Maryland Psychiatric Research Center (MPRC), School of Medicine, University of Maryland, College Park, MD (Kelly, McMahon, August, Feldman, Liu, Powell, Wehring, Buchanan, Gold, Carpenter); the Department of Orthopedics, School of Medicine, University of Maryland, College Park, MD (Demyanovich); the Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (Rodriguez, Eaton); the Department of Pathology, Division of Immunology, Immune Disorders Laboratory, Johns Hopkins University, Baltimore, MD (Cˇiháková, Talor); the Spring Grove Hospital Center, Baltimore, MD (Richardson, Vyas, Adams); the Center for Celiac Research and Treatment, Massachusetts General Hospital, Boston, MA (Fasano); the Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD (Cascella); the Department of Psychology, Kent State University, Kent, OH (Sayer)
| | - Alessio Fasano
- From the Maryland Psychiatric Research Center (MPRC), School of Medicine, University of Maryland, College Park, MD (Kelly, McMahon, August, Feldman, Liu, Powell, Wehring, Buchanan, Gold, Carpenter); the Department of Orthopedics, School of Medicine, University of Maryland, College Park, MD (Demyanovich); the Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (Rodriguez, Eaton); the Department of Pathology, Division of Immunology, Immune Disorders Laboratory, Johns Hopkins University, Baltimore, MD (Cˇiháková, Talor); the Spring Grove Hospital Center, Baltimore, MD (Richardson, Vyas, Adams); the Center for Celiac Research and Treatment, Massachusetts General Hospital, Boston, MA (Fasano); the Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD (Cascella); the Department of Psychology, Kent State University, Kent, OH (Sayer)
| | - Nicola G. Cascella
- From the Maryland Psychiatric Research Center (MPRC), School of Medicine, University of Maryland, College Park, MD (Kelly, McMahon, August, Feldman, Liu, Powell, Wehring, Buchanan, Gold, Carpenter); the Department of Orthopedics, School of Medicine, University of Maryland, College Park, MD (Demyanovich); the Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (Rodriguez, Eaton); the Department of Pathology, Division of Immunology, Immune Disorders Laboratory, Johns Hopkins University, Baltimore, MD (Cˇiháková, Talor); the Spring Grove Hospital Center, Baltimore, MD (Richardson, Vyas, Adams); the Center for Celiac Research and Treatment, Massachusetts General Hospital, Boston, MA (Fasano); the Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD (Cascella); the Department of Psychology, Kent State University, Kent, OH (Sayer)
| | - Stephanie M. Feldman
- From the Maryland Psychiatric Research Center (MPRC), School of Medicine, University of Maryland, College Park, MD (Kelly, McMahon, August, Feldman, Liu, Powell, Wehring, Buchanan, Gold, Carpenter); the Department of Orthopedics, School of Medicine, University of Maryland, College Park, MD (Demyanovich); the Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (Rodriguez, Eaton); the Department of Pathology, Division of Immunology, Immune Disorders Laboratory, Johns Hopkins University, Baltimore, MD (Cˇiháková, Talor); the Spring Grove Hospital Center, Baltimore, MD (Richardson, Vyas, Adams); the Center for Celiac Research and Treatment, Massachusetts General Hospital, Boston, MA (Fasano); the Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD (Cascella); the Department of Psychology, Kent State University, Kent, OH (Sayer)
| | - Fang Liu
- From the Maryland Psychiatric Research Center (MPRC), School of Medicine, University of Maryland, College Park, MD (Kelly, McMahon, August, Feldman, Liu, Powell, Wehring, Buchanan, Gold, Carpenter); the Department of Orthopedics, School of Medicine, University of Maryland, College Park, MD (Demyanovich); the Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (Rodriguez, Eaton); the Department of Pathology, Division of Immunology, Immune Disorders Laboratory, Johns Hopkins University, Baltimore, MD (Cˇiháková, Talor); the Spring Grove Hospital Center, Baltimore, MD (Richardson, Vyas, Adams); the Center for Celiac Research and Treatment, Massachusetts General Hospital, Boston, MA (Fasano); the Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD (Cascella); the Department of Psychology, Kent State University, Kent, OH (Sayer)
| | - MacKenzie A. Sayer
- From the Maryland Psychiatric Research Center (MPRC), School of Medicine, University of Maryland, College Park, MD (Kelly, McMahon, August, Feldman, Liu, Powell, Wehring, Buchanan, Gold, Carpenter); the Department of Orthopedics, School of Medicine, University of Maryland, College Park, MD (Demyanovich); the Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (Rodriguez, Eaton); the Department of Pathology, Division of Immunology, Immune Disorders Laboratory, Johns Hopkins University, Baltimore, MD (Cˇiháková, Talor); the Spring Grove Hospital Center, Baltimore, MD (Richardson, Vyas, Adams); the Center for Celiac Research and Treatment, Massachusetts General Hospital, Boston, MA (Fasano); the Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD (Cascella); the Department of Psychology, Kent State University, Kent, OH (Sayer)
| | - Megan M. Powell
- From the Maryland Psychiatric Research Center (MPRC), School of Medicine, University of Maryland, College Park, MD (Kelly, McMahon, August, Feldman, Liu, Powell, Wehring, Buchanan, Gold, Carpenter); the Department of Orthopedics, School of Medicine, University of Maryland, College Park, MD (Demyanovich); the Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (Rodriguez, Eaton); the Department of Pathology, Division of Immunology, Immune Disorders Laboratory, Johns Hopkins University, Baltimore, MD (Cˇiháková, Talor); the Spring Grove Hospital Center, Baltimore, MD (Richardson, Vyas, Adams); the Center for Celiac Research and Treatment, Massachusetts General Hospital, Boston, MA (Fasano); the Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD (Cascella); the Department of Psychology, Kent State University, Kent, OH (Sayer)
| | - Heidi J. Wehring
- From the Maryland Psychiatric Research Center (MPRC), School of Medicine, University of Maryland, College Park, MD (Kelly, McMahon, August, Feldman, Liu, Powell, Wehring, Buchanan, Gold, Carpenter); the Department of Orthopedics, School of Medicine, University of Maryland, College Park, MD (Demyanovich); the Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (Rodriguez, Eaton); the Department of Pathology, Division of Immunology, Immune Disorders Laboratory, Johns Hopkins University, Baltimore, MD (Cˇiháková, Talor); the Spring Grove Hospital Center, Baltimore, MD (Richardson, Vyas, Adams); the Center for Celiac Research and Treatment, Massachusetts General Hospital, Boston, MA (Fasano); the Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD (Cascella); the Department of Psychology, Kent State University, Kent, OH (Sayer)
| | - Robert W. Buchanan
- From the Maryland Psychiatric Research Center (MPRC), School of Medicine, University of Maryland, College Park, MD (Kelly, McMahon, August, Feldman, Liu, Powell, Wehring, Buchanan, Gold, Carpenter); the Department of Orthopedics, School of Medicine, University of Maryland, College Park, MD (Demyanovich); the Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (Rodriguez, Eaton); the Department of Pathology, Division of Immunology, Immune Disorders Laboratory, Johns Hopkins University, Baltimore, MD (Cˇiháková, Talor); the Spring Grove Hospital Center, Baltimore, MD (Richardson, Vyas, Adams); the Center for Celiac Research and Treatment, Massachusetts General Hospital, Boston, MA (Fasano); the Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD (Cascella); the Department of Psychology, Kent State University, Kent, OH (Sayer)
| | - James M. Gold
- From the Maryland Psychiatric Research Center (MPRC), School of Medicine, University of Maryland, College Park, MD (Kelly, McMahon, August, Feldman, Liu, Powell, Wehring, Buchanan, Gold, Carpenter); the Department of Orthopedics, School of Medicine, University of Maryland, College Park, MD (Demyanovich); the Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (Rodriguez, Eaton); the Department of Pathology, Division of Immunology, Immune Disorders Laboratory, Johns Hopkins University, Baltimore, MD (Cˇiháková, Talor); the Spring Grove Hospital Center, Baltimore, MD (Richardson, Vyas, Adams); the Center for Celiac Research and Treatment, Massachusetts General Hospital, Boston, MA (Fasano); the Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD (Cascella); the Department of Psychology, Kent State University, Kent, OH (Sayer)
| | - William T. Carpenter
- From the Maryland Psychiatric Research Center (MPRC), School of Medicine, University of Maryland, College Park, MD (Kelly, McMahon, August, Feldman, Liu, Powell, Wehring, Buchanan, Gold, Carpenter); the Department of Orthopedics, School of Medicine, University of Maryland, College Park, MD (Demyanovich); the Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (Rodriguez, Eaton); the Department of Pathology, Division of Immunology, Immune Disorders Laboratory, Johns Hopkins University, Baltimore, MD (Cˇiháková, Talor); the Spring Grove Hospital Center, Baltimore, MD (Richardson, Vyas, Adams); the Center for Celiac Research and Treatment, Massachusetts General Hospital, Boston, MA (Fasano); the Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD (Cascella); the Department of Psychology, Kent State University, Kent, OH (Sayer)
| | - William W. Eaton
- From the Maryland Psychiatric Research Center (MPRC), School of Medicine, University of Maryland, College Park, MD (Kelly, McMahon, August, Feldman, Liu, Powell, Wehring, Buchanan, Gold, Carpenter); the Department of Orthopedics, School of Medicine, University of Maryland, College Park, MD (Demyanovich); the Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (Rodriguez, Eaton); the Department of Pathology, Division of Immunology, Immune Disorders Laboratory, Johns Hopkins University, Baltimore, MD (Cˇiháková, Talor); the Spring Grove Hospital Center, Baltimore, MD (Richardson, Vyas, Adams); the Center for Celiac Research and Treatment, Massachusetts General Hospital, Boston, MA (Fasano); the Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD (Cascella); the Department of Psychology, Kent State University, Kent, OH (Sayer)
| |
Collapse
|
11
|
Yang H, Jiang Y, Chen Z, Wu J, Qiu C, Meng Q. A study of anti-gliadin antibodies in first-episode patients with schizophrenia among a Chinese population. Psychiatry Res 2019; 272:454-457. [PMID: 30611964 DOI: 10.1016/j.psychres.2018.12.161] [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: 05/21/2018] [Revised: 11/29/2018] [Accepted: 12/29/2018] [Indexed: 01/15/2023]
Abstract
A recent study suggested that digestion-resistant peptides derived from wheat gluten (mainly gliadin) could induce the secretion of anti-gliadin IgG antibodies in patients with schizophrenia. This research was then designed to replicate this initial finding in 134 drug-naïve patients with first-episode schizophrenia and 160 healthy controls. An enzyme-linked immunosorbent assay was developed in-house with 8 gliadin-derived peptide antigens to test anti-gliadin IgG antibodies in the circulation. The results showed that schizophrenia patients had significantly higher levels of plasma anti-AL2G2 IgG and anti-ABO3a IgG than healthy controls. Based on the specificity of 95%, anti-AL2G2 IgG assay had a sensitivity of 12.7% and anti-ABO3a IgG assay had a sensitivity of 17.2% for anti-ABO3a IgG assay. Increased levels of anti-AL2G2 and anti-ABC3a IgG antibodies were not correlated with total IgG levels in either the patient group or the control group. In conclusion, circulating IgG against AL2G2 and ABO3a may be useful biomarkers for identification of a gluten-sensitive subgroup of schizophrenia in the Chinese population although the present results are rather different from the work performed in a British population.
Collapse
Affiliation(s)
- Hua Yang
- Laboratory for Nursing Science & Institute of Laboratory Medicine, Guangdong Medical University, No.1 Xincheng Road, Dongguan 523808, China
| | - Yaling Jiang
- The Third People's Hospital of Jiangmen, Jiangmen 52900, China
| | - Zhenjian Chen
- The Third People's Hospital of Jiangmen, Jiangmen 52900, China
| | - Jingqing Wu
- The Third People's Hospital of Jiangmen, Jiangmen 52900, China
| | - Chaosen Qiu
- The Third People's Hospital of Jiangmen, Jiangmen 52900, China
| | - Qingyong Meng
- Laboratory for Nursing Science & Institute of Laboratory Medicine, Guangdong Medical University, No.1 Xincheng Road, Dongguan 523808, China.
| |
Collapse
|
12
|
Levinta A, Mukovozov I, Tsoutsoulas C. Use of a Gluten-Free Diet in Schizophrenia: A Systematic Review. Adv Nutr 2018; 9:824-832. [PMID: 30325398 PMCID: PMC6247287 DOI: 10.1093/advances/nmy056] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
We performed a systematic review of the literature to determine whether adherence to a gluten-free diet (GFD) leads to improved outcomes for patients with schizophrenia. We searched the AMED (Allied and Complementary Medicine; 1985-June 2016), MEDLINE (1946-June 2016), and Embase (1980-2016 week 24) databases using the terms "wheat" or "glutenin" or "gliadin" or "gluten" AND "schizophrenia." A total of 9 studies met the inclusion criteria for this review: 1 randomized controlled trial, 7 crossover studies, and 1 open-label pilot study. Six of the included studies demonstrated beneficial effects including improved functioning and decreased symptom severity after the course of a GFD, whereas 3 studies found no benefits. All of the included studies found that a GFD is well tolerated and can be adhered to by patients with schizophrenia. The findings of this systematic review should be interpreted with caution due to limitations inherent to nonrandomized trials, as well as the heterogeneity in the study design and the length of the GFD applied in each study. Publication bias is another potential limitation. Further research is required to examine the biomarkers of gluten sensitivity and inflammation to effectively target those patients with schizophrenia who will benefit most from this dietary intervention.
Collapse
Affiliation(s)
- Anastasia Levinta
- Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ilya Mukovozov
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
13
|
Analysis of gut microbiota diversity and auxiliary diagnosis as a biomarker in patients with schizophrenia: A cross-sectional study. Schizophr Res 2018; 197:470-477. [PMID: 29352709 DOI: 10.1016/j.schres.2018.01.002] [Citation(s) in RCA: 213] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 12/26/2017] [Accepted: 01/01/2018] [Indexed: 12/20/2022]
Abstract
With the advent of sequencing technology, characterization of schizophrenia with underlying probing of gut microbiome can provide abundant clues for diagnosis and prognosis of schizophrenia. In this study, we first compared the difference of gut microbiota between schizophrenia patients and healthy controls by 16S rRNA sequencing. We further explored whether gut microbiota can be used as a biomarker to assist in the diagnosis of schizophrenia. We restricted inclusion criteria strictly to control confounding bias. Finally, we investigated differences in fecal microbiota between 64 schizophrenia patients and 53 healthy controls. At the phylum level, we found that the abundance of Proteobacteria in the schizophrenia patients was significantly increased. At the genus level, the relative abundance of Succinivibrio, Megasphaera, Collinsella, Clostridium, Klebsiella and Methanobrevibacter was significantly higher whereas the abundance of Blautia, Coprococcus, Roseburia was decreased compared to health controls. The receiver operating characteristic curve analysis demonstrated that 12 significant microbiota biomarkers were capable of being used as diagnostic factors for distinguishing the schizophrenia cohort from those in the control cohort (AUC = 0.837). We performed PICRUSt analysis and found that several metabolic pathways differed significantly between healthy controls and schizophrenia patients, including vitamin B6 and fatty acid. In conclusion, there are some difference of gut microbiota between schizophrenia patients and healthy controls and the insights from this study could be used to develop microbiota-based diagnosis for schizophrenia.
Collapse
|
14
|
Gliadin-related antibodies in schizophrenia. Schizophr Res 2018; 195:585-586. [PMID: 28886891 DOI: 10.1016/j.schres.2017.08.051] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 08/25/2017] [Accepted: 08/27/2017] [Indexed: 11/20/2022]
|
15
|
Differential antibody responses to gliadin-derived indigestible peptides in patients with schizophrenia. Transl Psychiatry 2017; 7:e1121. [PMID: 28485731 PMCID: PMC5534957 DOI: 10.1038/tp.2017.89] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 02/24/2017] [Accepted: 03/16/2017] [Indexed: 01/21/2023] Open
Abstract
Gluten consumption has previously been implicated in the development of schizophrenia while an immunological link between gluten and schizophrenia was established by the detection of circulating antibodies against gliadin, a major component of wheat gluten. Several studies have reported an increase in circulating antibodies against native gliadin molecules that are unlikely to survive degradation in the digestive system. In this study, therefore, we measured plasma immunoglobulin G (IgG) and IgA antibodies against indigestible gliadin-derived peptide antigens using an in-house enzyme-linked immunosorbent assay (ELISA) among 169 patients with schizophrenia and 236 control subjects. We also examined the plasma levels of IgG and IgA antibodies against the mixture of native gliadins using commercially available ELISA kits. The results showed that patients with schizophrenia had the increased levels of plasma IgG against the γ-gliadin-derived fragment, namely AAQ6C, but decreased levels of plasma IgG against the α- and γ3-gliadin-derived antigens, as compared with control subjects. This study also demonstrated a uniform decrease in plasma IgA antibodies against gliadin-derived antigens. There was no significant difference in the levels of plasma antibodies against native gliadins between the patient group and the control group. Of eight gliadin-derived antigens tested, four showed a sensitivity of >20% against the specificity of ⩾95% for detection of their corresponding antibodies in plasma. These four tests may thus have a potential to serve as biomarkers for the identification of schizophrenia subgroups that may need an alternative therapy or precision treatment. Further investigation with clinical trials should be carried out to explore this possibility.
Collapse
|
16
|
Ergün C, Urhan M, Ayer A. A review on the relationship between gluten and schizophrenia: Is gluten the cause? Nutr Neurosci 2017; 21:455-466. [PMID: 28393621 DOI: 10.1080/1028415x.2017.1313569] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Schizophrenia is a chronic disease that possesses various clinical manifestations. It presents rather heterogeneous characteristics with respect to onset type, symptoms, and the course of the disease. Although the lifetime prevalence is as low as 1%, it can cause serious disability. Thus, it is very important to develop efficient treatment methods. In some studies, it is hypothesized that removing gluten from the diet leads to a significant improvement in disease symptoms. Epidemiological studies revealed that the prevalence of celiac disease among schizophrenic patients is almost two times higher than that of the general population. OBJECTIVE In this review, we evaluate the effects of gluten and celiac disease on the onset of schizophrenia. Efficacy of gluten-free diet applications, antibody response against gluten, and the interaction of the brain-gut axis and the presence of common genetic points are also investigated. METHODS Without any publication date restriction, Pubmed database searches were made for 'schizophrenia, gluten, gliadin, celiac disease, exorphin, brain-gut axis, psychiatric disorders.' The keywords and the articles about the schizophrenia-celiac disease relationship are included in our review. RESULTS Several studies presented evidence to suggest that symptoms associated with schizophrenia were minimized when gluten was excluded from patients' diets. Immunological searches revealed that most schizophrenic patients with increased anti-gliadin antibodies did not possess celiac disease; yet, the presence of increased antibodies against gliadin can be the share point of the immunological abnormalities found in both of the diseases. DISCUSSION There were no consistent results in the clinical, immunological, microbiological, and epidemiological studies that investigated the relationship between schizophrenia and celiac disease. This presents a need for a larger scale study to confirm the presence of this suggested correlation between schizophrenia and celiac disease. The underlying mechanisms between the two diseases should be explored.
Collapse
Affiliation(s)
- Can Ergün
- a Faculty of Health Sciences, Department of Nutrition and Dietetics , Bahçeşehir University , Beşiktaş, Istanbul , Turkey
| | - Murat Urhan
- b Manisa Mental Health and Diseases Hospital , Şehitler Street, 45020 Manisa , Turkey
| | - Ahmet Ayer
- b Manisa Mental Health and Diseases Hospital , Şehitler Street, 45020 Manisa , Turkey
| |
Collapse
|
17
|
Joseph J, Depp C, Shih PAB, Cadenhead KS, Schmid-Schönbein G. Modified Mediterranean Diet for Enrichment of Short Chain Fatty Acids: Potential Adjunctive Therapeutic to Target Immune and Metabolic Dysfunction in Schizophrenia? Front Neurosci 2017; 11:155. [PMID: 28396623 PMCID: PMC5366345 DOI: 10.3389/fnins.2017.00155] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 03/10/2017] [Indexed: 12/14/2022] Open
Abstract
Growing interest in gut and digestive processes and their potential link to brain and peripheral based inflammation or biobehavioral phenotypes has led to an increasing number of basic and translational scientific reports focused on the role of gut microbiota within the context of neuropsychiatric disorders. However, the effect of dietary modification on specific gut metabolites, in association with immune, metabolic, and psychopathological functioning in schizophrenia spectrum disorders has not been well characterized. The short chain fatty acids (SCFA) acetate, butyrate, and propionate, major metabolites derived from fermentation of dietary fibers by gut microbes, interact with multiple immune and metabolic pathways. The specific pathways that SCFA are thought to target, are dysregulated in cardiovascular disease, type II diabetes, and systemic inflammation. Most notably, these disorders are consistently linked to an attenuated lifespan in schizophrenia. Although, unhealthy dietary intake patterns and increased prevalence of immune and metabolic dysfunction has been observed in people with schizophrenia; dietary interventions have not been well utilized to target immune or metabolic illness. Prior schizophrenia patient trials primarily focused on the effects of gluten free diets. Findings from these studies indicate that a diet avoiding gluten benefits a limited subset of patients, individuals with celiac disease or non-celiac gluten sensitivity. Therefore, alternative dietary and nutritional modifications such as high-fiber, Mediterranean style, diets that enrich the production of SCFA, while being associated with a minimal likelihood of adverse events, may improve immune and cardiovascular outcomes linked to premature mortality in schizophrenia. With a growing literature demonstrating that SCFA can cross the blood brain barrier and target key inflammatory and metabolic pathways, this article highlights enriching dietary intake for SCFA as a potential adjunctive therapy for people with schizophrenia.
Collapse
Affiliation(s)
- Jamie Joseph
- Department of Psychiatry, University of CaliforniaSan Diego, La Jolla, CA, USA
| | - Colin Depp
- Department of Psychiatry, University of CaliforniaSan Diego, La Jolla, CA, USA
- Department of Psychology, VA San Diego Healthcare SystemSan Diego, CA, USA
| | - Pei-an B. Shih
- Department of Psychiatry, University of CaliforniaSan Diego, La Jolla, CA, USA
| | | | | |
Collapse
|
18
|
Rowland LM, Demyanovich HK, Wijtenburg SA, Eaton WW, Rodriguez K, Gaston F, Cihakova D, Talor MV, Liu F, McMahon RR, Hong LE, Kelly DL. Antigliadin Antibodies (AGA IgG) Are Related to Neurochemistry in Schizophrenia. Front Psychiatry 2017; 8:104. [PMID: 28674504 PMCID: PMC5474459 DOI: 10.3389/fpsyt.2017.00104] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 05/29/2017] [Indexed: 12/18/2022] Open
Abstract
Inflammation may play a role in schizophrenia; however, subgroups with immune regulation dysfunction may serve as distinct illness phenotypes with potential different treatment and prevention strategies. Emerging data show that about 30% of people with schizophrenia have elevated antigliadin antibodies of the IgG type, representing a possible subgroup of schizophrenia patients with immune involvement. Also, recent data have shown a high correlation of IgG-mediated antibodies between the periphery and cerebral spinal fluid in schizophrenia but not healthy controls, particularly AGA IgG suggesting that these antibodies may be crossing the blood-brain barrier with resulting neuroinflammation. Proton magnetic resonance spectroscopy (MRS) is a non-invasive technique that allows the quantification of certain neurochemicals in vivo that may proxy inflammation in the brain such as myoinositol and choline-containing compounds (glycerophosphorylcholine and phosphorylcholine). The objective of this exploratory study was to examine the relationship between serum AGA IgG levels and MRS neurochemical levels. We hypothesized that higher AGA IgG levels would be associated with higher levels of myoinositol and choline-containing compounds (glycerophosphorylcholine plus phosphorylcholine; GPC + PC) in the anterior cingulate cortex. Thirty-three participants with a DSM-IV diagnosis of schizophrenia or schizoaffective disorder had blood drawn and underwent neuroimaging using MRS within 9 months. We found that 10/33 (30%) had positive AGA IgG (≥20 U) similar to previous findings. While there were no significant differences in myoinositol and GPC + PC levels between patients with and without AGA IgG positivity, there were significant relationships between both myoinositol (r = 0.475, p = 0.007) and GPC + PC (r = 0.36, p = 0.045) with AGA IgG levels. This study shows a possible connection of AGA IgG antibodies to putative brain inflammation as measured by MRS in schizophrenia.
Collapse
Affiliation(s)
- Laura M Rowland
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Haley K Demyanovich
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States
| | - S Andrea Wijtenburg
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States
| | - William W Eaton
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Katrina Rodriguez
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Frank Gaston
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Daniela Cihakova
- Immunologic Disorders Laboratory, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Monica V Talor
- Immunologic Disorders Laboratory, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Fang Liu
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Robert R McMahon
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States
| | - L Elliot Hong
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Deanna L Kelly
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
19
|
Kynurenine and Tryptophan Levels in Patients With Schizophrenia and Elevated Antigliadin Immunoglobulin G Antibodies. Psychosom Med 2016; 78:931-939. [PMID: 27359171 PMCID: PMC5338470 DOI: 10.1097/psy.0000000000000352] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Several studies have reported an association between nonceliac gluten sensitivity and schizophrenia. Immune and kynurenine (KYN) pathways have also been implicated in the pathophysiology of schizophrenia, and certain proinflammatory immune mediators may increase KYN and reduce tryptophan (TRP) levels. METHODS We measured serum antigliadin immunoglobulin G (IgG), KYN, and TRP in 950 patients with schizophrenia. Patients with antibody level at the 90th percentile or higher of control participants (21.9% of all patients) were classified as having elevated antigliadin IgG. Independent t tests and linear regression models were used to compare TRP, KYN, and KYN-TRP ratio (indicator of TRP metabolism) between patients with and those without elevated antigliadin IgG. The correlation between antigliadin IgG and TRP, KYN, and the ratio was also evaluated in the patients. RESULTS KYN and KYN-TRP ratio were higher in patients with elevated antigliadin IgG (geometric mean [standard deviation {SD}] = 2.65 [0.25] µmol/L versus 2.25 [0.23] µmol/L [p < .001] and 0.05 [0.26] versus 0.04 [0.25; p = .001] respectively), findings robust to adjustment for potential demographic and clinical confounders. Antigliadin IgG positively correlated with KYN and KYN-TRP ratio (r = 0.12, p < .001; r = 0.11, p = .002). TRP did not differ between the two groups and did not correlate with antigliadin IgG. CONCLUSIONS Our results connect nonceliac gluten sensitivity with the KYN pathway of TRP metabolism in psychotic illness and hint toward potential individualized treatment targets.
Collapse
|
20
|
Pandey JP, Namboodiri AM, Elston RC. Immunoglobulin G genotypes and the risk of schizophrenia. Hum Genet 2016; 135:1175-9. [PMID: 27393575 PMCID: PMC5706111 DOI: 10.1007/s00439-016-1706-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 06/25/2016] [Indexed: 12/20/2022]
Abstract
Genes of the immune system are relevant to the etiology of schizophrenia. However, to our knowledge, no large-scale studies, using molecular methods, have been undertaken to investigate the role of highly polymorphic immunoglobulin GM (γ marker) genes in this disorder. In this investigation, we aimed to determine whether particular GM genotypes were associated with susceptibility to schizophrenia. Using a matched case-control study design, we analyzed DNA samples from 798 subjects-398 patients with schizophrenia and 400 controls-obtained from the U.S. National Institute of Mental Health Repository. GM alleles were determined by the TaqMan(®) genotyping assay. The GM 3/3; 23-/23- genotype was highly significantly associated with susceptibility to schizophrenia (p = 0.0002). Subjects with this genotype were over three times (OR 3.4; 95 % CI 1.7-6.7) as likely to develop schizophrenia as those without this genotype. Our results show that immunoglobulin GM genes are risk factors for the development of schizophrenia. Since GM alleles have been implicated in gluten sensitivity and in immunity to neurotropic viruses associated with cognitive impairment, the results presented here may help unify these two disparate areas of pathology affected in this disorder.
Collapse
Affiliation(s)
- Janardan P Pandey
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, 29425, USA.
| | - Aryan M Namboodiri
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | | |
Collapse
|
21
|
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
|
22
|
Trivedi MS, Hodgson NW, Walker SJ, Trooskens G, Nair V, Deth RC. Epigenetic effects of casein-derived opioid peptides in SH-SY5Y human neuroblastoma cells. Nutr Metab (Lond) 2015; 12:54. [PMID: 26664459 PMCID: PMC4673759 DOI: 10.1186/s12986-015-0050-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Accepted: 12/03/2015] [Indexed: 12/18/2022] Open
Abstract
Background Casein-free, gluten-free diets have been reported to mitigate some of the inflammatory gastrointestinal and behavioral traits associated with autism, but the mechanism for this palliative effect has not been elucidated. We recently showed that the opioid peptide beta-casomorphin-7, derived from bovine (bBCM7) milk, decreases cysteine uptake, lowers levels of the antioxidant glutathione (GSH) and decreases the methyl donor S-adenosylmethionine (SAM) in both Caco-2 human GI epithelial cells and SH-SY5Y human neuroblastoma cells. While human breast milk can also release a similar peptide (hBCM-7), the bBCM7 and hBCM-7 vary greatly in potency; as the bBCM-7 is highly potent and similar to morphine in it's effects. Since SAM is required for DNA methylation, we wanted to further investigate the epigenetic effects of these food-derived opioid peptides. In the current study the main objective was to characterize functional pathways and key genes responding to DNA methylation effects of food-derived opioid peptides. Methods SH-SY5Y neuroblastoma cells were treated with 1 μM hBCM7 and bBCM7 and RNA and DNA were isolated after 4 h with or without treatment. Transcriptional changes were assessed using a microarray approach and CpG methylation status was analyzed at 450,000 CpG sites. Functional implications from both endpoints were evaluated via Ingenuity Pathway Analysis 4.0 and KEGG pathway analysis was performed to identify biological interactions between transcripts that were significantly altered at DNA methylation or transcriptional levels (p < 0.05, FDR <0.1). Results Here we show that hBCM7 and bBCM7, as well as morphine, cause epigenetic changes affecting gene pathways related to gastrointestinal disease and inflammation. These epigenetic consequences exhibited the same potency order as opiate inhibition of cysteine uptake insofar as hBCM7 was less potent than bBCM7, which was less potent than morphine. Conclusion Our findings indicate that epigenetic effects of milk-derived opiate peptides may contribute to GI dysfunction and inflammation in sensitive individuals. While the current study was performed using SH-SY5Y neuronal cellular models, similar actions on other cells types might combine to cause symptoms of intolerance. These actions may provide a potential contributing mechanism for the beneficial effects of a casein-free diet in alleviating gastrointestinal symptoms in neurological conditions including autism and other conditions. Lastly, our study also contributes to the evolving awareness of a “gut-brain connection”. Electronic supplementary material The online version of this article (doi:10.1186/s12986-015-0050-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Malav S Trivedi
- Department of Pharmaceutical Sciences, Nova Southeastern University, Rm # 3103, HPD building, Fort Lauderdale, FL USA
| | - Nathaniel W Hodgson
- Department of Molecular and Cellular Biology, Harvard Medical School, Boston, MA USA
| | - Stephen J Walker
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston Salem, NC USA
| | - Geert Trooskens
- Department of Mathematical Modelling, Statistics and Bioinformatics, University of Ghent, Ghent, Belgium
| | - Vineeth Nair
- Department of Pharmaceutical Sciences, Nova Southeastern University, Rm # 3103, HPD building, Fort Lauderdale, FL USA
| | - Richard C Deth
- Department of Pharmaceutical Sciences, Nova Southeastern University, Rm # 3103, HPD building, Fort Lauderdale, FL USA
| |
Collapse
|
23
|
Lionetti E, Leonardi S, Franzonello C, Mancardi M, Ruggieri M, Catassi C. Gluten Psychosis: Confirmation of a New Clinical Entity. Nutrients 2015; 7:5532-5539. [PMID: 26184290 PMCID: PMC4517012 DOI: 10.3390/nu7075235] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Revised: 06/17/2015] [Accepted: 07/02/2015] [Indexed: 12/13/2022] Open
Abstract
Non-celiac gluten sensitivity (NCGS) is a syndrome diagnosed in patients with symptoms that respond to removal of gluten from the diet, after celiac disease and wheat allergy have been excluded. NCGS has been related to neuro-psychiatric disorders, such as autism, schizophrenia and depression. A singular report of NCGS presenting with hallucinations has been described in an adult patient. We report a pediatric case of a psychotic disorder clearly related to NCGS and investigate the causes by a review of literature. The pathogenesis of neuro-psychiatric manifestations of NCGS is unclear. It has been hypothesized that: (a) a "leaky gut" allows some gluten peptides to cross the intestinal membrane and the blood brain barrier, affecting the endogenous opiate system and neurotransmission; or (b) gluten peptides may set up an innate immune response in the brain similar to that described in the gut mucosa, causing exposure from neuronal cells of a transglutaminase primarily expressed in the brain. The present case-report confirms that psychosis may be a manifestation of NCGS, and may also involve children; the diagnosis is difficult with many cases remaining undiagnosed. Well-designed prospective studies are needed to establish the real role of gluten as a triggering factor in neuro-psychiatric disorders.
Collapse
Affiliation(s)
- Elena Lionetti
- Department of Pediatrics, University of Catania, Via S. Sofia 78, 95124 Catania, Italy.
| | - Salvatore Leonardi
- Department of Pediatrics, University of Catania, Via S. Sofia 78, 95124 Catania, Italy.
| | - Chiara Franzonello
- Department of Pediatrics, University of Catania, Via S. Sofia 78, 95124 Catania, Italy.
| | - Margherita Mancardi
- Pediatric Neuro-Psychiatric Unit, G. Gaslini Institute, Via Gerolamo Gaslini 5, 16147 Genova, Italy.
| | - Martino Ruggieri
- Department of Pediatrics, University of Catania, Via S. Sofia 78, 95124 Catania, Italy.
| | - Carlo Catassi
- Department of Pediatrics, Marche Polytechnic University, Ancona, Via Corridoni, 11, 60123 Ancona, Italy.
- The Division of Paediatric Gastroenterology and Nutrition and Center for Celiac Research, MassGeneral Hospital for Children, 55 Fruit Street, Boston, MA 02114, USA.
| |
Collapse
|
24
|
Avramopoulos D, Pearce BD, McGrath J, Wolyniec P, Wang R, Eckart N, Hatzimanolis A, Goes FS, Nestadt G, Mulle J, Coneely K, Hopkins M, Ruczinski I, Yolken R, Pulver AE. Infection and inflammation in schizophrenia and bipolar disorder: a genome wide study for interactions with genetic variation. PLoS One 2015; 10:e0116696. [PMID: 25781172 PMCID: PMC4363491 DOI: 10.1371/journal.pone.0116696] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 12/12/2014] [Indexed: 12/13/2022] Open
Abstract
Inflammation and maternal or fetal infections have been suggested as risk factors for schizophrenia (SZ) and bipolar disorder (BP). It is likely that such environmental effects are contingent on genetic background. Here, in a genome-wide approach, we test the hypothesis that such exposures increase the risk for SZ and BP and that the increase is dependent on genetic variants. We use genome-wide genotype data, plasma IgG antibody measurements against Toxoplasma gondii, Herpes simplex virus type 1, Cytomegalovirus, Human Herpes Virus 6 and the food antigen gliadin as well as measurements of C-reactive protein (CRP), a peripheral marker of inflammation. The subjects are SZ cases, BP cases, parents of cases and screened controls. We look for higher levels of our immunity/infection variables and interactions between them and common genetic variation genome-wide. We find many of the antibody measurements higher in both disorders. While individual tests do not withstand correction for multiple comparisons, the number of nominally significant tests and the comparisons showing the expected direction are in significant excess (permutation p=0.019 and 0.004 respectively). We also find CRP levels highly elevated in SZ, BP and the mothers of BP cases, in agreement with existing literature, but possibly confounded by our inability to correct for smoking or body mass index. In our genome-wide interaction analysis no signal reached genome-wide significance, yet many plausible candidate genes emerged. In a hypothesis driven test, we found multiple interactions among SZ-associated SNPs in the HLA region on chromosome 6 and replicated an interaction between CMV infection and genotypes near the CTNNA3 gene reported by a recent GWAS. Our results support that inflammatory processes and infection may modify the risk for psychosis and suggest that the genotype at SZ-associated HLA loci modifies the effect of these variables on the risk to develop SZ.
Collapse
Affiliation(s)
- Dimitrios Avramopoulos
- McKusick Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
- * E-mail:
| | - Brad D. Pearce
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, United States of America
| | - John McGrath
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Paula Wolyniec
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Ruihua Wang
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Nicole Eckart
- McKusick Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Alexandros Hatzimanolis
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Fernando S. Goes
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Gerald Nestadt
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Jennifer Mulle
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, United States of America
| | - Karen Coneely
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, United States of America
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Myfanwy Hopkins
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, United States of America
| | - Ingo Ruczinski
- Bloomberg School of Public Heath, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Robert Yolken
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Ann E. Pulver
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| |
Collapse
|
25
|
Antibody against α-gliadin 33-mer peptide: Is the key initiating factor for development of multiple sclerosis during gluten sensitivity? JOURNAL OF MEDICAL HYPOTHESES AND IDEAS 2015. [DOI: 10.1016/j.jmhi.2015.02.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
26
|
Severance EG, Gressitt KL, Alaedini A, Rohleder C, Enning F, Bumb JM, Müller JK, Schwarz E, Yolken RH, Leweke FM. IgG dynamics of dietary antigens point to cerebrospinal fluid barrier or flow dysfunction in first-episode schizophrenia. Brain Behav Immun 2015; 44:148-58. [PMID: 25241021 PMCID: PMC4275312 DOI: 10.1016/j.bbi.2014.09.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 08/29/2014] [Accepted: 09/08/2014] [Indexed: 12/30/2022] Open
Abstract
Schizophrenia is a complex brain disorder that may be accompanied by idiopathic inflammation. Classic central nervous system (CNS) inflammatory disorders such as viral encephalitis or multiple sclerosis can be characterized by incongruent serum and cerebrospinal fluid (CSF) IgG due in part to localized intrathecal synthesis of antibodies. The dietary antigens, wheat gluten and bovine milk casein, can induce a humoral immune response in susceptible individuals with schizophrenia, but the correlation between the food-derived serological and intrathecal IgG response is not known. Here, we measured IgG to wheat gluten and bovine milk casein in matched serum and CSF samples from 105 individuals with first-episode schizophrenia (n=75 antipsychotic-naïve), and 61 controls. We found striking correlations in the levels of IgG response to dietary proteins between serum and CSF of schizophrenia patients, but not controls (schizophrenia, R(2)=0.34-0.55, p⩽0.0001; controls R(2)=0.05-0.06, p>0.33). A gauge of blood-CSF barrier permeability and CSF flow rate, the CSF-to-serum albumin ratio, was significantly elevated in cases compared to controls (p⩽0.001-0.003). Indicators of intrathecal IgG production, the CSF IgG index and the specific Antibody Index, were not significantly altered in schizophrenia compared to controls. Thus, the selective diffusion of bovine milk casein and wheat gluten antibodies between serum and CSF in schizophrenia may be the function of a low-level anatomical barrier dysfunction or altered CSF flow rate, which may be transient in nature.
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,Correspondence: Emily G. Severance, , tel: +1 410-614-3918, fax: +1 410-955-3723
| | - Kristin L. Gressitt
- 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
| | - Armin Alaedini
- Department of Medicine, Columbia University Medical Center, 1130 Saint Nicholas Ave., ICRC 901B, New York, NY, 10032 U.S.A
| | - Cathrin Rohleder
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Frank Enning
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany,Department of Psychosomatics and Psychotherapeutic Medicine, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - J. Malte Bumb
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Juliane K. Müller
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Emanuel Schwarz
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - 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
| | - F. Markus Leweke
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
27
|
Schizophrenia and the gut-brain axis. Prog Neuropsychopharmacol Biol Psychiatry 2015; 56:155-60. [PMID: 25240858 DOI: 10.1016/j.pnpbp.2014.08.018] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 08/26/2014] [Accepted: 08/27/2014] [Indexed: 12/26/2022]
Abstract
Several risk factors for the development of schizophrenia can be linked through a common pathway in the intestinal tract. It is now increasingly recognized that bidirectional communication exists between the brain and the gut that uses neural, hormonal, and immunological routes. An increased incidence of gastrointestinal (GI) barrier dysfunction, food antigen sensitivity, inflammation, and the metabolic syndrome is seen in schizophrenia. These findings may be influenced by the composition of the gut microbiota. A significant subgroup of patients may benefit from the initiation of a gluten and casein-free diet. Antimicrobials and probiotics have therapeutic potential for reducing the metabolic dysfunction and immune dysregulation seen in patients with schizophrenia.
Collapse
|
28
|
Cook TB, Brenner LA, Cloninger CR, Langenberg P, Igbide A, Giegling I, Hartmann AM, Konte B, Friedl M, Brundin L, Groer MW, Can A, Rujescu D, Postolache TT. "Latent" infection with Toxoplasma gondii: association with trait aggression and impulsivity in healthy adults. J Psychiatr Res 2015; 60:87-94. [PMID: 25306262 DOI: 10.1016/j.jpsychires.2014.09.019] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Revised: 09/03/2014] [Accepted: 09/19/2014] [Indexed: 11/28/2022]
Abstract
BACKGROUND Latent chronic infection with Toxoplasma gondii (T. gondii), a common neurotropic pathogen, has been previously linked with suicidal self-directed violence (SSDV). We sought to determine if latent infection with T. gondii is associated with trait aggression and impulsivity, intermediate phenotypes for suicidal behavior, in psychiatrically healthy adults. METHODS Traits of aggression and impulsivity were analyzed in relationship to IgG antibody seropositivity for T. gondii and two other latent neurotropic infections, herpes simplex virus 1 (HSV1) and cytomegalovirus (CMV). One thousand community-residing adults residing in the Munich metropolitan area with no Axis I or II conditions by SCID for DSM-IV (510 men, 490 women, mean age 53.6 ± 15.8, range 20-74). Plasma samples were tested for IgG antibodies to T. gondii, HSV-1 and CMV by ELISA. Self-reported ratings of trait aggression scores (Questionnaire for Measuring Factors of Aggression [FAF]) and trait impulsivity (Sensation-Seeking Scale-V [SSS-V]) were analyzed using linear multivariate methods. RESULTS T. gondii IgG seropositivity was significantly associated with higher trait reactive aggression scores among women (p < .01), but not among men. T. gondii-positivity was also associated with higher impulsive sensation-seeking (SSS-V Disinhibition) among younger men (p < .01) aged 20-59 years old (median age = 60). All associations with HSV-1 and CMV were not significant. CONCLUSIONS Aggression and impulsivity, personality traits considered as endophenotypes for SSDV, are associated with latent T. gondii infection in a gender and age-specific manner, and could be further investigated as prognostic and treatment targets in T. gondii-positive individuals at risk for SSDV.
Collapse
Affiliation(s)
- Thomas B Cook
- Department of Public Health, Mercyhurst Institute for Public Health, Mercyhurst University, Erie, PA, USA
| | - Lisa A Brenner
- Veterans Integrated Service Network (VISN) 19, Mental Illness Research Education and Clinical Center (MIRECC), Denver, CO, USA; Department of Psychiatry, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA; Department of Physical Medicine and Rehabilitation, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA; Department of Neurology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - C Robert Cloninger
- Department of Psychiatry, Sansone Centre for Well-Being, Washington University, St. Louis, MO, USA
| | - Patricia Langenberg
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Ajirioghene Igbide
- DC Department of Behavioral Health, Saint Elizabeths Hospital, Psychiatry Residency Program, Washington, DC, USA
| | - Ina Giegling
- Department of Psychiatry, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Annette M Hartmann
- Department of Psychiatry, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Bettina Konte
- Department of Psychiatry, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Marion Friedl
- Department of Psychiatry, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Lena Brundin
- Division of Psychiatry and Behavioral Medicine, College of Human Medicine, Michigan State University, Van Andel Research Institute, Grand Rapids, MI, USA
| | | | - Adem Can
- Department of Psychiatry, University of Maryland-Baltimore School of Medicine, Baltimore, MD, USA
| | - Dan Rujescu
- Department of Psychiatry, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Teodor T Postolache
- Veterans Integrated Service Network (VISN) 19, Mental Illness Research Education and Clinical Center (MIRECC), Denver, CO, USA; Department of Psychiatry, University of Maryland-Baltimore School of Medicine, Baltimore, MD, USA; Veterans Integrated Service Network (VISN) 5, Mental Illness Research Education and Clinical Center (MIRECC), Baltimore, MD, USA.
| |
Collapse
|
29
|
Jackson J, Eaton W, Cascella N, Fasano A, Santora D, Sullivan K, Feldman S, Raley H, McMahon RP, Carpenter WT, Demyanovich H, Kelly DL. Gluten sensitivity and relationship to psychiatric symptoms in people with schizophrenia. Schizophr Res 2014; 159:539-42. [PMID: 25311778 PMCID: PMC4476307 DOI: 10.1016/j.schres.2014.09.023] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 09/09/2014] [Accepted: 09/11/2014] [Indexed: 12/20/2022]
Abstract
The relationship between gluten sensitivity and schizophrenia has been of increasing interest and novel mechanisms explaining this relationship continue to be described. Our study in 100 people with schizophrenia compared to 100 matched controls replicates a higher prevalence of gluten sensitivity and higher mean antigliadin IgG antibody levels schizophrenia (2.9 ± 7.7 vs. 1.3 ± 1.3, p = 0.046, controlled for age). Additionally, we examined symptoms within the schizophrenia group and found that while positive symptoms are significantly lower in people who have elevated antigliadin antibodies (AGA; 4.11 ± 1.36 vs. 6.39 ± 2.99, p = 0.020), no robust clinical profile differentiates between positive and negative antibody groups. Thus, identifying people in schizophrenia who may benefit from a gluten-free diet remains possible by blood test only.
Collapse
Affiliation(s)
- Jessica Jackson
- Temple University School of Medicine, 3500 North Broad Street, Philadelphia, PA 19140, USA
| | - William Eaton
- Johns Hopkins Bloomberg School of Public Health, 624 North Broadway, Baltimore, MD 21205, USA
| | - Nicola Cascella
- Neuropsychiatry Program Sheppard Pratt Hospital, 6501 North Charles Street, Baltimore, MD 21285, USA
| | - Alessio Fasano
- Massachusetts General Hospital for Children, 175 Cambridge Street, Boston, MA 02114, USA; Massachusetts General Hospital East, 16th Street, Charlestown, MA 02129, USA
| | - Debby Santora
- University of Maryland School of Medicine, 660 West Redwood Street, Baltimore, MD 21201, USA
| | - Kelli Sullivan
- University of Maryland School of Medicine, Department of Psychiatry, Maryland Psychiatric Research Center, 55 Wade Avenue, Catonsville, MD 21228, USA
| | - Stephanie Feldman
- University of Maryland School of Medicine, Department of Psychiatry, Maryland Psychiatric Research Center, 55 Wade Avenue, Catonsville, MD 21228, USA
| | - Heather Raley
- National Institute on Drug Abuse, 251 Bayview Blvd., Baltimore, MD 21223, USA
| | - Robert P McMahon
- University of Maryland School of Medicine, Department of Psychiatry, Maryland Psychiatric Research Center, 55 Wade Avenue, Catonsville, MD 21228, USA
| | - William T Carpenter
- University of Maryland School of Medicine, Department of Psychiatry, Maryland Psychiatric Research Center, 55 Wade Avenue, Catonsville, MD 21228, USA
| | - Haley Demyanovich
- University of Maryland School of Medicine, Department of Psychiatry, Maryland Psychiatric Research Center, 55 Wade Avenue, Catonsville, MD 21228, USA
| | - Deanna L Kelly
- University of Maryland School of Medicine, Department of Psychiatry, Maryland Psychiatric Research Center, 55 Wade Avenue, Catonsville, MD 21228, USA.
| |
Collapse
|
30
|
Porcelli B, Verdino V, Bossini L, Terzuoli L, Fagiolini A. Celiac and non-celiac gluten sensitivity: a review on the association with schizophrenia and mood disorders. AUTOIMMUNITY HIGHLIGHTS 2014; 5:55-61. [PMID: 26000156 PMCID: PMC4389040 DOI: 10.1007/s13317-014-0064-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 10/01/2014] [Indexed: 12/26/2022]
Abstract
An association between many psychiatric and gluten-related disorders has been known for some time. In the case of schizophrenia and mood disorders, the major psychiatric disorders, there is much evidence, not without contradictions, of a possible association between schizophrenia and celiac disease. The association between mood disorders and gluten-related disorders, especially celiac disease, has only been studied for depression, often coupled with anxiety, and very recently for bipolar disorder. Since non-celiac gluten sensitivity is now known to be different from celiac disease, many studies have shown that gluten sensitivity is also associated with major psychiatric disorders. Here we review the literature on the association between schizophrenia/mood disorders and celiac disease/gluten sensitivity, pointing out the differences between these associations.
Collapse
Affiliation(s)
- Brunetta Porcelli
- Biochemistry Division, Department of Medical Biotechnologies, Siena University, Polo Scientifico Universitario di San Miniato Via Alcide De Gasperi 2, 53100 Siena, Italy
| | - Valeria Verdino
- Psychiatry Division, Department of Molecular and Developmental Medicine, Siena University, Policlinico Le Scotte Viale Bracci 1, 53100 Siena, Italy
| | - Letizia Bossini
- Psychiatry Division, Department of Molecular and Developmental Medicine, Siena University, Policlinico Le Scotte Viale Bracci 1, 53100 Siena, Italy
| | - Lucia Terzuoli
- Biochemistry Division, Department of Medical Biotechnologies, Siena University, Polo Scientifico Universitario di San Miniato Via Alcide De Gasperi 2, 53100 Siena, Italy
| | - Andrea Fagiolini
- Psychiatry Division, Department of Molecular and Developmental Medicine, Siena University, Policlinico Le Scotte Viale Bracci 1, 53100 Siena, Italy
| |
Collapse
|
31
|
Trivedi MS, Shah JS, Al-Mughairy S, Hodgson NW, Simms B, Trooskens GA, Van Criekinge W, Deth RC. Food-derived opioid peptides inhibit cysteine uptake with redox and epigenetic consequences. J Nutr Biochem 2014; 25:1011-8. [PMID: 25018147 DOI: 10.1016/j.jnutbio.2014.05.004] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 04/07/2014] [Accepted: 05/05/2014] [Indexed: 12/17/2022]
Abstract
Dietary interventions like gluten-free and casein-free diets have been reported to improve intestinal, autoimmune and neurological symptoms in patients with a variety of conditions; however, the underlying mechanism of benefit for such diets remains unclear. Epigenetic programming, including CpG methylation and histone modifications, occurring during early postnatal development can influence the risk of disease in later life, and such programming may be modulated by nutritional factors such as milk and wheat, especially during the transition from a solely milk-based diet to one that includes other forms of nutrition. The hydrolytic digestion of casein (a major milk protein) and gliadin (a wheat-derived protein) releases peptides with opioid activity, and in the present study, we demonstrate that these food-derived proline-rich opioid peptides modulate cysteine uptake in cultured human neuronal and gastrointestinal (GI) epithelial cells via activation of opioid receptors. Decreases in cysteine uptake were associated with changes in the intracellular antioxidant glutathione and the methyl donor S-adenosylmethionine. Bovine and human casein-derived opioid peptides increased genome-wide DNA methylation in the transcription start site region with a potency order similar to their inhibition of cysteine uptake. Altered expression of genes involved in redox and methylation homeostasis was also observed. These results illustrate the potential of milk- and wheat-derived peptides to exert antioxidant and epigenetic changes that may be particularly important during the postnatal transition from placental to GI nutrition. Differences between peptides derived from human and bovine milk may contribute to developmental differences between breastfed and formula-fed infants. Restricted antioxidant capacity, caused by wheat- and milk-derived opioid peptides, may predispose susceptible individuals to inflammation and systemic oxidation, partly explaining the benefits of gluten-free or casein-free diets.
Collapse
Affiliation(s)
- Malav S Trivedi
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
| | - Jayni S Shah
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
| | - Sara Al-Mughairy
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
| | - Nathaniel W Hodgson
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
| | - Benjamin Simms
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
| | - Geert A Trooskens
- Department of Mathematical Modelling, Statistics and Bioinformatics, Faculty of Bioscience Engineering, Ghent University, Ghent 9000, Belgium
| | - Wim Van Criekinge
- Department of Mathematical Modelling, Statistics and Bioinformatics, Faculty of Bioscience Engineering, Ghent University, Ghent 9000, Belgium
| | - Richard C Deth
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA.
| |
Collapse
|
32
|
Okusaga O, Muravitskaja O, Fuchs D, Ashraf A, Hinman S, Giegling I, Hartmann AM, Konte B, Friedl M, Schiffman J, Hong E, Reeves G, Groer M, Dantzer R, Rujescu D, Postolache TT. Elevated levels of plasma phenylalanine in schizophrenia: a guanosine triphosphate cyclohydrolase-1 metabolic pathway abnormality? PLoS One 2014; 9:e85945. [PMID: 24465804 PMCID: PMC3897774 DOI: 10.1371/journal.pone.0085945] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 12/04/2013] [Indexed: 02/07/2023] Open
Abstract
Background Phenylalanine and tyrosine are precursor amino acids required for the synthesis of dopamine, the main neurotransmitter implicated in the neurobiology of schizophrenia. Inflammation, increasingly implicated in schizophrenia, can impair the function of the enzyme Phenylalanine hydroxylase (PAH; which catalyzes the conversion of phenylalanine to tyrosine) and thus lead to elevated phenylalanine levels and reduced tyrosine levels. This study aimed to compare phenylalanine, tyrosine, and their ratio (a proxy for PAH function) in a relatively large sample of schizophrenia patients and healthy controls. Methods We measured non-fasting plasma phenylalanine and tyrosine in 950 schizophrenia patients and 1000 healthy controls. We carried out multivariate analyses to compare log transformed phenylalanine, tyrosine, and phenylalanine:tyrosine ratio between patients and controls. Results Compared to controls, schizophrenia patients had higher phenylalanine (p<0.0001) and phenylalanine: tyrosine ratio (p<0.0001) but tyrosine did not differ between the two groups (p = 0.596). Conclusions Elevated phenylalanine and phenylalanine:tyrosine ratio in the blood of schizophrenia patients have to be replicated in longitudinal studies. The results may relate to an abnormal PAH function in schizophrenia that could become a target for novel preventative and interventional approaches.
Collapse
Affiliation(s)
- Olaoluwa Okusaga
- Mood and Anxiety Program, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Olesja Muravitskaja
- Mood and Anxiety Program, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Dietmar Fuchs
- Division of Biological Chemistry, Biocenter Innsbruck Medical University, Innsbruck, Austria
| | - Ayesha Ashraf
- Mood and Anxiety Program, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Sarah Hinman
- Mood and Anxiety Program, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Ina Giegling
- Section of Molecular and Clinical Neurobiology, Ludwig Maximilians University, Munich, Germany
| | - Annette M. Hartmann
- Section of Molecular and Clinical Neurobiology, Ludwig Maximilians University, Munich, Germany
| | - Bettina Konte
- Section of Molecular and Clinical Neurobiology, Ludwig Maximilians University, Munich, Germany
| | - Marion Friedl
- Section of Molecular and Clinical Neurobiology, Ludwig Maximilians University, Munich, Germany
| | - Jason Schiffman
- Department of Psychology, University of Maryland, Baltimore, Maryland, United States of America
| | - Elliot Hong
- Maryland Psychiatric Research Center (MPRC), Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Gloria Reeves
- Division of Child and Adolescent Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- University of Maryland Child and Adolescent Mental Health Innovations Center, Baltimore, Maryland, United States of America
| | - Maureen Groer
- University of South Florida, Tampa, Florida, United States of America
| | - Robert Dantzer
- The University of Texas, MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Dan Rujescu
- Section of Molecular and Clinical Neurobiology, Ludwig Maximilians University, Munich, Germany
- Department of Psychiatry, University of Halle-Wittenberg, Halle, Germany
| | - Teodor T. Postolache
- Mood and Anxiety Program, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- University of Maryland Child and Adolescent Mental Health Innovations Center, Baltimore, Maryland, United States of America
- VISN 5 Capitol Health Care Network Mental Illness Research Education and Clinical Center (MIRECC), Baltimore, Maryland, United States of America
- VISN 19 MIRECC, Denver, Colorado, United States of America
- * E-mail:
| |
Collapse
|
33
|
Okusaga O, Hamilton RG, Can A, Igbide A, Giegling I, Hartmann AM, Konte B, Friedl M, Reeves GM, Rujescu D, Postolache TT. Phadiatop Seropositivity in Schizophrenia Patients and Controls: A Preliminary Study. AIMS Public Health 2014; 1:43-50. [PMID: 25346942 PMCID: PMC4205967 DOI: 10.3934/publichealth.2014.2.43] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
There is a dearth of information on the association of atopy with schizophrenia. The few available studies used population-based registers to classify the atopy status of the patients but this strategy is not reliable. This study measured seropositivity with a multiallergen screen of allergen specific IgE antibodies in schizophrenia patients versus healthy controls. A subset of 66 schizophrenia patients and 34 healthy controls were randomly selected from a large comparative study of schizophrenia patients and controls. The Phadiatop multi-allergen screen was performed on sera from all the participants to assess their atopic status. Logistic regression was used to calculate the odds ratio for the association of schizophrenia with Phadiatop seropositivity as a measure of atopy. The prevalence of Phadiatop seropositivity was significantly lower (χ2 4.59, p = 0.032) and there was a reduced odds ratio for atopy in schizophrenia patients relative to controls (OR 0.40; 95% CI 0.17 to 0.94, p = 0.036). Though limited by a relatively small sample size and potentially confounded by anti-psychotic medications, this study suggests that the prevalence of atopy is lower in patients with schizophrenia. Replicating these results in larger samples could add to our growing understanding of immunological implications in mental illness.
Collapse
Affiliation(s)
- Olaoluwa Okusaga
- Mood and Anxiety Program, University of Maryland School of Medicine, Baltimore, MD USA ; Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, TX, USA
| | - Robert G Hamilton
- Johns Hopkins Dermatology, Allergy and Clinical Immunology Reference Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Adem Can
- Mood and Anxiety Program, University of Maryland School of Medicine, Baltimore, MD USA
| | - Ajirioghene Igbide
- Mood and Anxiety Program, University of Maryland School of Medicine, Baltimore, MD USA ; St. Elizabeths Hospital, Psychiatry Residency Training Program, Washington, DC, USA
| | - Ina Giegling
- Department of Psychiatry, University of Halle-Wittenberg, Germany
| | | | - Bettina Konte
- Department of Psychiatry, University of Halle-Wittenberg, Germany
| | - Marion Friedl
- Department of Psychiatry, University of Halle-Wittenberg, Germany
| | - Gloria M Reeves
- Division of Child and Adolescent Psychiatry, University of Maryland, School of Medicine, Baltimore, Maryland, USA ; University of Maryland Child and Adolescent Mental Health Innovations Center, Baltimore, Maryland, USA
| | - Dan Rujescu
- Department of Psychiatry, University of Halle-Wittenberg, Germany
| | - Teodor T Postolache
- Mood and Anxiety Program, University of Maryland School of Medicine, Baltimore, MD USA ; University of Maryland Child and Adolescent Mental Health Innovations Center, Baltimore, Maryland, USA
| |
Collapse
|