1
|
Wang H, Xie C, Deng B, Ding J, Li N, Kou Z, Jin M, He J, Wang Q, Wen H, Zhang J, Zhou Q, Chen S, Chen X, Yuan TF, Zhu S. Structural basis for antibody-mediated NMDA receptor clustering and endocytosis in autoimmune encephalitis. Nat Struct Mol Biol 2024:10.1038/s41594-024-01387-3. [PMID: 39227720 DOI: 10.1038/s41594-024-01387-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 08/07/2024] [Indexed: 09/05/2024]
Abstract
Antibodies against N-methyl-D-aspartate receptors (NMDARs) are most frequently detected in persons with autoimmune encephalitis (AE) and used as diagnostic biomarkers. Elucidating the structural basis of monoclonal antibody (mAb) binding to NMDARs would facilitate the development of targeted therapy for AE. Here, we reconstructed nanodiscs containing green fluorescent protein-fused NMDARs to label and sort individual immune B cells from persons with AE and further cloned and identified mAbs against NMDARs. This allowed cryo-electron microscopy analysis of NMDAR-Fab complexes, revealing that autoantibodies bind to the R1 lobe of the N-terminal domain of the GluN1 subunit. Small-angle X-ray scattering studies demonstrated NMDAR-mAb stoichiometry of 2:1 or 1:2, structurally suitable for mAb-induced clustering and endocytosis of NMDARs. Importantly, these mAbs reduced the surface NMDARs and NMDAR-mediated currents, without tonically affecting NMDAR channel gating. These structural and functional findings imply that the design of neutralizing antibody binding to the R1 lobe of NMDARs represents a potential therapy for AE treatment.
Collapse
Affiliation(s)
- Han Wang
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Chun Xie
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Bo Deng
- Department of Neurology, Huashan Hospital and Institute of Neurology, National Center for Neurological Disorders, Fudan University, Shanghai, China
| | - Jinjun Ding
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine and School of Psychology, Shanghai, China
| | - Na Li
- National Facility for Protein Science in Shanghai, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, China
| | - Zengwei Kou
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Mengmeng Jin
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jie He
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | | | - Han Wen
- DP Technology, Beijing, China
| | - Jinbao Zhang
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Qinming Zhou
- Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Sheng Chen
- Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Xiangjun Chen
- Department of Neurology, Huashan Hospital and Institute of Neurology, National Center for Neurological Disorders, Fudan University, Shanghai, China.
| | - Ti-Fei Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine and School of Psychology, Shanghai, China.
| | - Shujia Zhu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
2
|
Liu Z, Wang Y, Wang S, Wu J, Jia C, Tan X, Liu X, Huang X, Zhang L. Unraveling the causative connection between urticaria, inflammatory cytokines, and mental disorders: Perspectives from genetic evidence. Skin Res Technol 2024; 30:e13906. [PMID: 39300828 PMCID: PMC11413335 DOI: 10.1111/srt.13906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 07/05/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND The genetic association between urticaria and mental disorders and whether inflammatory cytokines mediate this process remains unclear. MATERIALS AND METHODS A Mendelian randomization (MR) approaches to elucidate the causal relationship between urticaria and mental disorders and to validate the mediation of inflammatory cytokines. Genome-wide association study (GWAS) databases used were obtained from Psychiatric Genomics Cooperation (PGC), GWAS Catalog, and FinnGen Consortium. Our study was conducted using inverse variance weighted (IVW) and Bayesian weighted MR (BWMR) methods for joint analysis. RESULTS The MR results showed that urticaria increased the risk of attention deficit hyperactivity disorder (ADHD) (odds ratio [OR] = $ = $ 1.088, 95% confidence interval [CI]: 1.026-1.154, p = $ = $ 0.0051); cholinergic urticaria increased the risk of bipolar disorder (BD) (OR = $ = $ 1.012, 95% CI: 1.001-1.022, p = $ = $ 0.0274); dermatographic urticaria increased the risk of ADHD (OR = $ = $ 1.057, 95% CI: 1.005-1.112, p = $ = $ 0.0323); idiopathic urticaria increased the risk of schizophrenia (SCZ) (OR = $ = $ 1.057, 95% CI: 1.005-1.112, p = $ = $ 0.0323); other unspecified urticaria increased the risk of ADHD (OR = $ = $ 1.085, 95% CI: 1.023-1.151, p = $ = $ 0.0063). We found that eight inflammatory cytokines were negatively associated with mental disorders and seven inflammatory cytokines were positively associated with mental disorders. Finally, our results suggested that inflammatory cytokines do not act as mediators between urticaria and mental disorders. CONCLUSIONS Our study reveals a causal relationship between urticaria and the increased risk of mental disorders. We suggest that the treatment of urticaria could incorporate psychiatric interventions and mental health assessment of patients.
Collapse
Affiliation(s)
- ZhiRong Liu
- Department of General SurgeryChengdu Second People's HospitalChengduChina
| | | | - ShiHao Wang
- School of Biological Science and TechnologyChengdu Medical CollegeChengduChina
| | - JiaXin Wu
- Graduate SchoolChengdu Medical CollegeChengduChina
| | - Cui Jia
- Development and Regeneration Key Laboratory of Sichuan ProvinceInstitute of NeuroscienceDepartment of Pathology and PathophysiologyChengdu Medical CollegeChengduChina
| | - Xuan Tan
- School of Biological Science and TechnologyChengdu Medical CollegeChengduChina
| | - XinLian Liu
- Development and Regeneration Key Laboratory of Sichuan ProvinceInstitute of NeuroscienceDepartment of Pathology and PathophysiologyChengdu Medical CollegeChengduChina
| | - XinWei Huang
- Shanghai Key Laboratory of Anesthesiology and Brain Functional ModulationClinical Research Center for Anesthesiology and Perioperative MedicineTranslational Research Institute of Brain and Brain‐Like IntelligenceShanghai Fourth People's HospitalSchool of MedicineTongji UniversityShanghaiChina
| | - LuShun Zhang
- Development and Regeneration Key Laboratory of Sichuan ProvinceInstitute of NeuroscienceDepartment of Pathology and PathophysiologyChengdu Medical CollegeChengduChina
| |
Collapse
|
3
|
McKeon A, Pittock SJ. Overview and Diagnostic Approach in Autoimmune Neurology. Continuum (Minneap Minn) 2024; 30:960-994. [PMID: 39088285 DOI: 10.1212/con.0000000000001447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/03/2024]
Abstract
OBJECTIVE The field of autoimmune neurology is rapidly evolving. This article reviews the epidemiology and pathophysiology as well as current approaches to clinical and paraclinical assessment, testing paradigms, and general principles of treatment. LATEST DEVELOPMENTS Improved recognition of autoimmune diagnoses among patients who have phenotypically diverse, subacute onset neurologic presentations is facilitated by disease-specific antibody biomarker discovery. These antibodies have varying associations with paraneoplastic causation (from no association to greater than 70% positive predictive value), immunotherapy responses, and outcomes. To simplify assessment in an increasingly complex discipline, neurologic phenotype-specific serum and CSF antibody evaluations are recommended. Clinical trials have led to the approval of monoclonal therapies for neuromyelitis optica spectrum disorder (NMOSD) and are underway for N-methyl-d-aspartate (NMDA) receptor and leucine-rich glioma inactivated protein 1 (LGI1) encephalitides. ESSENTIAL POINTS Autoimmune neurology is now a mainstream subspecialty, consisting of disorders with diverse presentations detectable using antibody testing of serum and CSF. Early and sustained immunotherapy (eg, corticosteroids, intravenous immunoglobulin [IVIg], plasma exchange) is recommended and may be supplemented by immune suppressants (eg, rituximab or cyclophosphamide) to sustain responses and optimize outcomes.
Collapse
|
4
|
Guma E, Chakravarty MM. Immune Alterations in the Intrauterine Environment Shape Offspring Brain Development in a Sex-Specific Manner. Biol Psychiatry 2024:S0006-3223(24)01260-5. [PMID: 38679357 PMCID: PMC11511788 DOI: 10.1016/j.biopsych.2024.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 03/20/2024] [Accepted: 04/17/2024] [Indexed: 05/01/2024]
Abstract
Exposure to immune dysregulation in utero or in early life has been shown to increase risk for neuropsychiatric illness. The sources of inflammation can be varied, including acute exposures due to maternal infection or acute stress, or persistent exposures due to chronic stress, obesity, malnutrition, or autoimmune diseases. These exposures may cause subtle alteration in brain development, structure, and function that can become progressively magnified across the life span, potentially increasing the likelihood of developing a neuropsychiatric conditions. There is some evidence that males are more susceptible to early-life inflammatory challenges than females. In this review, we discuss the various sources of in utero or early-life immune alteration and the known effects on fetal development with a sex-specific lens. To do so, we leveraged neuroimaging, behavioral, cellular, and neurochemical findings. Gaining clarity about how the intrauterine environment affects offspring development is critically important for informing preventive and early intervention measures that may buffer against the effects of these early-life risk factors.
Collapse
Affiliation(s)
- Elisa Guma
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health, Bethesda, Maryland; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts.
| | - M Mallar Chakravarty
- Computational Brain Anatomy Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada; Department of Biomedical Engineering, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
5
|
Kiyat P, Karti O, Gercik Ö, Şak T. Choroidal, retinal, and optic nerve changes in rheumatoid arthritis and primary sjogren's syndrome patients: comparıson with each other and healthy subjects. Int Ophthalmol 2024; 44:24. [PMID: 38324105 DOI: 10.1007/s10792-024-02970-9] [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: 05/09/2023] [Accepted: 12/04/2023] [Indexed: 02/08/2024]
Abstract
PURPOSE The present study aims to evaluate the optic nerve, macula, and choroidal changes in both rheumatoid arthritis (RA) and primary Sjögren's syndrome (SjS) patients, and to compare these findings with age-matched healthy volunteers. METHODS This study included 46 RA patients, 33 primary SjS patients, and 37 age-matched healthy volunteers. All of the patients underwent a thorough ophthalmological examination, during which measurements of the retinal nerve fiber layer (RNFL), ganglion cell layer(GCL), and subfoveal choroidal thickness (CT) were taken using OCT (optical coherence tomography). The measurements taken from the right eye of each patient were used to compare among the groups. RESULTS RNFL thickness in superior quadrant was found to be statistically significantly thinner in the eyes with RA when compared to the control group (p = 0.022). In the nasal quadrant, the RNFL thickness was significantly thinner in patients with primary SjS compared to healthy individuals (p = 0.036). Also, the temporal quadrant RNFL was significantly thinner in RA patients than in the primary SjS patients (p = 0.033). GCL thickness was observed to be thinner in all quadrants of both RA and primary SjS groups compared to the control group. However, the difference was not found to be statistically significant. Subfoveal CT was observed to be thicker in RA and SjS groups compared to the control group, but this difference was also not statistically significant. CONCLUSION Systemic autoimmune diseases like RA and primary SjS can lead to a decrease in RNLF and GCL thickness, which can impair visual acuity even in the absence of ocular symptoms. Therefore, monitoring changes in the optic nerve, retina, and choroid layer are crucial in these patients.
Collapse
Affiliation(s)
- Pelin Kiyat
- Department of Ophthalmology, İzmir Democracy University, Buca Seyfi Demirsoy Training and Research Hospital, Kozağaç Mah., Özmen Sok., No:147, Buca, İzmir, Turkey.
| | - Omer Karti
- Department of Ophthalmology, İzmir Democracy University, Buca Seyfi Demirsoy Training and Research Hospital, Kozağaç Mah., Özmen Sok., No:147, Buca, İzmir, Turkey
| | - Önay Gercik
- Department of Rheumatology, İzmir Democracy University, Buca Seyfi Demirsoy Training and Research Hospital, İzmir, Turkey
| | - Tuncer Şak
- Department of Rheumatology, İzmir Democracy University, Buca Seyfi Demirsoy Training and Research Hospital, İzmir, Turkey
| |
Collapse
|
6
|
Pittock SJ, Giometto B. Introduction and overview of immunopathological mechanisms and future directions. HANDBOOK OF CLINICAL NEUROLOGY 2024; 200:3-10. [PMID: 38494284 DOI: 10.1016/b978-0-12-823912-4.00029-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Paraneoplastic neurological disorders represent a significant part of the field of autoimmune neurology. Most neural autoantibodies discovered to date are associated with underlying malignancy and in that context are considered paraneoplastic antibody biomarkers. These autoantibodies can be divided into two major categories: those that target intracellular proteins (not pathogenic) and those that target plasma membrane proteins (pathogenic). Disorders accompanied by the former are mediated primarily by neural peptide-specific cytotoxic T-cells, are commonly associated with cancer, and are poorly responsive to immunotherapy. Disorders accompanied by the latter represent antibody-mediated diseases and are generally more responsive to immunotherapy. Areas of significant unmet need in the context of paraneoplastic neurological disorders include novel therapeutic options, as FDA-approved therapies are lacking. This chapter provides a brief overview of immunopathological mechanisms and potential future therapeutic targets. Our contributing authors and their chapters are also introduced.
Collapse
Affiliation(s)
- Sean J Pittock
- Department of Neurology and Laboratory Medicine and Pathology and Center for MS and Autoimmune Neurology, Mayo Clinic College of Medicine, Rochester, MN, United States.
| | - Bruno Giometto
- Department of Neurology, Azienda Provinciale per I Servizi Sanitari (APSS) and University of Trento (Italy), Santa Chiara Hospital, Trento, Italy
| |
Collapse
|
7
|
Chen CC, Lin CH, Lin MC. Maternal autoimmune disease and risk of offspring autism spectrum disorder - a nationwide population-based cohort study. Front Psychiatry 2023; 14:1254453. [PMID: 38025447 PMCID: PMC10654781 DOI: 10.3389/fpsyt.2023.1254453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Autism spectrum disorder (ASD) is a group of neurodevelopmental disorders which cause long term social and behavior impairment, and its prevalence is on the rise. Studies about the association between maternal autoimmune diseases and offspring ASD have controversial results. The aim of this study was to investigate whether maternal autoimmune diseases increase the risk of ASD in offspring from a population-based perspective. Methods The data sources were Taiwan's National Health Insurance Research Database (NHIRD) and Taiwan's Maternal and Child Health Database (MCHD), which were integrated and used to identify newborns whose mothers were diagnosed with autoimmune disease. Newborns were matched by maternal age, neonatal gender, and date of birth with controls whose mothers were without autoimmune disease using a ratio of 1:4 between 2004 and 2019. Data on diagnoses of autoimmune disease and autism spectrum disorders were retrieved from NHIRD. Patients who had at least 3 outpatient visits or at least 1 admission with a diagnosis of autoimmune disease and autism spectrum disorders were defined as incidence cases. The risks of ASD in offspring were compared between mothers with or without autoimmune disorders. Results We identified 20,865 newborns whose mothers had been diagnosed with autoimmune disease before pregnancy and matched them at a ratio of 1:4 with a total of 83,460 newborn whose mothers were without autoimmune disease, by maternal age, neonatal gender, and date of birth. They were randomly selected as the control group. The cumulative incidence rates of autism spectrum disorders (ASD) were significantly higher among the offspring of mothers with autoimmune diseases. After adjusting for cofactors, the risk of ASD remained significantly higher in children whose mother had autoimmune diseases. Regarding to specific maternal autoimmune disease, Sjögren's syndrome and rheumatoid arthritis were both associated with elevated risks of ASD in offspring. Conclusion Mother with autoimmune disease might be associated with increasing the risk of autism spectrum disorder in offspring.
Collapse
Affiliation(s)
- Ching-Chu Chen
- Children’s Medical Center, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Ching-Heng Lin
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Ming-Chih Lin
- Children’s Medical Center, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
- Department of Food and Nutrition, Providence University, Taichung, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| |
Collapse
|
8
|
Sørensen NV, Nilsson AC, Orlovska-Waast S, Jeppesen R, Christensen RHB, Benros ME. Antineuronal Autoantibodies in the Cerebrospinal Fluid and Serum From 106 Patients With Recent-Onset Depression Compared With 106 Individually Matched Healthy Control Subjects. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2023; 3:1116-1121. [PMID: 37881586 PMCID: PMC10593866 DOI: 10.1016/j.bpsgos.2022.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 09/04/2022] [Accepted: 10/19/2022] [Indexed: 03/05/2023] Open
Abstract
No large studies have investigated the prevalence of cerebrospinal fluid antineuronal autoantibodies in isolated depression. In this case-control study comparing 106 patients with isolated depression (ICD-10 code F32) with 106 healthy control subjects, cerebrospinal fluid and serum samples were tested for 7 immunoglobulin G autoantibodies using commercial fixed cell-based assays. To explore validity of methods, positive samples were retested twice by cell-based assays and once by tissue-based assays (monkey cerebellum). The prevalence of any of the antineuronal autoantibodies in cerebrospinal fluid was 0.0% in both groups and the seroprevalence was 0.9% in both groups, based on consistent findings in cell-based assays. However, all samples were negative by the tissue-based assay. Evaluation of antineuronal autoantibodies in cerebrospinal fluid cannot be recommended routinely for patients with isolated depression of moderate severity. Future studies of isolated depression should consider much larger sample sizes and evaluation of antineuronal autoantibodies using modalities other than commercial kits.
Collapse
Affiliation(s)
- Nina Vindegaard Sørensen
- Copenhagen Research Centre for Mental Health, Copenhagen University Hospital, Hellerup, Denmark
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anna Christine Nilsson
- Department of Clinical Immunology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Sonja Orlovska-Waast
- Copenhagen Research Centre for Mental Health, Copenhagen University Hospital, Hellerup, Denmark
| | - Rose Jeppesen
- Copenhagen Research Centre for Mental Health, Copenhagen University Hospital, Hellerup, Denmark
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Michael Eriksen Benros
- Copenhagen Research Centre for Mental Health, Copenhagen University Hospital, Hellerup, Denmark
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
9
|
Shiwaku H, Katayama S, Gao M, Kondo K, Nakano Y, Motokawa Y, Toyoda S, Yoshida F, Hori H, Kubota T, Ishikawa K, Kunugi H, Ikegaya Y, Okazawa H, Takahashi H. Analyzing schizophrenia-related phenotypes in mice caused by autoantibodies against NRXN1α in schizophrenia. Brain Behav Immun 2023; 111:32-45. [PMID: 37004758 DOI: 10.1016/j.bbi.2023.03.028] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 03/24/2023] [Accepted: 03/28/2023] [Indexed: 04/04/2023] Open
Abstract
The molecular pathological mechanisms underlying schizophrenia remain unclear; however, genomic analysis has identified genes encoding important risk molecules. One such molecule is neurexin 1α (NRXN1α), a presynaptic cell adhesion molecule. In addition, novel autoantibodies that target the nervous system have been found in patients with encephalitis and neurological disorders. Some of these autoantibodies inhibit synaptic antigen molecules. Studies have examined the association between schizophrenia and autoimmunity; however, the pathological data remain unclear. Here, we identified a novel autoantibody against NRXN1α in patients with schizophrenia (n = 2.1%) in a Japanese cohort (n = 387). None of the healthy control participants (n = 362) were positive for anti-NRXN1α autoantibodies. Anti-NRXN1α autoantibodies isolated from patients with schizophrenia inhibited the molecular interaction between NRXN1α and Neuroligin 1 (NLGN1) and between NRXN1α and Neuroligin 2 (NLGN2). Additionally, these autoantibodies reduced the frequency of the miniature excitatory postsynaptic current in the frontal cortex of mice. Administration of anti-NRXN1α autoantibodies from patients with schizophrenia into the cerebrospinal fluid of mice reduced the number of spines/synapses in the frontal cortex and induced schizophrenia-related behaviors such as reduced cognition, impaired pre-pulse inhibition, and reduced social novelty preference. These changes were improved through the removal of anti-NRXN1α autoantibodies from the IgG fraction of patients with schizophrenia. These findings demonstrate that anti-NRXN1α autoantibodies transferred from patients with schizophrenia cause schizophrenia-related pathology in mice. Removal of anti-NRXN1α autoantibodies may be a therapeutic target for a subgroup of patients who are positive for these autoantibodies.
Collapse
Affiliation(s)
- Hiroki Shiwaku
- Department of Psychiatry and Behavioral Sciences, Tokyo Medical and Dental University Graduate School, Tokyo 113-8510, Japan.
| | - Shingo Katayama
- Department of Psychiatry and Behavioral Sciences, Tokyo Medical and Dental University Graduate School, Tokyo 113-8510, Japan
| | - Mengxuan Gao
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Kanoh Kondo
- Department of Neuropathology, Medical Research Institute and Center for Brain Integration Research, Tokyo Medical and Dental University, 1-5-45, Tokyo 113-8510, Japan
| | - Yuri Nakano
- Department of Psychiatry and Behavioral Sciences, Tokyo Medical and Dental University Graduate School, Tokyo 113-8510, Japan
| | - Yukiko Motokawa
- Department of Psychiatry and Behavioral Sciences, Tokyo Medical and Dental University Graduate School, Tokyo 113-8510, Japan
| | - Saori Toyoda
- Department of Psychiatry and Behavioral Sciences, Tokyo Medical and Dental University Graduate School, Tokyo 113-8510, Japan
| | - Fuyuko Yoshida
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, 4-1-1, Tokyo 187-8553, Japan
| | - Hiroaki Hori
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, 4-1-1, Tokyo 187-8553, Japan
| | - Tetsuo Kubota
- Department of Medical Technology, Tsukuba International University, Ibaraki 300-0051, Japan
| | - Kinya Ishikawa
- The Center for Personalized Medicine for Healthy Aging, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Hiroshi Kunugi
- Department of Psychiatry, Teikyo University School of Medicine, Tokyo 173-8605, Japan
| | - Yuji Ikegaya
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; Institute for AI and Beyond, The University of Tokyo, Tokyo 113-0033, Japan; Center for Information and Neural Networks, National Institute of Information and Communications Technology, Suita City, Osaka 565-0871, Japan
| | - Hitoshi Okazawa
- Department of Neuropathology, Medical Research Institute and Center for Brain Integration Research, Tokyo Medical and Dental University, 1-5-45, Tokyo 113-8510, Japan
| | - Hidehiko Takahashi
- Department of Psychiatry and Behavioral Sciences, Tokyo Medical and Dental University Graduate School, Tokyo 113-8510, Japan
| |
Collapse
|
10
|
Daguano Gastaldi V, Bh Wilke J, Weidinger CA, Walter C, Barnkothe N, Teegen B, Luessi F, Stöcker W, Lühder F, Begemann M, Zipp F, Nave KA, Ehrenreich H. Factors predisposing to humoral autoimmunity against brain-antigens in health and disease: Analysis of 49 autoantibodies in over 7000 subjects. Brain Behav Immun 2023; 108:135-147. [PMID: 36323361 DOI: 10.1016/j.bbi.2022.10.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/24/2022] [Accepted: 10/22/2022] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Circulating autoantibodies (AB) against brain-antigens, often deemed pathological, receive increasing attention. We assessed predispositions and seroprevalence/characteristics of 49 AB in > 7000 individuals. METHODS Exploratory cross-sectional cohort study, investigating deeply phenotyped neuropsychiatric patients and healthy individuals of GRAS Data Collection for presence/characteristics of 49 brain-directed serum-AB. Predispositions were evaluated through GWAS of NMDAR1-AB carriers, analyses of immune check-point genotypes, APOE4 status, neurotrauma. Chi-square, Fisher's exact tests and logistic regression analyses were used. RESULTS Study of N = 7025 subjects (55.8 % male; 41 ± 16 years) revealed N = 1133 (16.13 %) carriers of any AB against 49 defined brain-antigens. Overall, age dependence of seroprevalence (OR = 1.018/year; 95 % CI [1.015-1.022]) emerged, but no disease association, neither general nor with neuropsychiatric subgroups. Males had higher AB seroprevalence (OR = 1.303; 95 % CI [1.144-1.486]). Immunoglobulin class (N for IgM:462; IgA:487; IgG:477) and titers were similar. Abundant were NMDAR1-AB (7.7 %). Low seroprevalence (1.25 %-0.02 %) was seen for most AB (e.g., amphiphysin, KCNA2, ARHGAP26, GFAP, CASPR2, MOG, Homer-3, KCNA1, GLRA1b, GAD65). Non-detectable were others. GWAS of NMDAR1-AB carriers revealed three genome-wide significant SNPs, two intergenic, one in TENM3, previously autoimmune disease-associated. Targeted analysis of immune check-point genotypes (CTLA4, PD1, PD-L1) uncovered effects on humoral anti-brain autoimmunity (OR = 1.55; 95 % CI [1.058-2.271]) and disease likelihood (OR = 1.43; 95 % CI [1.032-1.985]). APOE4 carriers (∼19 %) had lower seropositivity (OR = 0.766; 95 % CI [0.625-0.933]). Neurotrauma predisposed to NMDAR1-AB seroprevalence (IgM: OR = 1.599; 95 % CI [1.022-2.468]). CONCLUSIONS Humoral autoimmunity against brain-antigens, frequent across health and disease, is predicted by age, gender, genetic predisposition, and brain injury. Seroprevalence, immunoglobulin class, or titers do not predict disease.
Collapse
Affiliation(s)
- Vinicius Daguano Gastaldi
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, Göttingen, Germany
| | - Justus Bh Wilke
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, Göttingen, Germany
| | - Cosima A Weidinger
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, Göttingen, Germany
| | - Carolin Walter
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, Göttingen, Germany
| | - Nadine Barnkothe
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, Göttingen, Germany
| | - Bianca Teegen
- Institute for Experimental Immunology, Affiliated to Euroimmun, Lübeck, Germany
| | - Felix Luessi
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine‑Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Winfried Stöcker
- Institute for Experimental Immunology, Affiliated to Euroimmun, Lübeck, Germany
| | - Fred Lühder
- Institute of Neuroimmunology and Multiple Sclerosis Research, University Medical Center, of the Georg August University, Göttingen, Germany
| | - Martin Begemann
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, Göttingen, Germany
| | - Frauke Zipp
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine‑Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, City Campus, Göttingen, Germany
| | - Hannelore Ehrenreich
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, Göttingen, Germany.
| |
Collapse
|
11
|
Sun W, Feng Y, Li H, He X, Lu Y, Shan Z, Teng W, Li J. The effects of maternal anti-alpha-enolase antibody expression on the brain development in offspring. Clin Exp Immunol 2022; 210:187-198. [PMID: 36149061 PMCID: PMC9750830 DOI: 10.1093/cei/uxac086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/25/2022] [Accepted: 09/21/2022] [Indexed: 01/25/2023] Open
Abstract
Anti-alpha-enolase autoantibodies have not only been found to play an important role in autoimmune diseases but also cause neurological damage in adults. In this study, a pregnant mouse model with high serum alpha-enolase (ENO1)-specific antibody (ENO1Ab) was established by immunization with ENO1 protein to explore the effects of maternal circulatory ENO1Ab on the brain development in offspring. The pups showed impaired learning and memory abilities with obviously thinner tight junctions in the brain tissue. IgG deposits colocalized with both ENO1 protein and complement 3 (C3), and the membrane attack complex was obviously detectable in the brain tissues of pups from dams with high serum ENO1Ab expression. Our findings suggest that highly expressed ENO1Ab in the maternal circulation can pass through the blood-placenta-barrier and the compromised blood-brain barrier into the brain tissues of offspring and may cause neurological development impairment mainly through complement-dependent cytotoxicity.
Collapse
Affiliation(s)
- Wei Sun
- Department of Endocrinology and Metabolism, Institute of Endocrinology, NHC Key Laboratory of Diagnosis and Treatment of Thyroid Diseases, The First Hospital of China Medical University, Shenyang110001, PR China
| | - Yan Feng
- Department of Endocrinology and Metabolism, Institute of Endocrinology, NHC Key Laboratory of Diagnosis and Treatment of Thyroid Diseases, The First Hospital of China Medical University, Shenyang110001, PR China
| | - Hui Li
- Department of Endocrinology and Metabolism, Institute of Endocrinology, NHC Key Laboratory of Diagnosis and Treatment of Thyroid Diseases, The First Hospital of China Medical University, Shenyang110001, PR China
| | - Xiaoqing He
- Department of Endocrinology and Metabolism, Institute of Endocrinology, NHC Key Laboratory of Diagnosis and Treatment of Thyroid Diseases, The First Hospital of China Medical University, Shenyang110001, PR China
| | - Yihan Lu
- Department of Endocrinology and Metabolism, Institute of Endocrinology, NHC Key Laboratory of Diagnosis and Treatment of Thyroid Diseases, The First Hospital of China Medical University, Shenyang110001, PR China
| | - Zhongyan Shan
- Department of Endocrinology and Metabolism, Institute of Endocrinology, NHC Key Laboratory of Diagnosis and Treatment of Thyroid Diseases, The First Hospital of China Medical University, Shenyang110001, PR China
| | - Weiping Teng
- Department of Endocrinology and Metabolism, Institute of Endocrinology, NHC Key Laboratory of Diagnosis and Treatment of Thyroid Diseases, The First Hospital of China Medical University, Shenyang110001, PR China
| | - Jing Li
- Department of Endocrinology and Metabolism, Institute of Endocrinology, NHC Key Laboratory of Diagnosis and Treatment of Thyroid Diseases, The First Hospital of China Medical University, Shenyang110001, PR China
| |
Collapse
|
12
|
Mueller C, Elben S, Day GS, Alves P, Hebert J, Tang-Wai DF, Holtmann O, Iorio R, Perani D, Titulaer MJ, Hansen N, Bartsch T, Johnen A, Illes Z, Borm L, Willison AG, Wiendl H, Meuth SG, Kovac S, Bölte J, Melzer N. Review and meta-analysis of neuropsychological findings in autoimmune limbic encephalitis with autoantibodies against LGI1, CASPR2, and GAD65 and their response to immunotherapy. Clin Neurol Neurosurg 2022; 224:107559. [PMID: 36549220 DOI: 10.1016/j.clineuro.2022.107559] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 12/06/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022]
Abstract
OBJECTIVES It is assumed that autoimmune limbic encephalitis (ALE) demonstrates distinct neuropsychological manifestations with differential responses to immunotherapy according to which associated autoantibody (AAB), if any, is identified. Towards investigating whether this is the case, this study aims to summarize respective findings from the primary literature on ALE with AABs binding to cell surface neural antigens and ALE with AABs against intracellular neural antigens. METHODS We chose ALE with AABs against leucine-rich, glioma inactivated protein 1 (LGI1) and contactin-associated protein-like 2 (CASPR2) as the most frequent cell surface membrane antigens, and ALE with AABs to Embryonic Lethal, Abnormal Vision, Like 1 (ELAVL) proteins (anti-Hu) and glutamic acid decarboxylase 65 (GAD65) as the most frequent intracellular neural antigens. The PubMed and Scopus databases were searched on March 1st, 2021 for neuropsychological test and -screening data from patients with ALE of these AAB-types. Findings were reviewed according to AAB-type and immunotherapy status and are presented in a review section and are further statistically evaluated and presented in a meta-analysis section in this publication. RESULTS Of the 1304 initial hits, 32 studies on ALE with AABs against LGI1, CASPR2, and GAD65 reporting cognitive screening data could be included in a review. In ALE with AABs against LGI1, CASPR2 and GAD65, memory deficits are the most frequently reported deficits. However, deficits in attention and executive functions including working memory, fluency, and psychological function have also been reported. This review shows that ALE patients with AABs against both LGI1 and CASPR2 show higher percentages of neuropsychological deficits compared to ALE patients with AABs against GAD65 before and after initiation of immunotherapy. However, the methodologies used in these studies were heterogenous, and longitudinal studies were not comparable. Moreover, 21 studies including ALE patients with AABs against LGI1 and GAD65 were also suitable for meta-analysis. No suitable study on ALE with AABs against ELAVL proteins could be identified. Meta-Analyses could be executed for cognitive screening data and only partially, due to the small number of studies. However, in statistical analysis no consistent effect of AAB or immunotherapy on performance in cognitive screening tests could be found. CONCLUSION Currently, there is no definite evidence supporting the notion that different AAB-types of ALE exhibit distinct neuropsychological manifestations and respond differently to immunotherapy. Overall, we could not identify evidence for any effect of immunotherapy on cognition in ALE. More systematic, in-depth and longitudinal neuropsychological assessments of patients with different AAB-types of ALE are required in the future to investigate these aspects.
Collapse
Affiliation(s)
- Christoph Mueller
- Department of Neurology with Institute of Translational Neurology, Westfälische Wilhelms-University of Münster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany.
| | - Saskia Elben
- Department of Neurology, Medical Faculty, Heinrich-Heine University of Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany.
| | - Gregory S Day
- Department of Neurology, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, United States.
| | - Pedro Alves
- Serviço de Neurologia, Departamento de Neurociências e Saúde Mental, Hospital de Santa Maria, CHULN, Lisboa, Portugal; Laboratório de Estudos de Linguagem, Centro de Estudos Egas Moniz, Faculdade de Medicina, Universidade de Lisboa, Portugal.
| | - Julien Hebert
- Department of Medicine, Division of Neurology, University of Toronto, Toronto, Canada.
| | - David F Tang-Wai
- Department of Medicine, Division of Neurology, University of Toronto, Toronto, Canada; Memory Clinic, Toronto Western Hospital (University Health Network), Toronto, Canada.
| | - Olga Holtmann
- Institute of Medical Psychology and Systems Neuroscience, Westfälische Wilhelms-University of Münster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany.
| | - Raffaele Iorio
- Neurology Unit, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy; Università Cattolica del Sacro Cuore, Rome, Italy.
| | - Daniela Perani
- Division of Neuroscience, University Vita-Salute San Raffaele, Via Olgettina, 58, 20132 Milano, MI, Italy.
| | - Maarten J Titulaer
- Department of Neurology, Neuropsychology and Immunology, Erasmus Medical Center, Doctor Molewaterplein 40, 3015 GD Rotterdam, the Netherlands.
| | - Niels Hansen
- Department of Psychiatry and Psychotherapy, University of Göttingen, Von-Siebold-Str. 5, 37075 Göttingen, Germany.
| | - Thorsten Bartsch
- Department of Neurology, University Medical Center Schleswig-Holstein, Rosalind-Franklin-Straße 10, 24105 Kiel, Germany.
| | - Andreas Johnen
- Department of Neurology with Institute of Translational Neurology, Westfälische Wilhelms-University of Münster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany.
| | - Zslot Illes
- Department of Neurology with Institute of Clinical Research, University of Southern Denmark, J. B. Winsløws Vej 4, 5000 Odense, Denmark.
| | - Leah Borm
- Institute of Psychology, Westfälische Wilhelms-University of Münster, Fliednerstraße 21, 48149 Münster, Germany.
| | - Alice G Willison
- Department of Neurology, Medical Faculty, Heinrich-Heine University of Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany.
| | - Heinz Wiendl
- Department of Neurology with Institute of Translational Neurology, Westfälische Wilhelms-University of Münster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany.
| | - Sven G Meuth
- Department of Neurology with Institute of Translational Neurology, Westfälische Wilhelms-University of Münster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany; Department of Neurology, Medical Faculty, Heinrich-Heine University of Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany.
| | - Stjepana Kovac
- Department of Neurology with Institute of Translational Neurology, Westfälische Wilhelms-University of Münster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany.
| | - Jens Bölte
- Institute of Psychology, Westfälische Wilhelms-University of Münster, Fliednerstraße 21, 48149 Münster, Germany.
| | - Nico Melzer
- Department of Neurology with Institute of Translational Neurology, Westfälische Wilhelms-University of Münster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany; Department of Neurology, Medical Faculty, Heinrich-Heine University of Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany.
| |
Collapse
|
13
|
Bigotte M, Gimenez M, Gavoille A, Deligiannopoulou A, El Hajj A, Croze S, Goumaidi A, Malleret G, Salin P, Giraudon P, Ruiz A, Marignier R. Ependyma: a new target for autoantibodies in neuromyelitis optica? Brain Commun 2022; 4:fcac307. [PMID: 36751497 PMCID: PMC9897195 DOI: 10.1093/braincomms/fcac307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 08/26/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022] Open
Abstract
Neuromyelitis optica (NMO) is an autoimmune demyelinating disease of the central nervous system characterized by the presence of autoantibodies (called NMO-IgG) targeting aquaporin-4. Aquaporin-4 is expressed at the perivascular foot processes of astrocytes, in the glia limitans, but also at the ependyma. Most studies have focused on studying the pathogenicity of NMO-IgG on astrocytes, and NMO is now considered an astrocytopathy. However, periependymal lesions are observed in NMO suggesting that ependymal cells could also be targeted by NMO-IgG. Ependymal cells regulate CSF-parenchyma molecular exchanges and CSF flow, and are a niche for sub-ventricular neural stem cells. Our aim was to examine the effect of antibodies from NMO patients on ependymal cells. We exposed two models, i.e. primary cultures of rat ependymal cells and explant cultures of rat lateral ventricular wall whole mounts, to purified IgG of NMO patients (NMO-IgG) for 24 hours. We then evaluated the treatment effect using immunolabelling, functional assays, ependymal flow analysis and bulk RNA sequencing. For each experiment, the effects were compared with those of purified IgG from a healthy donors and non-treated cells. We found that: (i) NMO-IgG induced aquaporin-4 agglomeration at the surface of ependymal cells and induced cell enlargement in comparison to controls. In parallel, it induced an increase in gap junction connexin-43 plaque size; (ii) NMO-IgG altered the orientation of ciliary basal bodies and functionally impaired cilia motility; (iii) NMO-IgG activated the proliferation of sub-ventricular neural stem cells; (iv) treatment with NMO-IgG up-regulated the expression of pro-inflammatory cytokines and chemokines in the transcriptomic analysis. Our study showed that NMO-IgG can trigger an early and specific reactive phenotype in ependymal cells, with functional alterations of intercellular communication and cilia, activation of sub-ventricular stem cell proliferation and the secretion of pro-inflammatory cytokines. These findings suggest a key role for ependymal cells in the early phase of NMO lesion formation.
Collapse
Affiliation(s)
- Maxime Bigotte
- FORGETTING Team—Lyon Neuroscience Research Center, INSERM U1028, CNRS UMR 5292, Claude Bernard Lyon 1 University, 69675 Bron, France
| | - Marie Gimenez
- FORGETTING Team—Lyon Neuroscience Research Center, INSERM U1028, CNRS UMR 5292, Claude Bernard Lyon 1 University, 69675 Bron, France
| | - Antoine Gavoille
- Service de neurologie, sclérose en plaques, pathologies de la myéline et neuroinflammation, Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, 69500 Bron, France,Service de Biostatistique-Bioinformatique, Hospices Civils de Lyon, 69495 Pierre-Bénitem, France
| | - Adamantia Deligiannopoulou
- FORGETTING Team—Lyon Neuroscience Research Center, INSERM U1028, CNRS UMR 5292, Claude Bernard Lyon 1 University, 69675 Bron, France
| | - Aseel El Hajj
- FORGETTING Team—Lyon Neuroscience Research Center, INSERM U1028, CNRS UMR 5292, Claude Bernard Lyon 1 University, 69675 Bron, France
| | - Severine Croze
- Profilexpert, Genomic and Microgenomic Service, Claude Bernard Lyon 1 University, SFR santé LYON-EST, UCBL-INSERM US 7-CNRS UMS 3453, 69008 Lyon, France
| | | | - Gael Malleret
- FORGETTING Team—Lyon Neuroscience Research Center, INSERM U1028, CNRS UMR 5292, Claude Bernard Lyon 1 University, 69675 Bron, France
| | - Paul Salin
- FORGETTING Team—Lyon Neuroscience Research Center, INSERM U1028, CNRS UMR 5292, Claude Bernard Lyon 1 University, 69675 Bron, France
| | - Pascale Giraudon
- FORGETTING Team—Lyon Neuroscience Research Center, INSERM U1028, CNRS UMR 5292, Claude Bernard Lyon 1 University, 69675 Bron, France
| | - Anne Ruiz
- FORGETTING Team—Lyon Neuroscience Research Center, INSERM U1028, CNRS UMR 5292, Claude Bernard Lyon 1 University, 69675 Bron, France
| | - Romain Marignier
- Correspondence to: Romain Marignier Centre de référence des maladies inflammatoires rares du cerveau et de la moelle Service de neurologie, sclérose en plaques pathologies de la myéline et neuro-inflammation Hôpital Neurologique Pierre Wertheimer 59 boulevard Pinel, 69677 Bron cedex, France E-mail:
| |
Collapse
|
14
|
Teller J, Jung C, Wilke JB, Schimmelpfennig SD, Hindermann M, Hinken L, Gabriel MM, Fegbeutel C, Schäfer A, Laser H, Lichtinghagen R, Worthmann H, Weissenborn K, Ehrenreich H. Autoantibodies against NMDAR subunit NR1 disappear from blood upon anesthesia. Brain Behav Immun Health 2022; 24:100494. [PMID: 35965838 PMCID: PMC9372600 DOI: 10.1016/j.bbih.2022.100494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/22/2022] [Accepted: 07/27/2022] [Indexed: 11/24/2022] Open
Abstract
Anesthetics penetrate the blood-brain-barrier (BBB) and - as confirmed preclinically – transiently disrupt it. An analogous consequence in humans has remained unproven. In mice, we previously reported that upon BBB dysfunction, the brain acts as ‘immunoprecipitator’ of autoantibodies against N-methyl-D-aspartate-receptor subunit-NR1 (NMDAR1-AB). We thus hypothesized that during human anesthesia, pre-existing NMDAR1-AB will specifically bind to brain. Screening of N = 270 subjects undergoing general anesthesia during cardiac surgery for serum NMDAR1-AB revealed N = 25 NMDAR1-AB seropositives. Only N = 14 remained positive post-surgery. No changes in albumin, thyroglobulin or CRP were associated with reduction of serum NMDAR1-AB. Thus, upon anesthesia, BBB opening likely occurs also in humans. Whether the blood brain barrier opens on general anesthesia in humans is unclear. Serum NMDAR1-AB titers drop upon anesthesia during cardiac surgery. Drop of serum NMDAR1-AB after anesthesia indicates ‘immunoprecipitation’ by brain. Immunoprecipitation needs brain access of NMDAR1-AB, indicating barrier opening. Neither hemodilution nor inflammation explain this loss of NMDAR1-AB from serum.
Collapse
|
15
|
Ramirez-Celis A, Croen LA, Yoshida CK, Alexeeff SE, Schauer J, Yolken RH, Ashwood P, Van de Water J. Maternal autoantibody profiles as biomarkers for ASD and ASD with co-occurring intellectual disability. Mol Psychiatry 2022; 27:3760-3767. [PMID: 35618885 PMCID: PMC9708563 DOI: 10.1038/s41380-022-01633-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/05/2022] [Accepted: 05/12/2022] [Indexed: 02/08/2023]
Abstract
Maternal autoantibody-related ASD (MAR ASD) is a subtype of autism in which pathogenic maternal autoantibodies (IgG) cross the placenta, access the developing brain, and cause neurodevelopmental alterations and behaviors associated with autism in the exposed offspring. We previously reported maternal IgG response to eight proteins (CRMP1, CRMP2, GDA LDHA, LDHB, NSE, STIP1, and YBOX) and that reactivity to nine specific combinations of these proteins (MAR ASD patterns) was predictive of ASD risk. The aim of the current study was to validate the previously identified MAR ASD patterns (CRMP1 + GDA, CRMP1 + CRMP2, NSE + STIP1, CRMP2 + STIP1, LDHA + YBOX, LDHB + YBOX, GDA + YBOX, STIP1 + YBOX, and CRMP1 + STIP1) and their accuracy in predicting ASD risk in a prospective cohort employing maternal samples collected prior to parturition. We used prenatal plasma from mothers of autistic children with or without co-occurring intellectual disability (ASD = 540), intellectual disability without autism (ID = 184) and general population controls (GP = 420) collected by the Early Markers for Autism (EMA) study. We found reactivity to one or more of the nine previously identified MAR ASD patterns in 10% of the ASD group compared with 4% of the ID group and 1% of the GP controls (ASD vs GP: Odds Ratio (OR) = 7.81, 95% Confidence Interval (CI) 3.32 to 22.43; ASD vs ID: OR = 2.77, 95% CI (1.19-7.47)) demonstrating that the MAR ASD patterns are strongly associated with the ASD group and could be used to assess ASD risk prior to symptom onset. The pattern most strongly associated with ASD was CRMP1 + CRMP2 and increased the odds for an ASD diagnosis 16-fold (3.32 to >999.99). In addition, we found that several of these specific MAR ASD patterns were strongly associated with ASD with intellectual disability (ASD + ID) and others associated with ASD without ID (ASD-no ID). Prenatal screening for these MAR patterns may lead to earlier identification of ASD and facilitate access to the appropriate early intervention services based on each child's needs.
Collapse
Affiliation(s)
- Alexandra Ramirez-Celis
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, One Shields Avenue, University of California, Davis, CA, 95616, USA
| | - Lisa A Croen
- Kaiser Permanente Division of Research, 2000 Broadway, Oakland, CA, 94612, USA
| | - Cathleen K Yoshida
- Kaiser Permanente Division of Research, 2000 Broadway, Oakland, CA, 94612, USA
| | - Stacey E Alexeeff
- Kaiser Permanente Division of Research, 2000 Broadway, Oakland, CA, 94612, USA
| | - Joseph Schauer
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, One Shields Avenue, University of California, Davis, CA, 95616, USA
| | - Robert H Yolken
- Department of Psychiatry and Behavioral Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Paul Ashwood
- UC Davis MIND Institute, 2825 50th St, Sacramento, CA, 95817, USA
- Department of Medical Microbiology and Immunology, One Shields Avenue, University of California, Davis, CA, 95616, USA
| | - Judy Van de Water
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, One Shields Avenue, University of California, Davis, CA, 95616, USA.
- UC Davis MIND Institute, 2825 50th St, Sacramento, CA, 95817, USA.
| |
Collapse
|
16
|
Jianing W, Jingyi X, Pingting Y. Neuropsychiatric lupus erythematosus: Focusing on autoantibodies. J Autoimmun 2022; 132:102892. [PMID: 36030137 DOI: 10.1016/j.jaut.2022.102892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 08/05/2022] [Indexed: 10/15/2022]
Abstract
Patients with systemic lupus erythematosus (SLE) frequently suffer from nervous system complications, termed neuropsychiatric lupus erythematosus (NPLE). NPLE accounts for the poor prognosis of SLE. Correct attribution of NP events to SLE is the primary principle in managing NPLE. The vascular injuries and neuroinflammation are the fundamental neuropathologic changes in NPLE. Specific autoantibody-mediated central nerve system (CNS) damages distinguish NPLE from other CNS disorders. Though the central antibodies in NPLE are generally thought to be raised from the periphery immune system, they may be produced in the meninges and choroid plexus. On this basis, abnormal activation of microglia and disease-associated microglia (DAM) should be the common mechanisms of NPLE and other CNS disturbances. Improved understanding of both characteristic and sharing features of NPLE might yield further options for managing this disease.
Collapse
Affiliation(s)
- Wang Jianing
- Department of Rheumatology and Immunology, The First Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Xu Jingyi
- Department of Rheumatology and Immunology, The First Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Yang Pingting
- Department of Rheumatology and Immunology, The First Hospital of China Medical University, Shenyang, 110001, People's Republic of China.
| |
Collapse
|
17
|
Shiwaku H, Katayama S, Kondo K, Nakano Y, Tanaka H, Yoshioka Y, Fujita K, Tamaki H, Takebayashi H, Terasaki O, Nagase Y, Nagase T, Kubota T, Ishikawa K, Okazawa H, Takahashi H. Autoantibodies against NCAM1 from patients with schizophrenia cause schizophrenia-related behavior and changes in synapses in mice. Cell Rep Med 2022; 3:100597. [PMID: 35492247 PMCID: PMC9043990 DOI: 10.1016/j.xcrm.2022.100597] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 01/31/2022] [Accepted: 03/14/2022] [Indexed: 12/12/2022]
Abstract
From genetic and etiological studies, autoimmune mechanisms underlying schizophrenia are suspected; however, the details remain unclear. In this study, we describe autoantibodies against neural cell adhesion molecule (NCAM1) in patients with schizophrenia (5.4%, cell-based assay; 6.7%, ELISA) in a Japanese cohort (n = 223). Anti-NCAM1 autoantibody disrupts both NCAM1-NCAM1 and NCAM1-glial cell line-derived neurotrophic factor (GDNF) interactions. Furthermore, the anti-NCAM1 antibody purified from patients with schizophrenia interrupts NCAM1-Fyn interaction and inhibits phosphorylation of FAK, MEK1, and ERK1 when introduced into the cerebrospinal fluid of mice and also reduces the number of spines and synapses in frontal cortex. In addition, it induces schizophrenia-related behavior in mice, including deficient pre-pulse inhibition and cognitive impairment. In conclusion, anti-NCAM1 autoantibodies in patients with schizophrenia cause schizophrenia-related behavior and changes in synapses in mice. These antibodies may be a potential therapeutic target and serve as a biomarker to distinguish a small but treatable subgroup in heterogeneous patients with schizophrenia. Some patients with schizophrenia are positive for anti-NCAM1 autoantibodies Anti-NCAM1 antibody from schizophrenia patients inhibits NCAM1-NCAM1 interactions Anti-NCAM1 antibody from schizophrenia patients reduces spines and synapses in mice Anti-NCAM1 antibody from patients induces schizophrenia-related behavior in mice
Collapse
Affiliation(s)
- Hiroki Shiwaku
- Department of Psychiatry and Behavioral Sciences, Tokyo Medical and Dental University Graduate School, Tokyo 113-8510, Japan.
| | - Shingo Katayama
- Department of Psychiatry and Behavioral Sciences, Tokyo Medical and Dental University Graduate School, Tokyo 113-8510, Japan
| | - Kanoh Kondo
- Department of Neuropathology, Medical Research Institute and Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Yuri Nakano
- Department of Psychiatry and Behavioral Sciences, Tokyo Medical and Dental University Graduate School, Tokyo 113-8510, Japan
| | - Hikari Tanaka
- Department of Neuropathology, Medical Research Institute and Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Yuki Yoshioka
- Department of Neuropathology, Medical Research Institute and Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Kyota Fujita
- Department of Neuropathology, Medical Research Institute and Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Haruna Tamaki
- Department of Neurology and Neurological Science, Tokyo Medical and Dental University Graduate School, Tokyo 113-8510, Japan
| | | | | | | | | | - Tetsuo Kubota
- Department of Medical Technology, Tsukuba International University, Ibaraki 300-0051, Japan
| | - Kinya Ishikawa
- The Center for Personalized Medicine for Healthy Aging, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Hitoshi Okazawa
- Department of Neuropathology, Medical Research Institute and Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Hidehiko Takahashi
- Department of Psychiatry and Behavioral Sciences, Tokyo Medical and Dental University Graduate School, Tokyo 113-8510, Japan.
| |
Collapse
|
18
|
Hampe CS, Mitoma H. A Breakdown of Immune Tolerance in the Cerebellum. Brain Sci 2022; 12:brainsci12030328. [PMID: 35326284 PMCID: PMC8946792 DOI: 10.3390/brainsci12030328] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 11/21/2022] Open
Abstract
Cerebellar dysfunction can be associated with ataxia, dysarthria, dysmetria, nystagmus and cognitive deficits. While cerebellar dysfunction can be caused by vascular, traumatic, metabolic, genetic, inflammatory, infectious, and neoplastic events, the cerebellum is also a frequent target of autoimmune attacks. The underlying cause for this vulnerability is unclear, but it may be a result of region-specific differences in blood–brain barrier permeability, the high concentration of neurons in the cerebellum and the presence of autoantigens on Purkinje cells. An autoimmune response targeting the cerebellum—or any structure in the CNS—is typically accompanied by an influx of peripheral immune cells to the brain. Under healthy conditions, the brain is protected from the periphery by the blood–brain barrier, blood–CSF barrier, and blood–leptomeningeal barrier. Entry of immune cells to the brain for immune surveillance occurs only at the blood-CSF barrier and is strictly controlled. A breakdown in the barrier permeability allows peripheral immune cells uncontrolled access to the CNS. Often—particularly in infectious diseases—the autoimmune response develops because of molecular mimicry between the trigger and a host protein. In this review, we discuss the immune surveillance of the CNS in health and disease and also discuss specific examples of autoimmunity affecting the cerebellum.
Collapse
Affiliation(s)
- Christiane S. Hampe
- Department of Medicine, University of Washington, Seattle, WA 98195, USA
- Correspondence: ; Tel.: +1-206-554-9181
| | - Hiroshi Mitoma
- Department of Medical Education, Tokyo Medical University, Tokyo 160-0023, Japan;
| |
Collapse
|
19
|
NMDAR1 autoantibodies amplify behavioral phenotypes of genetic white matter inflammation: a mild encephalitis model with neuropsychiatric relevance. Mol Psychiatry 2022; 27:4974-4983. [PMID: 34866134 PMCID: PMC9763107 DOI: 10.1038/s41380-021-01392-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 09/28/2021] [Accepted: 11/12/2021] [Indexed: 02/06/2023]
Abstract
Encephalitis has an estimated prevalence of ≤0.01%. Even with extensive diagnostic work-up, an infectious etiology is identified or suspected in <50% of cases, suggesting a role for etiologically unclear, noninfectious processes. Mild encephalitis runs frequently unnoticed, despite slight neuroinflammation detectable postmortem in many neuropsychiatric illnesses. A widely unexplored field in humans, though clearly documented in rodents, is genetic brain inflammation, particularly that associated with myelin abnormalities, inducing primary white matter encephalitis. We hypothesized that "autoimmune encephalitides" may result from any brain inflammation concurring with the presence of brain antigen-directed autoantibodies, e.g., against N-methyl-D-aspartate-receptor NR1 (NMDAR1-AB), which are not causal of, but may considerably shape the encephalitis phenotype. We therefore immunized young female Cnp-/- mice lacking the structural myelin protein 2'-3'-cyclic nucleotide 3'-phosphodiesterase (Cnp) with a "cocktail" of NMDAR1 peptides. Cnp-/- mice exhibit early low-grade inflammation of white matter tracts and blood-brain barrier disruption. Our novel mental-time-travel test disclosed that Cnp-/- mice are compromised in what-where-when orientation, but this episodic memory readout was not further deteriorated by NMDAR1-AB. In contrast, comparing wild-type and Cnp-/- mice without/with NMDAR1-AB regarding hippocampal learning/memory and motor balance/coordination revealed distinct stair patterns of behavioral pathology. To elucidate a potential contribution of oligodendroglial NMDAR downregulation to NMDAR1-AB effects, we generated conditional NR1 knockout mice. These mice displayed normal Morris water maze and mental-time-travel, but beam balance performance was similar to immunized Cnp-/-. Immunohistochemistry confirmed neuroinflammation/neurodegeneration in Cnp-/- mice, yet without add-on effect of NMDAR1-AB. To conclude, genetic brain inflammation may explain an encephalitic component underlying autoimmune conditions.
Collapse
|
20
|
McLellan J, Kim DHJ, Bruce M, Ramirez-Celis A, Van de Water J. Maternal Immune Dysregulation and Autism-Understanding the Role of Cytokines, Chemokines and Autoantibodies. Front Psychiatry 2022; 13:834910. [PMID: 35722542 PMCID: PMC9201050 DOI: 10.3389/fpsyt.2022.834910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 04/20/2022] [Indexed: 11/29/2022] Open
Abstract
Autism spectrum disorder (ASD) is acknowledged as a highly heterogeneous, behaviorally defined neurodevelopmental disorder with multiple etiologies. In addition to its high heritability, we have come to recognize a role for maternal immune system dysregulation as a prominent risk factor for the development of ASD in the child. Examples of these risk factors include altered cytokine/chemokine activity and the presence of autoantibodies in mothers that are reactive to proteins in the developing brain. In addition to large clinical studies, the development of pre-clinical models enables the ability to evaluate the cellular and molecular underpinnings of immune-related pathology. For example, the novel animal models of maternal autoantibody-related (MAR) ASD described herein will serve as a preclinical platform for the future testing of targeted therapeutics for one 'type' of ASD. Identification of the cellular targets will advance precision medicine efforts toward tailored therapeutics and prevention. This minireview highlights emerging evidence for the role of maternal immune dysregulation as a potential biomarker, as well as a pathologically relevant mechanism for the development of ASD in offspring. Further, we will discuss the current limitations of these models as well as potential avenues for future research.
Collapse
Affiliation(s)
- Janna McLellan
- Division of Rheumatology, Department of Internal Medicine, Allergy, and Clinical Immunology, University of California, Davis, Davis, CA, United States
| | - Danielle H J Kim
- Division of Rheumatology, Department of Internal Medicine, Allergy, and Clinical Immunology, University of California, Davis, Davis, CA, United States
| | - Matthew Bruce
- Division of Rheumatology, Department of Internal Medicine, Allergy, and Clinical Immunology, University of California, Davis, Davis, CA, United States
| | - Alexandra Ramirez-Celis
- Division of Rheumatology, Department of Internal Medicine, Allergy, and Clinical Immunology, University of California, Davis, Davis, CA, United States
| | - Judy Van de Water
- Division of Rheumatology, Department of Internal Medicine, Allergy, and Clinical Immunology, University of California, Davis, Davis, CA, United States.,MIND Institute, University of California, Davis, Davis, CA, United States
| |
Collapse
|
21
|
Steiner J, Vasilevska V. Inflammation und psychische Erkrankung. INFO NEUROLOGIE + PSYCHIATRIE 2021. [PMCID: PMC8601778 DOI: 10.1007/s15005-021-2121-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Affiliation(s)
- Johann Steiner
- Universitätsklinikum Magdeburg, Klinik für Psychiatrie und Psychotherapie & Labor für Translationale Psychiatrie, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Veronika Vasilevska
- Universitätsklinikum Magdeburg, Klinik für Psychiatrie und Psychotherapie & Labor für Translationale Psychiatrie, Leipziger Str. 44, 39120 Magdeburg, Germany
| |
Collapse
|
22
|
N-methyl-D-aspartate receptor antibody and the choroid plexus in schizophrenia patients with tardive dyskinesia. J Psychiatr Res 2021; 142:290-298. [PMID: 34411812 DOI: 10.1016/j.jpsychires.2021.08.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/02/2021] [Accepted: 08/09/2021] [Indexed: 11/21/2022]
Abstract
BACKGROUND Immune disturbance has been postulated to be one of the mechanisms underlying the pathogenesis of tardive dyskinesia (TD). Recently, the role of autoimmune abnormality in TD has been increasingly recognized. Autoantibodies against neuronal N-methyl-D-aspartate receptor (NMDAR) may be cross-reactive in the brain in neuropsychiatric disorders, and the choroid plexus (CP) is a crucial immune barrier in the central nervous system (CNS). We supposed that NMDAR antibodies might underlie the pathophysiological process of TD through the mediation of CP. METHODS Serum NMDAR antibody levels were assessed by enzyme-linked immunosorbent assay, CP and ventricle volumes were assessed by magnetic resonance imaging in schizophrenia patients with TD (n = 61), without TD (NTD, n = 61), and in healthy controls (n = 74). Psychopathology and TD severity were assessed by the Positive and Negative Syndrome Scale and Abnormal Involuntary Movement Scale (AIMS). RESULTS NMDAR antibody levels were significantly higher, CP volumes were larger in the TD group than in the NTD group (p = 0.022; p = 0.019, respectively). In the TD group, higher NMDAR antibody level was correlated with larger CP volume (β = 0.406, p = 0.002). An elevated NMDAR antibody level and enlarged CP volume were correlated with orofacial AIMS score (β = 0.331, p = 0.011; β = 0.459, p = 3.34 × 10-4, respectively). In a mediation model, the effect of NMDAR antibody level on the orofacial AIMS score was mediated by the CP volume (indirect effect: β = 0.08, 95% confidence interval = 0.002-0.225; direct effect: β = 0.14, p = 0.154). CONCLUSIONS Our findings highlight a potential NMDAR antibody-associated mechanism in orofacial TD, which may be mediated by increased CP volume.
Collapse
|
23
|
Campana M, Strauß J, Münz S, Oviedo-Salcedo T, Fernando P, Eichhorn P, Falkai P, Hasan A, Wagner E. Cerebrospinal Fluid Pathologies in Schizophrenia-Spectrum Disorder-A Retrospective Chart Review. Schizophr Bull 2021; 48:47-55. [PMID: 34480476 PMCID: PMC8781327 DOI: 10.1093/schbul/sbab105] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND The role of inflammatory processes in the etiology of schizophrenia is increasingly being investigated. A link between psychosis and inflammation measured with different biomarkers has been reported in the literature and needs to be further explored. To investigate the presence of inflammatory biomarkers in first-episode psychosis (FEP) we analyzed the largest available FEP cohort to date regarding routine CSF and blood diagnostics. METHODS We report a retrospective analysis of clinical data from all inpatients that were admitted to our tertiary care hospital with a ICD-10 diagnosis of F2x (schizophrenia-spectrum) between January 1, 2008 and August 1, 2018 and underwent a lumbar puncture. RESULTS A total of n = 314 FEP patients were included in our sample. 42.7% patients (134/314) showed cerebrospinal fluid (CSF) alterations. Oligoclonal bands in the CSF were present in 21.8% of patients (67/307) with 12.4% (27/217) of patients presenting OCBs type 2 or 3. 15.8% (49/310) of our cohort revealed signs of blood-brain-barrier (BBB) dysfunction with increased albumin ratios. Mean serum CRP levels were 2.4 mg/l (SD = 9.5). CRP elevation was present in 116/280 cases (41.4%). CONCLUSIONS This large retrospective analysis on FEP cohort greatly enriches the clinical data available on this population and contributes to the discussion around inflammation in psychosis. Of note, even though several inflammatory alterations were found both in CSF and in blood tests, we found no evidence for a significant relationship between peripheral inflammation and inflammatory CSF. Furthermore, no significant relationship between CSF alterations and peripheral inflammation measured with CRP could be established.
Collapse
Affiliation(s)
- Mattia Campana
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany,To whom correspondence should be addressed; Department of Psychiatry and, Psychotherapy, LMU Munich, Nussbaumstraße 7, D-80336 Munich, Germany; tel.: 49-89-4400-55505, fax: 49-89-4400-55530, e-mail:
| | - Johanna Strauß
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| | - Susanne Münz
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| | - Tatiana Oviedo-Salcedo
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| | - Piyumi Fernando
- Department of Psychiatry, Psychotherapy and Psychosomatics, University of Augsburg, Augsburg, Germany
| | - Peter Eichhorn
- Institute of Laboratory Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| | - Alkomiet Hasan
- Department of Psychiatry, Psychotherapy and Psychosomatics, University of Augsburg, Augsburg, Germany
| | - Elias Wagner
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
24
|
Taraschenko O, Fox HS, Eldridge E, Wang W, Dowd SW, Al-Saleem F, Kattala CD, Dessain SK, Dingledine R. Monoclonal Antibodies From Anti-NMDA Receptor Encephalitis Patient as a Tool to Study Autoimmune Seizures. Front Neurosci 2021; 15:710650. [PMID: 34512245 PMCID: PMC8427020 DOI: 10.3389/fnins.2021.710650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 07/20/2021] [Indexed: 01/27/2023] Open
Abstract
Anti-N-methyl-D-aspartate (NMDA) receptor encephalitis manifests with precipitous cognitive decline, abnormal movements, and severe seizures that can be challenging to control with conventional anti-seizure medications. We previously demonstrated that intracerebroventricular (i.c.v.) administration of cerebrospinal fluid from affected patients, or purified NMDA receptor antibodies from encephalitis patients to mice precipitated seizures, thereby confirming that antibodies are directly pathogenic for seizures. Although different repertoires of anti-NMDA receptor antibodies could contribute to the distinct clinical manifestations in encephalitis patients, the role of specific antibodies in the expression of seizure, motor, and cognitive phenotypes remains unclear. Using three different patient-derived monoclonal antibodies with distinct epitopes within the N-terminal domain (NTD) of the NMDA receptor, we characterized the seizure burden, motor activity and anxiety-related behavior in mice. We found that continuous administration of 5F5, 2G6 or 3C11 antibodies for 2 weeks precipitated seizures, as measured with continuous EEG using cortical screw electrodes. The seizure burden was comparable in all three antibody-treated groups. The seizures were accompanied by increased hippocampal C-C chemokine ligand 2 (CCL2) mRNA expression 3 days after antibody infusion had stopped. Antibodies did not affect the motor performance or anxiety scores in mice. These findings suggest that neuronal antibodies targeting different epitopes within the NMDA receptor may result in a similar seizure phenotype.
Collapse
Affiliation(s)
- Olga Taraschenko
- Department of Neurological Sciences, Division of Epilepsy, University of Nebraska Medical Center, Omaha, NE, United States
| | - Howard S. Fox
- Department of Neurological Sciences, Division of Epilepsy, University of Nebraska Medical Center, Omaha, NE, United States
| | - Ember Eldridge
- Department of Neurological Sciences, Division of Epilepsy, University of Nebraska Medical Center, Omaha, NE, United States
| | - Wenyi Wang
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - Samuel W. Dowd
- Department of Neurological Sciences, Division of Epilepsy, University of Nebraska Medical Center, Omaha, NE, United States
| | - Fetweh Al-Saleem
- Lankenau Institute for Medical Research, Wynnewood, PA, United States
| | | | - Scott K. Dessain
- Lankenau Institute for Medical Research, Wynnewood, PA, United States
| | - Raymond Dingledine
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
25
|
Wollmuth LP, Chan K, Groc L. The diverse and complex modes of action of anti-NMDA receptor autoantibodies. Neuropharmacology 2021; 194:108624. [PMID: 34081993 PMCID: PMC8693782 DOI: 10.1016/j.neuropharm.2021.108624] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/13/2021] [Accepted: 05/15/2021] [Indexed: 12/21/2022]
Abstract
NMDA receptors are ligand-gated ion channels that are found throughout the brain and are required for both brain development and many higher order functions. A variety of human patients with diverse clinical phenotypes have been identified that carry autoantibodies directed against NMDA receptor subunits. Here we focus on two general classes of autoantibodies, anti-GluN1 antibodies associated with anti-NMDA receptor encephalitis and anti-GluN2 antibodies associated with systemic lupus erythematosus (SLE). These two general classes of anti-NMDA receptor autoantibodies display a wide range of pathophysiological mechanisms from altering synaptic composition to gating of NMDARs. While we have made progress in understanding how these autoantibodies work at the molecular and cellular level, many unanswered questions remain including their long-term actions on brain function, the significance of clonal variations, and their effects on different NMDA receptor-expressing cell types in local circuits. This information will be needed to define fully the transition from anti-NMDA receptor autoantibodies to a clinical phenotype.
Collapse
Affiliation(s)
- Lonnie P Wollmuth
- Department of Neurobiology & Behavior, USA; Department of Biochemistry & Cell Biology, USA; Center for Nervous System Disorders. Stony Brook University, Stony Brook, NY, 11794-5230, USA.
| | - Kelvin Chan
- Graduate Program in Neuroscience, USA; Medical Scientist Training Program (MSTP), USA; Department of Neurobiology & Behavior, USA
| | - Laurent Groc
- Univ. de Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000, Bordeaux, France; CNRS, IINS UMR, 5297, Bordeaux, France
| |
Collapse
|
26
|
Zhao X, Yang P. Hydroxychloroquine alleviates the neurotoxicity induced by anti-ribosomal P antibodies. J Neuroimmunol 2021; 358:577648. [PMID: 34229206 DOI: 10.1016/j.jneuroim.2021.577648] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 05/27/2021] [Accepted: 06/27/2021] [Indexed: 12/11/2022]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by a wide spectrum of autoantibodies, among which anti-ribosomal P (anti-P) antibodies are considered to be closely related to the neuropsychiatric SLE (NPSLE). Hydroxychloroquine (HCQ) has been proven to be effective against a variety of autoimmune diseases and is an essential drug for the treatment of SLE. In this study, we investigated the effects of anti-ribosomal P (anti-P) antibodies on neural cells and determined whether hydroxychloroquine (HCQ) influenced the anti-P antibodies-induced changes. The results showed that the binding of anti-P antibodies with mouse neuroblastoma- 2a (N2a) cells and rat primary neurons resulted in elevated intracellular calcium levels, inducing decreased cell viability and cell apoptosis. These inhibitory effects were alleviated by HCQ in a concentration-dependent manner by reducing the intracellular calcium levels and modulating the expression of apoptotic proteins. In summary, our study demonstrates that anti-P antibodies induce neural cell damage. HCQ could ease the damage effects and may play a neuroprotective role in NPSLE.
Collapse
Affiliation(s)
- Xinnan Zhao
- Department of Rheumatology and Immunology, the First Affiliated Hospital of China Medical University, Nanjing North Street 155, Heping District, Shenyang, Liaoning, China
| | - Pingting Yang
- Department of Rheumatology and Immunology, the First Affiliated Hospital of China Medical University, Nanjing North Street 155, Heping District, Shenyang, Liaoning, China.
| |
Collapse
|
27
|
Nkiliza A, Joshi U, Evans JE, Ait-Ghezala G, Parks M, Crawford F, Mullan M, Abdullah L. Adaptive Immune Responses Associated with the Central Nervous System Pathology of Gulf War Illness. Neurosci Insights 2021; 16:26331055211018458. [PMID: 34104887 PMCID: PMC8155779 DOI: 10.1177/26331055211018458] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 04/26/2021] [Indexed: 11/17/2022] Open
Abstract
Gulf War Illness is a multisymptomatic condition which affects 30% of veterans
from the 1991 Gulf War. While there is evidence for a role of peripheral
cellular and humoral adaptive immune responses in Gulf War Illness, a potential
role of the adaptive immune system in the central nervous system pathology of
this condition remains unknown. Furthermore, many of the clinical features of
Gulf War Illness resembles those of autoimmune diseases, but the biological
processes are likely different as the etiology of Gulf War Illness is linked to
hazardous chemical exposures specific to the Gulf War theatre. This review
discusses Gulf War chemical–induced maladaptive immune responses and a potential
role of cellular and humoral immune responses that may be relevant to the
central nervous system symptoms and pathology of Gulf War Illness. The
discussion may stimulate investigations into adaptive immunity for developing
novel therapies for Gulf War Illness.
Collapse
|
28
|
Kim K, Wang X, Ragonnaud E, Bodogai M, Illouz T, DeLuca M, McDevitt RA, Gusev F, Okun E, Rogaev E, Biragyn A. Therapeutic B-cell depletion reverses progression of Alzheimer's disease. Nat Commun 2021; 12:2185. [PMID: 33846335 PMCID: PMC8042032 DOI: 10.1038/s41467-021-22479-4] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 03/12/2021] [Indexed: 01/16/2023] Open
Abstract
The function of B cells in Alzheimer's disease (AD) is not fully understood. While immunoglobulins that target amyloid beta (Aβ) may interfere with plaque formation and hence progression of the disease, B cells may contribute beyond merely producing immunoglobulins. Here we show that AD is associated with accumulation of activated B cells in circulation, and with infiltration of B cells into the brain parenchyma, resulting in immunoglobulin deposits around Aβ plaques. Using three different murine transgenic models, we provide counterintuitive evidence that the AD progression requires B cells. Despite expression of the AD-fostering transgenes, the loss of B cells alone is sufficient to reduce Aβ plaque burden and disease-associated microglia. It reverses behavioral and memory deficits and restores TGFβ+ microglia, respectively. Moreover, therapeutic depletion of B cells at the onset of the disease retards AD progression in mice, suggesting that targeting B cells may also benefit AD patients.
Collapse
Affiliation(s)
- Ki Kim
- Immunoregulation Section, Laboratory of Immunology and Molecular Biology, National Institute on Aging, Baltimore, MD, USA
| | - Xin Wang
- Immunoregulation Section, Laboratory of Immunology and Molecular Biology, National Institute on Aging, Baltimore, MD, USA
| | - Emeline Ragonnaud
- Immunoregulation Section, Laboratory of Immunology and Molecular Biology, National Institute on Aging, Baltimore, MD, USA
| | - Monica Bodogai
- Immunoregulation Section, Laboratory of Immunology and Molecular Biology, National Institute on Aging, Baltimore, MD, USA
| | - Tomer Illouz
- The Mina and Everard Goodman faculty of Life Sciences, Ramat Gan, Israel
- The Gonda Brain Research Center, Bar Ilan University, Ramat Gan, Israel
- The Paul Feder Laboratory on Alzheimer's disease research, Bar Ilan University, Ramat Gan, Israel
| | - Marisa DeLuca
- Immunoregulation Section, Laboratory of Immunology and Molecular Biology, National Institute on Aging, Baltimore, MD, USA
| | - Ross A McDevitt
- Mouse Phenotyping Unit, Comparative Medicine Section, National Institute on Aging, Baltimore, MD, USA
| | - Fedor Gusev
- Department of Genomics and Human Genetics, Institute of General Genetics, Russian Academy of Sciences, Moscow, Russia
| | - Eitan Okun
- The Mina and Everard Goodman faculty of Life Sciences, Ramat Gan, Israel
- The Gonda Brain Research Center, Bar Ilan University, Ramat Gan, Israel
- The Paul Feder Laboratory on Alzheimer's disease research, Bar Ilan University, Ramat Gan, Israel
| | - Evgeny Rogaev
- Department of Genomics and Human Genetics, Institute of General Genetics, Russian Academy of Sciences, Moscow, Russia
- Center for Genetics and Genetic Technologies, Faculty of Biology, Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
- Department of Psychiatry, University of Massachusetts Medical School, Worcester, MA, USA
- Sirius University of Science and Technology, Sochi, Russia
| | - Arya Biragyn
- Immunoregulation Section, Laboratory of Immunology and Molecular Biology, National Institute on Aging, Baltimore, MD, USA.
| |
Collapse
|
29
|
Saini L, Sondhi V. CNS autoimmunity in children: An unwanted wrinkle in a smooth narrative. Med J Armed Forces India 2021; 77:138-146. [PMID: 33867628 DOI: 10.1016/j.mjafi.2021.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 03/13/2021] [Indexed: 11/25/2022] Open
Abstract
The emerging paradigm of childhood autoimmune neurological disorders has exploded in recent times due to reliable diagnostic methods and their ease of availability, well-defined diagnostic criteria, and universal awareness about these disorders. The most important aspect of these disorders is a considerable recovery in response to early targeted immunotherapy. If left untreated and/or ill-treated, these can lead to mortality or lifelong morbidity. Autoantibodies can target any part of the central nervous system (CNS), ranging from superficial structures like myelin to deep intracellular ion channels like voltage-gated potassium channels, resulting in contrasting and at times overlapping symptomatology. Though neuroimaging characteristics and serological tests confirm these disorders' diagnosis, it is essential to suspect them clinically and start management before the reports are available for minimizing morbidity and mortality. In the pediatric age group, several metabolic conditions, like mitochondrial disorders and enzyme deficiencies like HMG-CoA-lyase deficiency, can develop neuroimaging patterns similar to those seen in childhood CNS autoimmune disorders and may also show a favorable response to steroids in acute phases. Hence, the clinician must suspect and work up the index patient appropriately. Here, we briefly discuss the pathophysiology, clinical clues, and potential therapeutic targets related to pediatric CNS autoimmune disorders.
Collapse
Affiliation(s)
- Lokesh Saini
- Assistant Professor (Pediatrics), Pediatric Neurology Unit, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Vishal Sondhi
- Associate Professor, Department of Pediatrics, Armed Forces Medical College, Pune, India
| |
Collapse
|
30
|
Host genetics influences the relationship between the gut microbiome and psychiatric disorders. Prog Neuropsychopharmacol Biol Psychiatry 2021; 106:110153. [PMID: 33130294 DOI: 10.1016/j.pnpbp.2020.110153] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 10/10/2020] [Accepted: 10/22/2020] [Indexed: 12/15/2022]
Abstract
The gut microbiome is associated with psychiatric disorders; however, the molecular mechanisms mediating this association are poorly understood. The ability of host genetics to modulate the gut microbiome may be an important factor in understanding the association. In this study, we aimed to evaluate the role of genetic variants associated with the gut microbiome in the susceptibility of individuals to four psychiatric disorders: schizophrenia (SCZ), attention-deficit/hyperactivity disorder (ADHD), autism spectrum disorder (ASD), and major depressive disorder (MDD). A total of 201 host genetic markers associated with microbiome outcomes and reported in available genome-wide association studies (GWAS) were included in the analyses. We searched for these variants in the summary statistics of the largest GWAS on these disorders to date, which were published by the Psychiatric Genomic Consortium, and performed gene-based and gene set association analyses. Two variants were significantly associated with ASD (rs9401458 and rs9401452) and one with MDD (rs75036654). For the gene-based association analysis, eight genes were associated with SCZ (ASIC2, KCND3, ITSN1, SIPA1L3, RBMS3, BANK1, CSMD1, and LHFPL3), one with MDD (ACTL8), two with ADHD (C14orf39 and FBXL17), and one with ASD (PINX). The gene set comprising 83 genes was associated with SCZ (p = 0.047). These findings suggest that genes related to microbiome composition may affect the susceptibility of individuals to psychiatric disorders, mainly schizophrenia. Although less robust, the associations with ASD, ADHD, and MDD cannot be discarded.
Collapse
|
31
|
Beutgen VM, Pfeiffer N, Grus FH. Serological Levels of Anti-clathrin Antibodies Are Decreased in Patients With Pseudoexfoliation Glaucoma. Front Immunol 2021; 12:616421. [PMID: 33679756 PMCID: PMC7933590 DOI: 10.3389/fimmu.2021.616421] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 02/01/2021] [Indexed: 11/13/2022] Open
Abstract
Evidence for immunologic contribution to glaucoma pathophysiology is steadily increasing in ophthalmic research. Particularly, an altered abundance of circulating autoantibodies to ocular antigens is frequently observed. Here, we report an analysis of autoantibody abundancies to selected antigens in sera of open-angle glaucoma patients, subdivided into normal-tension glaucoma (N = 31), primary open-angle glaucoma (N = 43) and pseudoexfoliation glaucoma (N = 45), vs. a non-glaucomatous control group (N = 46). Serum samples were analyzed by protein microarray, including 38 antigens. Differences in antibody levels were assessed by ANOVA. Five serological antibodies showed significantly altered levels among the four groups (P < 0.05), which can be used to cluster the subjects in groups consisting mainly of PEXG or POAG/NTG samples. Among the altered autoantibodies, anti-Clathrin antibodies were identified as most important subgroup predictors, enhancing prospective glaucoma subtype prediction. As a second aim, we wanted to gain further insights into the characteristics of previously identified glaucoma-related antigens and their role in glaucoma pathogenesis. To this end, we used the bioinformatics toolset of Metascape to construct protein-protein interaction networks and GO enrichment analysis. Glaucoma-related antigens were significantly enriched in 13 biological processes, including mRNA metabolism, protein folding, blood coagulation and apoptosis, proposing a link of glaucoma-associated pathways to changes in the autoantibody repertoire. In conclusion, our study provides new aspects of the involvement of natural autoimmunity in glaucoma pathomechanisms and promotes advanced opportunities toward new diagnostic approaches.
Collapse
Affiliation(s)
- Vanessa M Beutgen
- Experimental and Translational Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Norbert Pfeiffer
- Experimental and Translational Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Franz H Grus
- Experimental and Translational Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| |
Collapse
|
32
|
Autoantibodies against NMDA receptor 1 modify rather than cause encephalitis. Mol Psychiatry 2021; 26:7746-7759. [PMID: 34331009 PMCID: PMC8872987 DOI: 10.1038/s41380-021-01238-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/06/2021] [Accepted: 07/09/2021] [Indexed: 02/07/2023]
Abstract
The etiology and pathogenesis of "anti-N-methyl-D-aspartate-receptor (NMDAR) encephalitis" and the role of autoantibodies (AB) in this condition are still obscure. While NMDAR1-AB exert NMDAR-antagonistic properties by receptor internalization, no firm evidence exists to date that NMDAR1-AB by themselves induce brain inflammation/encephalitis. NMDAR1-AB of all immunoglobulin classes are highly frequent across mammals with multiple possible inducers and boosters. We hypothesized that "NMDAR encephalitis" results from any primary brain inflammation coinciding with the presence of NMDAR1-AB, which may shape the encephalitis phenotype. Thus, we tested whether following immunization with a "cocktail" of 4 NMDAR1 peptides, induction of a spatially and temporally defined sterile encephalitis by diphtheria toxin-mediated ablation of pyramidal neurons ("DTA" mice) would modify/aggravate the ensuing phenotype. In addition, we tried to replicate a recent report claiming that immunizing just against the NMDAR1-N368/G369 region induced brain inflammation. Mice after DTA induction revealed a syndrome comprising hyperactivity, hippocampal learning/memory deficits, prefrontal cortical network dysfunction, lasting blood brain-barrier impairment, brain inflammation, mainly in hippocampal and cortical regions with pyramidal neuronal death, microgliosis, astrogliosis, modest immune cell infiltration, regional atrophy, and relative increases in parvalbumin-positive interneurons. The presence of NMDAR1-AB enhanced the hyperactivity (psychosis-like) phenotype, whereas all other readouts were identical to control-immunized DTA mice. Non-DTA mice with or without NMDAR1-AB were free of any encephalitic signs. Replication of the reported NMDAR1-N368/G369-immunizing protocol in two large independent cohorts of wild-type mice completely failed. To conclude, while NMDAR1-AB can contribute to the behavioral phenotype of an underlying encephalitis, induction of an encephalitis by NMDAR1-AB themselves remains to be proven.
Collapse
|
33
|
Vasilevska V, Guest PC, Schlaaff K, Incesoy EI, Prüss H, Steiner J. Potential Cross-Links of Inflammation With Schizophreniform and Affective Symptoms: A Review and Outlook on Autoimmune Encephalitis and COVID-19. Front Psychiatry 2021; 12:729868. [PMID: 34650454 PMCID: PMC8507462 DOI: 10.3389/fpsyt.2021.729868] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 08/23/2021] [Indexed: 01/08/2023] Open
Abstract
Based on current implications of the SARS-CoV-2 pandemic with regards to mental health, we show that biological links exist between inflammation and mental illness in addition to psychoreactive effects. We describe key principles of the biological interaction of the immune system and the mind, as well as the possible routes of viral entry into the brain. In addition, we provide a stepwise scheme for the diagnosis and therapy of autoimmune-encephalitis with schizophrenia-like symptomatology as a general guide for clinical practice and in the specialized scenario of infections, such as those caused by the SARS-CoV-2 virus.
Collapse
Affiliation(s)
- Veronika Vasilevska
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany.,Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Paul C Guest
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, University of Campinas, Campinas, Brazil
| | - Konstantin Schlaaff
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany.,Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Enise I Incesoy
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany.,Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany.,German Center for Neurodegenerative Diseases, Magdeburg, Germany.,Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke-University, Magdeburg, Germany
| | - Harald Prüss
- German Center for Neurodegenerative Diseases, Berlin, Germany.,Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Johann Steiner
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany.,Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany.,Center for Behavioral Brain Sciences, Magdeburg, Germany.,German Center for Mental Health, Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health, Magdeburg, Germany
| |
Collapse
|
34
|
Ehrenreich H, Wilke J, Steixner-Kumar AA. Spontaneous serum autoantibody fluctuations: To be or not to be. Mol Psychiatry 2021; 26:1723-1725. [PMID: 32968237 PMCID: PMC8440174 DOI: 10.1038/s41380-020-00883-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/20/2020] [Accepted: 09/04/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Hannelore Ehrenreich
- Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany.
| | - Justus Wilke
- grid.419522.90000 0001 0668 6902Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Agnes A. Steixner-Kumar
- grid.419522.90000 0001 0668 6902Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| |
Collapse
|
35
|
Fouad K, Popovich PG, Kopp MA, Schwab JM. The neuroanatomical-functional paradox in spinal cord injury. Nat Rev Neurol 2021; 17:53-62. [PMID: 33311711 PMCID: PMC9012488 DOI: 10.1038/s41582-020-00436-x] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2020] [Indexed: 12/13/2022]
Abstract
Although lesion size is widely considered to be the most reliable predictor of outcome after CNS injury, lesions of comparable size can produce vastly different magnitudes of functional impairment and subsequent recovery. This neuroanatomical-functional paradox is likely to contribute to the many failed attempts to independently replicate findings from animal models of neurotrauma. In humans, the analogous clinical-radiological paradox could explain why individuals with similar injuries can respond differently to rehabilitation. We describe the neuroanatomical-functional paradox in the context of traumatic spinal cord injury (SCI) and discuss the underlying mechanisms of the paradox, including the concepts of lesion-affected and recovery-related networks. We also consider the various secondary complications that further limit the accuracy of outcome prediction in SCI and provide suggestions for how to increase the predictive, translational value of preclinical SCI models.
Collapse
Affiliation(s)
- Karim Fouad
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, AB, Canada
- Institute for Neuroscience and Mental Health, University of Alberta, Edmonton, AB, Canada
| | - Phillip G Popovich
- Belford Center for Spinal Cord Injury, The Ohio State University, Wexner Medical Center, Columbus, OH, USA
- Center for Brain and Spinal Cord Repair, The Ohio State University, Wexner Medical Center, Columbus, OH, USA
- Department of Neuroscience, The Ohio State University, Wexner Medical Center, Columbus, OH, USA
- The Neurological Institute, The Ohio State University, Wexner Medical Center, Columbus, OH, USA
| | - Marcel A Kopp
- Clinical & Experimental Spinal Cord Injury Research, Department of Neurology with Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health (QUEST-Center for Transforming Biomedical Research), Berlin, Germany
| | - Jan M Schwab
- Belford Center for Spinal Cord Injury, The Ohio State University, Wexner Medical Center, Columbus, OH, USA.
- Center for Brain and Spinal Cord Repair, The Ohio State University, Wexner Medical Center, Columbus, OH, USA.
- Department of Neuroscience, The Ohio State University, Wexner Medical Center, Columbus, OH, USA.
- The Neurological Institute, The Ohio State University, Wexner Medical Center, Columbus, OH, USA.
- Clinical & Experimental Spinal Cord Injury Research, Department of Neurology with Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.
- Spinal Cord Injury Medicine (Neuroplegiology), Department of Neurology, The Ohio State University, Wexner Medical Center, Columbus, OH, USA.
- Department of Physical Medicine and Rehabilitation, The Ohio State University, Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
36
|
Pan H, Steixner-Kumar AA, Seelbach A, Deutsch N, Ronnenberg A, Tapken D, von Ahsen N, Mitjans M, Worthmann H, Trippe R, Klein-Schmidt C, Schopf N, Rentzsch K, Begemann M, Wienands J, Stöcker W, Weissenborn K, Hollmann M, Nave KA, Lühder F, Ehrenreich H. Multiple inducers and novel roles of autoantibodies against the obligatory NMDAR subunit NR1: a translational study from chronic life stress to brain injury. Mol Psychiatry 2021; 26:2471-2482. [PMID: 32089545 PMCID: PMC8440197 DOI: 10.1038/s41380-020-0672-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 01/13/2020] [Accepted: 01/23/2020] [Indexed: 12/03/2022]
Abstract
Circulating autoantibodies (AB) of different immunoglobulin classes (IgM, IgA, and IgG), directed against the obligatory N-methyl-D-aspartate-receptor subunit NR1 (NMDAR1-AB), belong to the mammalian autoimmune repertoire, and appear with age-dependently high seroprevalence across health and disease. Upon access to the brain, they can exert NMDAR-antagonistic/ketamine-like actions. Still unanswered key questions, addressed here, are conditions of NMDAR1-AB formation/boosting, intraindividual persistence/course in serum over time, and (patho)physiological significance of NMDAR1-AB in modulating neuropsychiatric phenotypes. We demonstrate in a translational fashion from mouse to human that (1) serum NMDAR1-AB fluctuate upon long-term observation, independent of blood-brain barrier (BBB) perturbation; (2) a standardized small brain lesion in juvenile mice leads to increased NMDAR1-AB seroprevalence (IgM + IgG), together with enhanced Ig-class diversity; (3) CTLA4 (immune-checkpoint) genotypes, previously found associated with autoimmune disease, predispose to serum NMDAR1-AB in humans; (4) finally, pursuing our prior findings of an early increase in NMDAR1-AB seroprevalence in human migrants, which implicated chronic life stress as inducer, we independently replicate these results with prospectively recruited refugee minors. Most importantly, we here provide the first experimental evidence in mice of chronic life stress promoting serum NMDAR1-AB (IgA). Strikingly, stress-induced depressive-like behavior in mice and depression/anxiety in humans are reduced in NMDAR1-AB carriers with compromised BBB where NMDAR1-AB can readily reach the brain. To conclude, NMDAR1-AB may have a role as endogenous NMDAR antagonists, formed or boosted under various circumstances, ranging from genetic predisposition to, e.g., tumors, infection, brain injury, and stress, altogether increasing over lifetime, and exerting a spectrum of possible effects, also including beneficial functions.
Collapse
Affiliation(s)
- Hong Pan
- grid.419522.90000 0001 0668 6902Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Agnes A. Steixner-Kumar
- grid.419522.90000 0001 0668 6902Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Anna Seelbach
- grid.419522.90000 0001 0668 6902Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Nadine Deutsch
- grid.10423.340000 0000 9529 9877Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Anja Ronnenberg
- grid.419522.90000 0001 0668 6902Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Daniel Tapken
- grid.5570.70000 0004 0490 981XDepartment of Biochemistry I–Receptor Biochemistry, Ruhr University, Bochum, Germany
| | - Nico von Ahsen
- grid.411984.10000 0001 0482 5331Institute of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Marina Mitjans
- grid.419522.90000 0001 0668 6902Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Hans Worthmann
- grid.10423.340000 0000 9529 9877Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Ralf Trippe
- grid.5570.70000 0004 0490 981XDepartment of Biochemistry I–Receptor Biochemistry, Ruhr University, Bochum, Germany
| | - Christina Klein-Schmidt
- grid.5570.70000 0004 0490 981XDepartment of Biochemistry I–Receptor Biochemistry, Ruhr University, Bochum, Germany
| | - Nadine Schopf
- grid.419522.90000 0001 0668 6902Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Kristin Rentzsch
- Institute for Experimental Immunology, Euroimmun, Lübeck, Germany
| | - Martin Begemann
- grid.419522.90000 0001 0668 6902Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany ,grid.411984.10000 0001 0482 5331Department of Psychiatry & Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Jürgen Wienands
- grid.7450.60000 0001 2364 4210Institute for Cellular and Molecular Immunology, Georg August University, Göttingen, Germany
| | - Winfried Stöcker
- Institute for Experimental Immunology, Euroimmun, Lübeck, Germany
| | - Karin Weissenborn
- grid.10423.340000 0000 9529 9877Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Michael Hollmann
- grid.5570.70000 0004 0490 981XDepartment of Biochemistry I–Receptor Biochemistry, Ruhr University, Bochum, Germany
| | - Klaus-Armin Nave
- grid.419522.90000 0001 0668 6902Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Fred Lühder
- grid.411984.10000 0001 0482 5331Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Göttingen, Germany
| | - Hannelore Ehrenreich
- Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany.
| |
Collapse
|
37
|
Ramirez-Celis A, Becker M, Nuño M, Schauer J, Aghaeepour N, Van de Water J. Risk assessment analysis for maternal autoantibody-related autism (MAR-ASD): a subtype of autism. Mol Psychiatry 2021; 26:1551-1560. [PMID: 33483694 PMCID: PMC8159732 DOI: 10.1038/s41380-020-00998-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 12/08/2020] [Accepted: 12/09/2020] [Indexed: 11/16/2022]
Abstract
The incidence of autism spectrum disorder (ASD) has been rising, however ASD-risk biomarkers remain lacking. We previously identified the presence of maternal autoantibodies to fetal brain proteins specific to ASD, now termed maternal autoantibody-related (MAR) ASD. The current study aimed to create and validate a serological assay to identify ASD-specific maternal autoantibody patterns of reactivity against eight previously identified proteins (CRMP1, CRMP2, GDA, NSE, LDHA, LDHB, STIP1, and YBOX) that are highly expressed in developing brain, and determine the relationship of these reactivity patterns with ASD outcome severity. We used plasma from mothers of children diagnosed with ASD (n = 450) and from typically developing children (TD, n = 342) to develop an ELISA test for each of the protein antigens. We then determined patterns of reactivity a highly significant association with ASD, and discovered several patterns that were ASD-specific (18% in the training set and 10% in the validation set vs. 0% TD). The three main patterns associated with MAR ASD are CRMP1 + GDA (ASD% = 4.2 vs. TD% = 0, OR 31.04, p = <0.0001), CRMP1 + CRMP2 (ASD% = 3.6 vs. TD% = 0, OR 26.08, p = 0.0005) and NSE + STIP1 (ASD% = 3.1 vs. TD% = 0, OR 22.82, p = 0.0001). Additionally, we found that maternal autoantibody reactivity to CRMP1 significantly increases the odds of a child having a higher Autism Diagnostic Observation Schedule (ADOS) severity score (OR 2.3; 95% CI: 1.358-3.987, p = 0.0021). This is the first report that uses machine learning subgroup discovery to identify with 100% accuracy MAR ASD-specific patterns as potential biomarkers of risk for a subset of up to 18% of ASD cases in this study population.
Collapse
Affiliation(s)
- Alexandra Ramirez-Celis
- grid.27860.3b0000 0004 1936 9684Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, One Shields Avenue, University of California, Davis, CA 95616 USA
| | - Martin Becker
- grid.168010.e0000000419368956Department of Anesthesiology, Pain, and Perioperative Medicine, Stanford University, Palo Alto, CA 94305 USA ,grid.168010.e0000000419368956Department of Pediatrics, Stanford University, Palo Alto, CA 94305 USA ,grid.168010.e0000000419368956Department of Biomedical Data Sciences, Stanford University, Palo Alto, CA 94305 USA
| | - Miriam Nuño
- grid.27860.3b0000 0004 1936 9684Department of Public Health Sciences, Division of Biostatistics, One Shields Avenue, University of California, Davis, CA 95616 USA
| | - Joseph Schauer
- grid.27860.3b0000 0004 1936 9684Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, One Shields Avenue, University of California, Davis, CA 95616 USA
| | - Nima Aghaeepour
- grid.168010.e0000000419368956Department of Anesthesiology, Pain, and Perioperative Medicine, Stanford University, Palo Alto, CA 94305 USA ,grid.168010.e0000000419368956Department of Pediatrics, Stanford University, Palo Alto, CA 94305 USA ,grid.168010.e0000000419368956Department of Biomedical Data Sciences, Stanford University, Palo Alto, CA 94305 USA
| | - Judy Van de Water
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, One Shields Avenue, University of California, Davis, CA, 95616, USA.
| |
Collapse
|
38
|
Oral spirochetes: Pathogenic mechanisms in periodontal disease. Microb Pathog 2020; 144:104193. [PMID: 32304795 DOI: 10.1016/j.micpath.2020.104193] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 04/07/2020] [Indexed: 12/14/2022]
Abstract
Periodontitis is an infectious inflammatory disease resulting from infection of biofilm forming bacteria. Several bacterial factors regulate inflammatory response and cause to tissue damage and loss of connection between gingival and tooth. Since bacterial virulence factors and also host immune responses have role, understanding of periodontal disease is complex, in overall we can say that in this disease epithelium is deleted by bacteria. Oral spirochetes are related to periodontitis, among them, Treponema denticola, have been associated with periodontal diseases such as early-onset periodontitis, necrotizing ulcerative gingivitis, and acute pericoronitis. This review will analyse mechanisms of pathogenesis of spirochetes in periodontitis. Microorganisms cause destruction of gingival tissue by two mechanisms. In one, damage results from the direct action of bacterial enzymes and cytotoxic products of bacterial metabolism. In the other, only bacterial components have role, and tissue destruction is the inevitable side effect of a subverted and exaggerated host inflammatory response to plaque antigens.
Collapse
|
39
|
Neuroprotective effect of microglia against impairments of auditory steady-state response induced by anti-P IgG from SLE patients in naïve mice. J Neuroinflammation 2020; 17:31. [PMID: 31973738 PMCID: PMC6977246 DOI: 10.1186/s12974-020-1716-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Accepted: 01/13/2020] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVE Autoantibodies against ribosomal P proteins (anti-P antibodies) are strongly associated with the neuropsychiatric manifestations of systemic lupus erythematosus (NPSLE). The present study was designed to assess whether anti-P antibodies can induce abnormal brain electrical activities in mice and investigate the potential cytopathological mechanism. METHODS Affinity-purified human anti-ribosomal P antibodies were injected intravenously into mice after blood-brain barrier (BBB) disruption. The auditory steady-state response (ASSR) was evaluated based on electroencephalography (EEG) signals in response to 40-Hz click-train stimuli, which were recorded from electrodes implanted in the skull of mice. Immunofluorescence staining was used to examine the morphology and density of neurons and glia in the hippocampus and cortex. The presence of apoptosis in the brain tissues was studied using the TUNEL assay. A PLX3397 diet was used to selectively eliminate microglia from the brains of mice. RESULTS Circulating anti-P antibodies caused an enhancement of the ASSR and the activation of microglia through the disrupted BBB, while no obvious neural apoptosis was observed. In contrast, when microglia were depleted, anti-P antibodies induced a serious reduction in the ASSR and neural apoptosis. CONCLUSION Our study indicates that anti-P antibodies can directly induce the dysfunction of auditory-evoked potentials in the brain and that microglia are involved in the protection of neural activity after the invasion of anti-P antibodies, which could have important implications for NPSLE.
Collapse
|
40
|
Benros ME, Mortensen PB. Role of Infection, Autoimmunity, Atopic Disorders, and the Immune System in Schizophrenia: Evidence from Epidemiological and Genetic Studies. Curr Top Behav Neurosci 2020; 44:141-159. [PMID: 30895532 DOI: 10.1007/7854_2019_93] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
An immunologic component to schizophrenia has been increasingly recognized, where infections and chronic inflammatory diseases as atopic disorders and autoimmune diseases could be involved in the pathogenesis of schizophrenia. Psychotic symptoms can be directly triggered by infections reaching the CNS, or be secondary to systemic inflammation indirectly affecting the brain through immune components, such as brain-reactive antibodies and cytokines. Large-scale epidemiological studies have consistently displayed that infections, autoimmune diseases, and atopic disorders are associated with increased risk of schizophrenia and that schizophrenia is associated with increased levels of immune markers at diagnosis. However, since there is also an increased risk of immune-related diseases after the diagnosis with schizophrenia and in family members of individuals with schizophrenia, parts of the association could also be due to heritable factors. Shared genetic factor might account for some of this increased prevalence of immune-related diseases among individuals with schizophrenia, and indeed the most pronounced genetic association with schizophrenia lies within the HLA region, which is one of the most important regions for the immune system. However, genetic studies have shown that the common genetic variants associated with schizophrenia do not seem to increase the susceptibility for acquiring infections. Nonetheless, shared genes with the susceptibility for acquiring infections not captured by the polygenic risk score for schizophrenia could still influence the association.
Collapse
Affiliation(s)
- Michael E Benros
- Mental Health Centre Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark.
- National Centre for Register Based Research, Aarhus University, Aarhus, Denmark.
| | - Preben B Mortensen
- National Centre for Register Based Research, Aarhus University, Aarhus, Denmark
| |
Collapse
|
41
|
Major depressive disorders accompanying autoimmune diseases - Response to treatment. Prog Neuropsychopharmacol Biol Psychiatry 2019; 95:109678. [PMID: 31238086 DOI: 10.1016/j.pnpbp.2019.109678] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/10/2019] [Accepted: 06/21/2019] [Indexed: 12/22/2022]
Abstract
MDDs (major depressive disorders) belong to the most frequently diagnosed mental diseases and affect approximately 350 million people all over the world. A growing body of evidence suggests that inflammatory processes may play a significant role in the pathophysiology and progression of the disease. The comorbidity of MDDs with many other medical conditions, for example autoimmune diseases (ADs) caused by inflammation, has been observed on numerous occasions. In both cases, increased levels of pro-inflammatory cytokines, chemokines and other inflammatory agents are observed. Furthermore, higher rates of inflammatory markers are associated with a poorer response to antidepressant treatment. Additionally, the presence of any AD is associated with higher prevalence of depression and may reduce the chance of effective therapy. Interestingly, the administration of several anti-inflammatory agents used in AD treatment is positively correlated with a reduction of depressive symptoms. In conclusion, the factors contributing to the coexistence of depression as well as affecting antidepressant treatment effectiveness may lead to an alteration of the cytokine profiles in many autoimmune diseases.
Collapse
|
42
|
Chiarugi A. A Popperian View on Anti‐CGRP Biologics in Migraine. Headache 2019; 59:1855-1860. [DOI: 10.1111/head.13695] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2019] [Indexed: 12/23/2022]
Affiliation(s)
- Alberto Chiarugi
- Headache Center Careggi University Hospital University of Florence Florence Italy
- Department of Health Sciences Section of Clinical Pharmacology and Oncology University of Florence Florence Italy
| |
Collapse
|
43
|
Joshi U, Pearson A, Evans JE, Langlois H, Saltiel N, Ojo J, Klimas N, Sullivan K, Keegan AP, Oberlin S, Darcey T, Cseresznye A, Raya B, Paris D, Hammock B, Vasylieva N, Hongsibsong S, Stern LJ, Crawford F, Mullan M, Abdullah L. A permethrin metabolite is associated with adaptive immune responses in Gulf War Illness. Brain Behav Immun 2019; 81:545-559. [PMID: 31325531 PMCID: PMC7155744 DOI: 10.1016/j.bbi.2019.07.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/17/2019] [Accepted: 07/11/2019] [Indexed: 10/31/2022] Open
Abstract
Gulf War Illness (GWI), affecting 30% of veterans from the 1991 Gulf War (GW), is a multi-symptom illness with features similar to those of patients with autoimmune diseases. The objective of the current work is to determine if exposure to GW-related pesticides, such as permethrin (PER), activates peripheral and central nervous system (CNS) adaptive immune responses. In the current study, we focused on a PER metabolite, 3-phenoxybenzoic acid (3-PBA), as this is a common metabolite previously shown to form adducts with endogenous proteins. We observed the presence of 3-PBA and 3-PBA modified lysine of protein peptides in the brain, blood and liver of pyridostigmine bromide (PB) and PER (PB+PER) exposed mice at acute and chronic post-exposure timepoints. We tested whether 3-PBA-haptenated albumin (3-PBA-albumin) can activate immune cells since it is known that chemically haptenated proteins can stimulate immune responses. We detected autoantibodies against 3-PBA-albumin in plasma from PB + PER exposed mice and veterans with GWI at chronic post-exposure timepoints. We also observed that in vitro treatment of blood with 3-PBA-albumin resulted in the activation of B- and T-helper lymphocytes and that these immune cells were also increased in blood of PB + PER exposed mice and veterans with GWI. These immune changes corresponded with elevated levels of infiltrating monocytes in the brain and blood of PB + PER exposed mice which coincided with alterations in the markers of blood-brain barrier disruption, brain macrophages and neuroinflammation. These studies suggest that pesticide exposure associated with GWI may have resulted in the activation of the peripheral and CNS adaptive immune responses, possibly contributing to an autoimmune-type phenotype in veterans with GWI.
Collapse
Affiliation(s)
- Utsav Joshi
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,Open University, Milton Keynes, UK,James A. Haley VA Hospital, Tampa, FL, USA
| | - Andrew Pearson
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,Open University, Milton Keynes, UK,James A. Haley VA Hospital, Tampa, FL, USA
| | - James E. Evans
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,James A. Haley VA Hospital, Tampa, FL, USA
| | - Heather Langlois
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,James A. Haley VA Hospital, Tampa, FL, USA
| | - Nicole Saltiel
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,James A. Haley VA Hospital, Tampa, FL, USA
| | - Joseph Ojo
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,Open University, Milton Keynes, UK,James A. Haley VA Hospital, Tampa, FL, USA
| | - Nancy Klimas
- NOVA Southeastern University, Ft. Lauderdale, FL, USA,Miami VAMC, Miami, FL, USA
| | | | | | - Sarah Oberlin
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,James A. Haley VA Hospital, Tampa, FL, USA
| | - Teresa Darcey
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,James A. Haley VA Hospital, Tampa, FL, USA
| | - Adam Cseresznye
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,James A. Haley VA Hospital, Tampa, FL, USA
| | - Balaram Raya
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,James A. Haley VA Hospital, Tampa, FL, USA
| | - Daniel Paris
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,Open University, Milton Keynes, UK,James A. Haley VA Hospital, Tampa, FL, USA
| | - Bruce Hammock
- Department of Entomology and Nematology, UCD Comprehensive Cancer Center, University of California Davis, Davis, CA, USA
| | - Natalia Vasylieva
- Department of Entomology and Nematology, UCD Comprehensive Cancer Center, University of California Davis, Davis, CA, USA
| | - Surat Hongsibsong
- Environment and Health Research Unit, Research Institute for Health Science, Chiang Mai University, Chiang, Thailand
| | - Lawrence J. Stern
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, USA,Department of Pathology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Fiona Crawford
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,Open University, Milton Keynes, UK,James A. Haley VA Hospital, Tampa, FL, USA
| | - Michael Mullan
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,Open University, Milton Keynes, UK,James A. Haley VA Hospital, Tampa, FL, USA
| | - Laila Abdullah
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA; Open University, Milton Keynes, UK; James A. Haley VA Hospital, Tampa, FL, USA.
| |
Collapse
|
44
|
Kello N, Anderson E, Diamond B. Cognitive Dysfunction in Systemic Lupus Erythematosus: A Case for Initiating Trials. Arthritis Rheumatol 2019; 71:1413-1425. [PMID: 31102496 PMCID: PMC6716992 DOI: 10.1002/art.40933] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 05/14/2019] [Indexed: 12/25/2022]
Abstract
Cognitive dysfunction (CD) is an insidious and underdiagnosed manifestation of systemic lupus erythematosus (SLE) that has a considerable impact on quality of life, which can be devastating. Given the inconsistencies in the modes of assessment and the difficulties in attribution to SLE, the reported prevalence of CD ranges from 5% to 80%. Although clinical studies of SLE-related CD have been hampered by heterogeneous subject populations and a lack of sensitive and standardized cognitive tests or other validated objective biomarkers for CD, there are, nonetheless, strong data from mouse models and from the clinical arena that show CD is related to known disease mechanisms. Several cytokines, inflammatory molecules, and antibodies have been associated with CD. Proposed mechanisms for antibody- and cytokine-mediated neuronal injury include the abrogation of blood-brain barrier integrity with direct access of soluble molecules in the circulation to the brain and ensuing neurotoxicity and microglial activation. No treatments for SLE-mediated CD exist, but potential candidates include agents that inhibit microglial activation, such as angiotensin-converting enzyme inhibitors, or that protect blood-brain barrier integrity, such as C5a receptor blockers. Structural and functional neuroimaging data have shown a range of regional abnormalities in metabolism and white matter microstructural integrity in SLE patients that correlate with CD and could in the future become diagnostic tools and outcome measures in clinical trials aimed at preserving cognitive function in SLE.
Collapse
Affiliation(s)
- Nina Kello
- Institute of Molecular Medicine, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, USA
| | - Erik Anderson
- Elmezzi Graduate School, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, USA
| | - Betty Diamond
- Institute of Molecular Medicine, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, USA
| |
Collapse
|
45
|
Konstantinou GN, Konstantinou GN. Psychiatric comorbidity in chronic urticaria patients: a systematic review and meta-analysis. Clin Transl Allergy 2019; 9:42. [PMID: 31462988 PMCID: PMC6706894 DOI: 10.1186/s13601-019-0278-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 07/29/2019] [Indexed: 01/06/2023] Open
Abstract
Background Dermatological illness can affect the quality of life and may coexist with psychiatric disorders. Objective The aim of this review was to systematically evaluate the published evidence of any psychiatric disorders that may coexist with chronic urticaria (CU) and any effect psychiatric interventions may have on CU. Methods Following the Cochrane guidance, we conducted a systematic literature search using web-based search engines provided by PubMed (for Medline database), Google Scholar and Scopus for studies that have investigated the existence of psychiatric comorbidity in patients with CU. To be included, a study had to possess features, such as: (1) distinction between chronic urticaria and allergic conditions, (2) direct collection of diagnostic psychiatric data by using clinical interview and standardized questionnaires, (3) International Classification of Disorders criteria or the Diagnostic and Statistical Manual of Mental Disorders criteria for the diagnosis of mental disorders, and (4) manuscripts written or published in the English language. Unpublished research and research in progress were not included. All the eligible studies were scrutinized for any reported psychiatric interventions that had any effect on CU. The systematic review has been registered on PROSPERO (registration number CRD42019122811) and was conducted following the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA). Results Twenty-five studies were identified. Almost one out of three CU patients have at least one underlying psychiatric disorder. None of the studies clarified whether the psychiatric disorders pre-existed the CU onset, and no association was found between CU severity and duration, and psychological functioning. Only one case report and two case series mentioned that treatment of psychiatric disorders with either anti-depressants, anti-anxiety drugs or psychological interventions might result in improvement of urticaria. Conclusions Patients with CU frequently experience psychiatric disorders. This highlights the need for a multidisciplinary therapeutic approach involving prompt recognition and management of any potential psychiatric disorder in addition to urticaria treatment. Further studies are needed to assess whether psychiatric disorders coexist with CU independently or follow urticaria onset and whether any psychological or psychiatric intervention may help in CU control.
Collapse
Affiliation(s)
| | - George N Konstantinou
- Department of Allergy and Clinical Immunology, 424 General Military Training Hospital, 11 Eleftheriou Benizelou Street, Kalamaria, 55 133 Thessaloniki, Greece
| |
Collapse
|
46
|
Breithaupt L, Köhler-Forsberg O, Larsen JT, Benros ME, Thornton LM, Bulik CM, Petersen L. Association of Exposure to Infections in Childhood With Risk of Eating Disorders in Adolescent Girls. JAMA Psychiatry 2019; 76:800-809. [PMID: 31017632 PMCID: PMC6487907 DOI: 10.1001/jamapsychiatry.2019.0297] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
IMPORTANCE Infections are recognized as playing a critical role in the risk of psychiatric disorders and suicidal behavior; however, few studies have evaluated the risk of eating disorders. OBJECTIVE To evaluate the association of hospitalization for infections and treatment with anti-infective agents with the risk of an eating disorder diagnosis. DESIGN, SETTING, AND PARTICIPANTS A nationwide, population-based, prospective cohort study of 525 643 girls born from January 1, 1989, to December 31, 2006, and followed up until December 31, 2012, was conducted using individual-level data drawn from Danish longitudinal registers. Data were analyzed from January 15 to June 15, 2018, using survival analysis models and adjusted for age, calendar period, parental educational level, and parental history of psychiatric illness. EXPOSURES Hospital admission for infections and prescribed anti-infective agents for infections. MAIN OUTCOMES AND MEASURES The main outcome of interest was diagnosis of an eating disorder (anorexia nervosa, bulimia nervosa, or eating disorder not otherwise specified) in a hospital, outpatient clinic, or emergency department setting. Cox proportional hazards regression models were used to estimate hazard ratios (HRs) and accompanying 95% CIs. RESULTS The study population consisted of 525 643 adolescent girls: 2131 received a diagnosis of anorexia nervosa (median [range] age, 15.2 [8.6-21.3] years), 711 received a diagnosis of bulimia nervosa (median [range] age, 17.9 [13.4-22.7] years), and 1398 received a diagnosis of an eating disorder not otherwise specified (median [range] age, 15.6 [8.6-21.6] years). A total of 525 643 adolescent girls were followed up for 4 601 720.4 person-years until a mean age of 16.2 years (range, 10.5-22.7 years). Severe infections that required hospitalization were associated with an increased risk of a subsequent diagnosis of anorexia nervosa by 22% (HR, 1.22; 95% CI, 1.10-1.35), bulimia nervosa by 35% (HR, 1.35; 95% CI, 1.13-1.60), and eating disorder not otherwise specified by 39% (HR, 1.39; 95% CI, 1.23-1.57) compared with adolescent girls without hospitalizations for infections. Infections treated with anti-infective agents were associated with an increased risk of a subsequent diagnosis of anorexia nervosa by 23% (HR, 1.23; 95% CI, 1.10-1.37), bulimia nervosa by 63% (HR, 1.63; 95% CI, 1.32-2.02), and eating disorder not otherwise specified by 45% (HR, 1.45; 95% CI, 1.25-1.67) compared with adolescent girls without infections treated with anti-infective agents. CONCLUSIONS AND RELEVANCE The findings suggest that hospital-treated infections and less severe infections treated with anti-infective agents are associated with increased risk of subsequent anorexia nervosa, bulimia nervosa, and eating disorders not otherwise specified and that future studies should investigate whether these associations are causal and identify the exact mechanisms between infections and subsequent inflammatory processes with eating disorders.
Collapse
Affiliation(s)
- Lauren Breithaupt
- Department of Psychology, George Mason University, Fairfax, Virginia,Eating Disorders Clinical and Research Program, Massachusetts General Hospital, Boston,Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - Ole Köhler-Forsberg
- Psychosis Research Unit, Aarhus University Hospital–Psychiatry, Risskov, Denmark,The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Aarhus University, Aarhus, Denmark,Centre for Integrated Register-Based Research, Aarhus University, Aarhus, Denmark,Department of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Janne Tidselbak Larsen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Aarhus University, Aarhus, Denmark,National Centre for Register-Based Research, School of Business and Social Sciences, Aarhus University, Aarhus, Denmark
| | - Michael E. Benros
- National Centre for Register-Based Research, School of Business and Social Sciences, Aarhus University, Aarhus, Denmark,Mental Health Centre Copenhagen, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Laura Marie Thornton
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill
| | - Cynthia M. Bulik
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill,Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill,Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Liselotte Petersen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Aarhus University, Aarhus, Denmark,National Centre for Register-Based Research, School of Business and Social Sciences, Aarhus University, Aarhus, Denmark
| |
Collapse
|
47
|
Predictors of damage accrual in systemic lupus erythematosus: a longitudinal observational study with focus on neuropsychological factors and anti-neuronal antibodies. Clin Rheumatol 2019; 38:3129-3137. [PMID: 31367942 DOI: 10.1007/s10067-019-04707-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 06/27/2019] [Accepted: 07/22/2019] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Central nervous system disease occurs in over 20% of patients with systemic lupus erythematosus (SLE) resulting in major morbidity and damage. Cognitive dysfunction is common in SLE, but the cause remains uncertain and treatment options are limited. This study explores the influence of clinical, neuropsychological factors and anti-neuronal antibodies on lupus damage accrual. METHOD A prospective cohort with 99 SLE patients recruited between 2008 and 2013 and followed up in 2016 was established. Baseline evaluations were depression (MINI-Plus), cognitive function evaluating attention, visuospatial memory and executive functions, and anti-neuronal antibodies. Activity index (SLEDAI-2K) and SLICC/ACR Damage Index (SDI) were assessed at baseline and last follow-up. RESULTS At baseline, median (interquartile range) age was 36.0 years (27.0-45.0), disease duration 3.7 years (0.4-12.4), SLEDAI-2K 6.0 (3.0-12.0), and SDI score 1.0 (0-1.0). Major depression was present in 23%, cognitive deficit in 18%, and received immunomodulators in 36%. Anti-dsDNA/N-methyl-D-aspartate receptor antibodies were present in 19%, anti-ribosomal P in 12%, and anti-neuronal surface P antigen (NSPA) in 5%. After a median follow-up of 55 months (interquartile range 39-78), 11% had damage accrual. In a multivariate analysis, baseline SDI, SLEDAI-2K, and immunomodulators use were associated with final damage, whereas SLEDAI-2K and immunomodulator use were also associated with accrual damage. Models including anti-NSPA showed impact on final and accrual damage. Cognitive deficit, depression, and other autoantibodies were not predictors. CONCLUSIONS Disease activity and immunomodulator use associate with lupus damage. Of the anti-neuronal antibodies examined, anti-NSPA emerged as a potential poor prognostic factor, probably related to severe SLE onset requiring elevated corticosteroid doses. Key Points • Anti-NSPA may be a worse prognostic factor in SLE. • Other neuropsychological factors do not influence damage.
Collapse
|
48
|
Mané-Damas M, Hoffmann C, Zong S, Tan A, Molenaar PC, Losen M, Martinez-Martinez P. Autoimmunity in psychotic disorders. Where we stand, challenges and opportunities. Autoimmun Rev 2019; 18:102348. [PMID: 31323365 DOI: 10.1016/j.autrev.2019.102348] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 02/26/2019] [Indexed: 12/24/2022]
Abstract
Psychotic disorders are debilitating mental illnesses associated with abnormalities in various neurotransmitter systems. The development of disease-modifing therapies has been hampered by the mostly unknown etiologies and pathophysiologies. Autoantibodies against several neuronal antigens are responsible for autoimmune encephalitis. These autoantibodies disrupt neurotransmission within the brain, resulting in a wide range of psychiatric and neurologic manifestations, including psychosis. The overlap of symptoms of autoimmune encephalitis with psychotic disorders raised the question as to whether autoantibodies against a number of receptors, ion channel and associated proteins could ultimately be responsible for some forms of psychosis. Here we review our current knowledge, on antibody mediated autoimmunity in psychotic disorders, the different diagnostic methods and their limitations, as well as on varying therapeutic approaches targeting the immune system.
Collapse
Affiliation(s)
- Marina Mané-Damas
- Department of Psychiatry and Neuropsychology, School of Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.
| | - Carolin Hoffmann
- Department of Psychiatry and Neuropsychology, School of Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.
| | - Shenghua Zong
- Department of Psychiatry and Neuropsychology, School of Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.
| | - Amanda Tan
- Department of Psychiatry and Neuropsychology, School of Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Peter C Molenaar
- Department of Psychiatry and Neuropsychology, School of Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.
| | - Mario Losen
- Department of Psychiatry and Neuropsychology, School of Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.
| | - Pilar Martinez-Martinez
- Department of Psychiatry and Neuropsychology, School of Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
49
|
Gata-Garcia A, Diamond B. Maternal Antibody and ASD: Clinical Data and Animal Models. Front Immunol 2019; 10:1129. [PMID: 31191521 PMCID: PMC6547809 DOI: 10.3389/fimmu.2019.01129] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 05/03/2019] [Indexed: 12/26/2022] Open
Abstract
Over the past several decades there has been an increasing interest in the role of environmental factors in the etiology of neuropsychiatric and neurodevelopmental disorders. Epidemiologic studies have shifted from an exclusive focus on the identification of genetic risk alleles for such disorders to recognizing and understanding the contribution of xenobiotic exposures, infections, and the maternal immune system during the prenatal and early post-natal periods. In this review we discuss the growing literature regarding the effects of maternal brain-reactive antibodies on fetal brain development and their contribution to the development of neuropsychiatric and neurodevelopmental disorders. Autoimmune diseases primarily affect women and are more prevalent in mothers of children with neurodevelopmental disorders. For example, mothers of children with Autism Spectrum Disorder (ASD) are significantly more likely to have an autoimmune disease than women of neurotypically developing children. Moreover, they are four to five times more likely to harbor brain-reactive antibodies than unselected women of childbearing age. Many of these women exhibit no apparent clinical consequence of harboring these antibodies, presumably because the antibodies never access brain tissue. Nevertheless, these maternal brain-reactive antibodies can access the fetal brain, and some may be capable of altering brain development when present during pregnancy. Several animal models have provided evidence that in utero exposure to maternal brain-reactive antibodies can permanently alter brain anatomy and cause persistent behavioral or cognitive phenotypes. Although this evidence supports a contribution of maternal brain-reactive antibodies to neurodevelopmental disorders, an interplay between antibodies, genetics, and other environmental factors is likely to determine the specific neurodevelopmental phenotypes and their severity. Additional modulating factors likely also include the microbiome, sex chromosomes, and gonadal hormones. These interactions may help to explain the sex-bias observed in neurodevelopmental disorders. Studies on this topic provide a unique opportunity to learn how to identify and protect at risk pregnancies while also deciphering critical pathways in neurodevelopment.
Collapse
Affiliation(s)
- Adriana Gata-Garcia
- Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Manhasset, NY, United States.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | - Betty Diamond
- Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Manhasset, NY, United States
| |
Collapse
|
50
|
Abstract
Purpose of review Humoral autoimmunity has gained highest interest in neurology and psychiatry. Despite numerous recent articles on this hot topic, however, the biological significance of natural autoantibodies (AB) and the normal autoimmune repertoire of mammals remained quite obscure. AB may contribute to disorder-relevant phenotypes and are even believed to induce diseases themselves, but the circumstances under which AB become pathogenic are not fully understood. This review will focus on the highly frequent AB against the N-methyl-d-aspartate receptor 1 (NMDAR1-AB) as an illustrating example and provide a critical overview of current work (please note that the new nomenclature, GluN1, is disregarded here for consistency with the AB literature). In particular, it will demonstrate how little is known at this point and how many conclusions are drawn based on small numbers of individuals, fragmentary experimental approaches or missing controls. Recent findings NMDAR1-AB were investigated by clinicians world-wide with numerous small studies and case reports appearing yearly. Many publications were on ‘anti-NMDAR encephalitis’ cases or tried to separate those from other NMDAR1-AB associated conditions. Original exclusivity claims (e.g. electroencephalogram, EEG or functional magnetic resonance imaging, fMRI findings) turned out not to be exclusive for ‘anti-NMDAR encephalitis’. Systematic analyses of representative NMDAR1-AB positive sera of all immunoglobulin (Ig) classes showed comparable distribution of different epitopes, often polyspecific/polyclonal, across health and disease. Sophisticated imaging tools provided findings on synapse trafficking changes induced by NMDAR1-AB from psychotic subjects but still lack epitope data to support any claimed disorder link. Persistently high titers of NMDAR1-AB (IgG) in immunized mice with open blood–brain barrier (BBB)-induced psychosis-like symptoms but failed to induce inflammation in the brain. Knowledge on peripheral NMDAR, for example in the immune system, and on potential inducers of NMDAR1-AB is only slowly increasing. Summary The present knowledge on the (patho) physiological role of NMDAR1-AB is very limited and still characterized by adamant rumors. Much more experimental work and more solid and informative clinical reports, including large numbers of subjects and adequate control groups, follow-up investigations and interdisciplinary approaches will be necessary to obtain a better understanding of the significance of humoral autoimmunity in general (in focus here: NMDAR1-AB) and its disease-relevance in particular.
Collapse
|