51
|
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: 6.4] [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
|
52
|
Biological evaluation of newly synthesized quinoline–based compound PPQ-8 in acute and chronic toxoplasmosis: An experimental study. Exp Parasitol 2019; 206:107756. [DOI: 10.1016/j.exppara.2019.107756] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 07/31/2019] [Accepted: 09/03/2019] [Indexed: 01/01/2023]
|
53
|
Alexopoulos H, Dalakas MC. The immunobiology of autoimmune encephalitides. J Autoimmun 2019; 104:102339. [PMID: 31611142 DOI: 10.1016/j.jaut.2019.102339] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 09/25/2019] [Indexed: 12/17/2022]
Abstract
Autoimmune encephalitides, with an estimated incidence of 1.5 per million population per year, although described only 15 years ago, have already had a remarkable impact in neurology and paved the field to autoimmune neuropsychiatry. Many patients traditionally presented with aberrant behavior, especially of acute or subacute onset, and treated with anti-psychotic therapies, turn out to have a CNS autoimmune disease with pathogenic autoantibodies against synaptic antigens responding to immunotherapies. The review describes the clinical spectrum of these disorders, and the pathogenetic role of key autoantibodies directed against: a) cell surface synaptic antigens and receptors, including NMDAR, GABAa, GABAb, AMPA and glycine receptors; b) channels such as AQP4 water-permeable channel or voltage-gated potassium channels; c) proteins that stabilize voltage-gated potassium channel complex into the membrane, like the LGI1 and CASPR2; and d) enzymes that catalyze the formation of neurotransmitters such as Glutamic Acid Decarboxylase (GAD). These antibodies, effectively target excitatory or inhibitory synapses in the limbic system, basal ganglia or brainstem altering synaptic function and resulting in uncontrolled neurological excitability disorder clinically manifested with psychosis, agitation, behavioral alterations, depression, sleep disturbances, seizure-like phenomena, movement disorders such as ataxia, chorea and dystonia, memory changes or coma. Some of the identified triggering factors include: viruses, especially herpes simplex, accounting for the majority of relapses occurring after viral encephalitis, which respond to immunotherapy rather than antiviral agents; tumors especially teratoma, SCLC and thymomas; and biological cancer therapies (immune-check-point inhibitors). As anti-synaptic antibodies persist after viral infections or tumor removal, augmentation of autoreactive B cells which release autoantigens to draining lymph nodes, molecular mimicry and infection-induced bystander immune activation products play a role in autoimmunization process or perpetuating autoimmune neuroinflammation. The review stresses the importance of early detection, clinical recognition, proper antibody testing and early therapy initiation as these disorders, regardless of a known or not trigger, are potentially treatable responding to systemic immunotherapy with intravenous steroids, IVIg, rituximab or even bortezomid.
Collapse
Affiliation(s)
- Harry Alexopoulos
- Neuroimmunology Unit, Department of Pathophysiology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Marinos C Dalakas
- Neuroimmunology Unit, Department of Pathophysiology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Department of Neurology, Thomas Jefferson University, Philadelphia, USA.
| |
Collapse
|
54
|
Nissen J, Trabjerg B, Pedersen MG, Banasik K, Pedersen OB, Sørensen E, Nielsen KR, Erikstrup C, Petersen MS, Paarup HM, Bruun-Rasmussen P, Westergaard D, Hansen TF, Pedersen CB, Werge T, Torrey F, Hjalgrim H, Mortensen PB, Yolken R, Brunak S, Ullum H, Burgdorf KS. Herpes Simplex Virus Type 1 infection is associated with suicidal behavior and first registered psychiatric diagnosis in a healthy population. Psychoneuroendocrinology 2019; 108:150-154. [PMID: 31284079 DOI: 10.1016/j.psyneuen.2019.06.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 06/24/2019] [Accepted: 06/24/2019] [Indexed: 12/29/2022]
Abstract
Increasing evidence shows that latent infections and inflammation is associated with cognitive and behavioral changes in humans. This case-control study investigates the association between Herpes Simplex Virus Type 1 (HSV-1) infection and C-reactive Protein (CRP) levels, and psychiatric disorders and suicidal behavior. Public health register data from 81,912 participants in the Danish Blood Donor Study, were reviewed to identify individuals registered with an ICD-10 code of any psychiatric diagnosis, or who had attempted or committed suicide. We found 1,504 psychiatric cases and 353 suicidal cases; for all cases, controls were frequency-matched by age and sex, resulting in 5,336 participants. Plasma samples were analyzed for IgG-class antibodies against HSV-1 and CRP. HSV-1 infection was associated with suicidal behavior (odds-ratio, 1.40; 95% confidence interval [CI] 1.11-1.77). Accounting for temporality, HSV-1 infection was associated with having first psychiatric disorder after the date of blood collection (incidence rate ration, 1.44; 95% CI, 1.05-1.95). No association between CRP and psychiatric disorders or suicidal behavior was found. The finding that HSV-1 was associated with suicidal behavior and first psychiatric disorder indicates that infection may play a role in the etiology and pathogenesis of suicidal behavior and development of psychiatric disorders.
Collapse
Affiliation(s)
- Janna Nissen
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.
| | - Betina Trabjerg
- National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark; The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Denmark
| | - M G Pedersen
- National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark; The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Denmark; Centre for Integrated Register-based Research, CIRRAU, Aarhus University, Aarhus, Denmark
| | - Karina Banasik
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | | | - Erik Sørensen
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Kaspar René Nielsen
- Department of Clinical Immunology, Aalborg University Hospital, Aalborg, Denmark
| | - Christian Erikstrup
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| | | | | | - Peter Bruun-Rasmussen
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - David Westergaard
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - T F Hansen
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark; Danish Headache Center, Department of Neurology, Rigshospitalet-Glostrup, Denmark; Institute of Biological Psychiatry, Mental Health Centre Sct. Hans, Copenhagen University Hospital, Roskilde, Denmark
| | - Carsten B Pedersen
- National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark; The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Denmark; Centre for Integrated Register-based Research, CIRRAU, Aarhus University, Aarhus, Denmark
| | - Thomas Werge
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Denmark; Institute of Biological Psychiatry, Mental Health Centre Sct. Hans, Copenhagen University Hospital, Roskilde, Denmark
| | - Fuller Torrey
- Stanley Medical Research Institute, Kensington, MD School of Medicine, Baltimore, USA
| | - Henrik Hjalgrim
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark; Department of Hematology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Preben Bo Mortensen
- National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark; The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Denmark; Centre for Integrated Register-based Research, CIRRAU, Aarhus University, Aarhus, Denmark
| | - Robert Yolken
- Stanley Division of Developmental Neurovirology, Stanley Neurovirology Laboratory, Johns Hopkins University, USA
| | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Henrik Ullum
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - K S Burgdorf
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
55
|
Sharma S, Akundi RS. Mitochondria: A Connecting Link in the Major Depressive Disorder Jigsaw. Curr Neuropharmacol 2019; 17:550-562. [PMID: 29512466 PMCID: PMC6712299 DOI: 10.2174/1570159x16666180302120322] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 02/02/2018] [Accepted: 02/27/2018] [Indexed: 12/19/2022] Open
Abstract
Background Depression is a widespread phenomenon with varying degrees of pathology in different patients. Various hypotheses have been proposed for the cause and continuance of depression. Some of these include, but not limited to, the monoamine hypothesis, the neuroendocrine hypothesis, and the more recent epigenetic and inflammatory hypotheses. Objective In this article, we review all the above hypotheses with a focus on the role of mitochondria as the connecting link. Oxidative stress, respiratory activity, mitochondrial dynamics and metabolism are some of the mitochondria-dependent factors which are affected during depression. We also propose exogenous ATP as a contributing factor to depression. Result Literature review shows that pro-inflammatory markers are elevated in depressive individuals. The cause for elevated levels of cytokines in depression is not completely understood. We propose exogenous ATP activates purinergic receptors which in turn increase the levels of various pro-inflammatory factors in the pathophysiology of depression. Conclusion Mitochondria are integral to the function of neurons and undergo dysfunction in major depressive disorder patients. This dysfunction is reflected in all the various hypotheses that have been proposed for depression. Among the newer targets identified, which also involve mitochondria, includes the role of exogenous ATP. The diversity of purinergic receptors, and their differential expression among various individuals in the population, due to genetic and environmental (prenatal) influences, may influence the susceptibility and severity of depression. Identifying specific receptors involved and using patient-specific purinergic receptor antagonist may be an appropriate therapeutic course in the future.
Collapse
Affiliation(s)
- Shilpa Sharma
- Neuroinflammation Research Lab, Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi, India
| | - Ravi S Akundi
- Neuroinflammation Research Lab, Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi, India
| |
Collapse
|
56
|
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: 52] [Impact Index Per Article: 8.7] [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
|
57
|
Quinlan S, Merino-Serrais P, Di Grande A, Dussmann H, Prehn JHM, Ní Chonghaile T, Henshall DC, Jimenez-Mateos EM. The Anti-inflammatory Compound Candesartan Cilexetil Improves Neurological Outcomes in a Mouse Model of Neonatal Hypoxia. Front Immunol 2019; 10:1752. [PMID: 31396238 PMCID: PMC6667988 DOI: 10.3389/fimmu.2019.01752] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 07/10/2019] [Indexed: 11/15/2022] Open
Abstract
Recent studies suggest that mild hypoxia-induced neonatal seizures can trigger an acute neuroinflammatory response leading to long-lasting changes in brain excitability along with associated cognitive and behavioral deficits. The cellular elements and signaling pathways underlying neuroinflammation in this setting remain incompletely understood but could yield novel therapeutic targets. Here we show that brief global hypoxia-induced neonatal seizures in mice result in transient cytokine production, a selective expansion of microglia and long-lasting changes to the neuronal structure of pyramidal neurons in the hippocampus. Treatment of neonatal mice after hypoxia-seizures with the novel anti-inflammatory compound candesartan cilexetil suppressed acute seizure-damage and mitigated later-life aggravated seizure responses and hippocampus-dependent learning deficits. Together, these findings improve our understanding of the effects of neonatal seizures and identify potentially novel treatments to protect against short and long-lasting harmful effects.
Collapse
Affiliation(s)
- Sean Quinlan
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Paula Merino-Serrais
- Division for Neurogeriatrics, Department of Neurobiology Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden.,Departamento de Neurobiologia Funcional y de Sistemas, Instituto Cajal, Consejo Superior de Investigaciones Cientificas, Madrid, Spain
| | - Alessandra Di Grande
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Heiko Dussmann
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Jochen H M Prehn
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland.,FutureNeuro Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Tríona Ní Chonghaile
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - David C Henshall
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland.,FutureNeuro Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland.,INFANT Research Centre, UCC, Cork, Ireland
| | - Eva M Jimenez-Mateos
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland.,Department of Physiology, School of Medicine, Trinity College Dublin, The University of Dublin, Dublin, Ireland
| |
Collapse
|
58
|
Khandaker GM, Meyer U, Jones PB. From Infection to the Microbiome: An Evolving Role of Microbes in Schizophrenia. Curr Top Behav Neurosci 2019; 44:67-84. [PMID: 30847804 PMCID: PMC6732248 DOI: 10.1007/7854_2018_84] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The study of microorganisms such as bacteria, viruses, archaea, fungi, and protozoa in the context of psychiatric disorders may be surprising to some. This intersection of disciplines, however, has a rich history and is currently revitalized by newfound functions of the microbiome and the gut-brain axis in human diseases. Schizophrenia, in particular, fits this model as a disorder with gene and environmental roots that may be anchored in the immune system. In this context, the combination of a precisely timed pathogen exposure in a person with genetically encoded altered immunity may have especially destructive consequences for the central nervous system (CNS). Furthermore, significant components of immunity, such as the development of the immune response and the concept of immune tolerance, are largely dictated by the commensal residents of the microbiome. When this community of microbes is imbalanced, perhaps as the result of a pathogen invasion, stress, or immune gene deficiency, a pathological cycle of localized inflammation, endothelial barrier compromise, translocation of gut-derived products, and systemic inflammation may ensue. If these pathologies enable access of gut and microbial metabolites and immune molecules to the CNS across the blood-brain barrier (BBB), and studies of the gut-brain axis support this hypothesis, a worsening of cognitive deficits and psychiatric symptoms is predicted to occur in susceptible individuals with schizophrenia. In this chapter, we review the role of microbes in various stages of this model and how these organisms may contribute to documented phenotypes of schizophrenia. An increased understanding of the role of pathogens and the microbiome in psychiatric disorders will better guide the development of microbial and immune-based therapeutics for disease prevention and treatment.
Collapse
Affiliation(s)
- Golam M. Khandaker
- grid.5335.00000000121885934Department of Psychiatry, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Urs Meyer
- grid.5801.c0000 0001 2156 2780Verhaltensneurobiologie, ETH Zürich, Schwerzenbach, Switzerland
| | - Peter B. Jones
- grid.5335.00000000121885934Department of Psychiatry, University of Cambridge School of Clinical Medicine, Cambridge, UK
| |
Collapse
|
59
|
Stratification and prediction of remission in first-episode psychosis patients: the OPTiMiSE cohort study. Transl Psychiatry 2019; 9:20. [PMID: 30655509 PMCID: PMC6336802 DOI: 10.1038/s41398-018-0366-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 12/10/2018] [Indexed: 11/09/2022] Open
Abstract
Early response to first-line antipsychotic treatments is strongly associated with positive long-term symptomatic and functional outcome in psychosis. Unfortunately, attempts to identify reliable predictors of treatment response in first-episode psychosis (FEP) patients have not yet been successful. One reason for this could be that FEP patients are highly heterogeneous in terms of symptom expression and underlying disease biological mechanisms, thereby impeding the identification of one-size-fits-all predictors of treatment response. We have used a clustering approach to stratify 325 FEP patients into four clinical subtypes, termed C1A, C1B, C2A and C2B, based on their symptoms assessed using the Positive and Negative Syndrome Scale (PANSS) scale. Compared to C1B, C2A and C2B patients, those from the C1A subtype exhibited the most severe symptoms and were the most at risk of being non-remitters when treated with the second-generation antipsychotic drug amisulpride. Before treatment, C1A patients exhibited higher serum levels of several pro-inflammatory cytokines and inflammation-associated biomarkers therefore validating our stratification approach on external biological measures. Most importantly, in the C1A subtype, but not others, lower serum levels of interleukin (IL)-15, higher serum levels of C-X-C motif chemokine 12 (CXCL12), previous exposure to cytomegalovirus (CMV), use of recreational drugs and being younger were all associated with higher odds of being non-remitters 4 weeks after treatment. The predictive value of this model was good (mean area under the curve (AUC) = 0.73 ± 0.10), and its specificity and sensitivity were 45 ± 0.09% and 83 ± 0.03%, respectively. Further validation and replication of these results in clinical trials would pave the way for the development of a blood-based assisted clinical decision support system in psychosis.
Collapse
|
60
|
Basic Concept of Microglia Biology and Neuroinflammation in Relation to Psychiatry. Curr Top Behav Neurosci 2019; 44:9-34. [PMID: 30739307 DOI: 10.1007/7854_2018_83] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The hypothesis that the neuroimmune system plays a role in the pathogenesis of different psychiatric disorders, including schizophrenia, depression, and bipolar disease, has attained increasing interest over the past years. Previously thought to have the sole purpose of protecting the central nervous system (CNS) from harmful stimuli, it is now known that the central immune system is critically involved in regulating physiological processes including neurodevelopment, synaptic plasticity, and circuit maintenance. Hence, alterations in microglia - the main immune cell of the CNS - and/or inflammatory factors do not unequivocally connote ongoing neuroinflammation or neuroinflammatory processes per se but rather might signify changes in brain homoeostasis. Despite this, psychiatric research tends to equate functional changes in microglia or alterations in other immune mediators with neuroinflammation. It is the main impetus of this chapter to overcome some of the current misconceptions and possible oversimplifications with respect to neuroinflammation and microglia activity in psychiatry. In order to do so, we will first provide an overview of the basic concepts of neuroinflammation and neuroinflammatory processes. We will then focus on microglia with respect to their ontogeny and immunological and non-immunological functions presenting novel insights on how microglia communicate with other cell types of the central nervous system to ensure proper brain functioning. And lastly, we will delineate the non-immunological functions of inflammatory cytokines in order to address the possible misconception of equating alterations in central cytokine levels with ongoing central inflammation. We hereby hope to help unravel the functional relevance of neuroimmune dysfunctions in psychiatric illnesses and provide future research directions in the field of psychoneuroimmunology.
Collapse
|
61
|
Belz M, Rehling N, Schmidt U, Wiltfang J, Kis B, Wolff-Menzler C. Bacterial infections among patients with psychiatric disorders: Relation with hospital stay, age, and psychiatric diagnoses. PLoS One 2018; 13:e0208458. [PMID: 30513128 PMCID: PMC6279031 DOI: 10.1371/journal.pone.0208458] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 11/17/2018] [Indexed: 11/17/2022] Open
Abstract
The prevalence of infections is supposed to be higher in older patients and to extend the length of hospital stays. This study aimed, first, to test this supposition within a large psychiatric population which we divided into four clusters of psychiatric ICD-10 diagnoses: F00-F03 (dementias), F10 (substance disorders), F20-29 (schizophrenia, schizophreniform and other non-mood psychotic disorders), F32-F33 (major depressive disorders). Second, despite the increasing evidence for the role of infections in psychiatric disorders, it is, to the best of our knowledge, largely unknown whether the rates of infections with pathogens of the four most frequent germ families differ between psychiatric diseases. Thus, in a retrospective study, the results of clinical routine examinations (pap smear, analysis of midstream urine, stool) dependent on symptoms in 8545 patients of a German psychiatric clinic were analyzed in a 12-year dataset. Results show that a longer hospital stay was associated with an increased number of microbiological tests, but led to no significant difference between positive vs. negative findings. Consistent with previous studies, patients with infections were older than patients without infections. For the F10 diagnosis cluster we found a significantly reduced (F10: Staphylococcaceae) and for the F20-29 cluster a heightened risk of infections (Staphylococcaceae, Corynebacteriaceae). Furthermore, patients belonging to the F00-F03 cluster exhibited elevated rates of infections with all four germ families. The latter can be ascribed to patients' age as we found higher age to be associated with these infections, independently of the presence of dementia. Our results suggest that different psychiatric diagnoses are associated with a heightened or lowered risk of bacterial infections and, furthermore, that clinical routine infection-screenings for elderly psychiatric patients seems to be reasonable.
Collapse
Affiliation(s)
- Michael Belz
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Göttingen, Lower-Saxony, Germany
| | - Nico Rehling
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Göttingen, Lower-Saxony, Germany
| | - Ulrike Schmidt
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Göttingen, Lower-Saxony, Germany
| | - Jens Wiltfang
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Göttingen, Lower-Saxony, Germany.,German Center for Neurodegenerative Diseases (DZNE), Goettingen, Lower-Saxony, Germany.,Institute for Biomedicine (iBiMED), Medical Sciences Department, University of Aveiro, Aveiro, Portugal
| | - Bernhard Kis
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Göttingen, Lower-Saxony, Germany
| | - Claus Wolff-Menzler
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Göttingen, Lower-Saxony, Germany
| |
Collapse
|
62
|
Kannan G, Prandovszky E, Severance E, Yolken RH, Pletnikov MV. A New T. gondii Mouse Model of Gene-Environment Interaction Relevant to Psychiatric Disease. SCIENTIFICA 2018; 2018:7590958. [PMID: 30631636 PMCID: PMC6305013 DOI: 10.1155/2018/7590958] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 10/11/2018] [Indexed: 06/09/2023]
Abstract
Infection with the protozoan parasite, Toxoplasma gondii (T. gondii), was linked to several psychiatric disorders. The exact mechanisms of a hypothesized contribution of T. gondii infection are poorly understood, and it appears that only a subset of seropositive individuals go on to develop a mental illness, suggesting genetic vulnerability. In order to stimulate mechanistic studies of how exposure to T. gondii could interact with genetic predisposition to psychiatric disorders, we have generated and characterized a mouse model of chronic T. gondii infection in BALB/c mice with inducible forebrain neuronal expression of a C-terminus truncated dominant-negative form of disrupted-in-schizophrenia 1 (DN-DISC1). In this gene-environment interaction (GxE) model, exposing control and DN-DISC1 male and female mice to T. gondii produced sex-dependent abnormalities in locomotor activity and prepulse inhibition of the acoustic startle. No genotype- or sex-dependent effects were found on levels of anti-Toxoplasma IgG antibodies or anti-NMDAR or C1q antibodies. Our work demonstrates that a psychiatric genetic risk factor, DN-DISC1, modulates the neurobehavioral effects of chronic T. gondii infection in a sex-dependent manner. The present T. gondii model of GxE provides a valuable experimental system for future mechanistic studies and evaluation of new treatments.
Collapse
Affiliation(s)
- Geetha Kannan
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Emese Prandovszky
- Stanley Neurovirology Laboratory, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Emily Severance
- Stanley Neurovirology Laboratory, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Robert H. Yolken
- Stanley Neurovirology Laboratory, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Mikhail V. Pletnikov
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
63
|
Wang LY, Chen SF, Chiang JH, Hsu CY, Shen YC. Autoimmune diseases are associated with an increased risk of schizophrenia: A nationwide population-based cohort study. Schizophr Res 2018; 202:297-302. [PMID: 29925476 DOI: 10.1016/j.schres.2018.06.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 06/08/2018] [Accepted: 06/11/2018] [Indexed: 01/13/2023]
Abstract
OBJECTIVES Studies have suggested a possible autoimmune contribution in a subset of patients with schizophrenia. The purpose of this study was to determine if a history of autoimmune diseases (AD) is associated with an increased risk of later onset of schizophrenia. METHODS Taiwan's National Health Insurance Research Database was used to identify a total of 64,817 AD patients and an equal number of age-matched control patients. The incidence rates of schizophrenia with a maximum follow-up period of 10 years between patients with and without AD were compared using a Cox proportional hazard model to estimate the hazard ratio (HR) and 95% confidence interval (95% CI). RESULTS The main finding was the discovery of a higher incidence of subsequent schizophrenia in patients with AD (HR: 1.72, 95% CI: 1.23-2.4) after adjustment for other demographic characteristics. Specifically, the risk of schizophrenia was observed to be a significant increase in systemic lupus erythematosus (3.73, 2.07-6.72), rheumatoid arthritis (2.89, 1.97-4.23), dermatomyositis (5.85, 1.32-25.94) and autoimmune vasculitis (2.44, 1.17-5.06). Also, this study revealed some potential risk factors for developing schizophrenia, including younger age (less than or equal to 50 years) and some comorbidities (hypertension, chronic obstructive pulmonary disease, and alcohol use disorder). Conversely, this study found that steroid use was a potential protective factor for the development of schizophrenia. CONCLUSIONS This study found that AD were associated with an increased risk of developing schizophrenia, suggesting that the abnormal autoimmune process was associated with an increase in the expression of psychiatric disturbances.
Collapse
Affiliation(s)
- Ling-Yi Wang
- Epidemiology and Biostatistics Consulting Center, Department of Medical Research and Department of Pharmacy, Tzu Chi General Hospital, Hualien, Taiwan
| | - Shih-Fen Chen
- Center of Medical Genetics, Tzu Chi General Hospital, Hualien, Taiwan
| | - Jen-Huai Chiang
- Management Office for Health Data, China Medical University Hospital at Taichung, and College of Medicine, China Medical University, Taichung, Taiwan
| | - Chung-Y Hsu
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
| | - Yu-Chih Shen
- Department of Psychiatry, Tzu Chi General Hospital at Hualien, and School of Medicine, Tzu Chi University, Hualien, Taiwan.
| |
Collapse
|
64
|
Severance EG, Yolken RH. Deciphering microbiome and neuroactive immune gene interactions in schizophrenia. Neurobiol Dis 2018; 135:104331. [PMID: 30471416 DOI: 10.1016/j.nbd.2018.11.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 10/29/2018] [Accepted: 11/19/2018] [Indexed: 02/07/2023] Open
Abstract
The body's microbiome represents an actively regulated network of novel mechanisms that potentially underlie the etiology and pathophysiology of a wide range of diseases. For complex brain disorders such as schizophrenia, understanding the cellular and molecular pathways that intersect the bidirectional gut-brain axis is anticipated to lead to new methods of treatment. The means by which the microbiome might differ across neuropsychiatric and neurological disorders are not known. Brain disorders as diverse as schizophrenia, major depression, Parkinson's disease and multiple sclerosis appear to share a common pathology of an imbalanced community of commensal microbiota, often measured in terms of a leaky gut phenotype accompanied by low level systemic inflammation. While environmental factors associated with these disease states might contribute to intestinal pathologies, products from a perturbed microbiome may also directly promote specific signs, symptoms and etiologies of individual disorders. We hypothesize that in schizophrenia, it is the putatively unique susceptibility related to genes that modulate the immune system and the gut-brain pleiotropy of these genes which leads to a particularly neuropathological response when challenged by a microbiome in dysbiosis. Consequences from exposure to this dysbiosis may occur during pre- or post-natal time periods and thus may interfere with normal neurodevelopment in those who are genetically predisposed. Here, we review the evidence from the literature which supports the idea that the intersection of the microbiome and immune gene susceptibility in schizophrenia is relevant etiologically and for disease progression. Figuring prominently at both ends of the gut-brain axis and at points in between are proteins encoded by genes found in the major histocompatibility complex (MHC), including select MHC as well as non-MHC complement pathway genes.
Collapse
Affiliation(s)
- Emily G Severance
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| | - Robert H Yolken
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
65
|
Orhan F, Schwieler L, Fatouros-Bergman H, Malmqvist A, Cervenka S, Collste K, Flyckt L, Farde L, Sellgren CM, Piehl F, Engberg G, Erhardt S. Increased number of monocytes and plasma levels of MCP-1 and YKL-40 in first-episode psychosis. Acta Psychiatr Scand 2018; 138:432-440. [PMID: 30132802 DOI: 10.1111/acps.12944] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/10/2018] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Accumulating evidence implicates immune activation in the development of schizophrenia. Here, monocyte numbers, monocyte chemoattractant protein-1 (MCP-1) and chitinase-3-like protein 1 (YKL-40) were investigated in plasma and cerebrospinal fluid (CSF) in first-episode psychosis (FEP) patients. METHOD CSF and blood were sampled from 42 first-episode psychosis (FEP) patients and 22 healthy controls. The levels of YKL-40 and MCP-1 were measured using electrochemiluminescence assay, and blood monocytes were counted using an XN-9000-hematology analyzer. RESULTS We found higher plasma levels of MCP-1 and YKL-40 in FEP patients compared with healthy controls, a condition that was unrelated to antipsychotic and/or anxiolytic medication. This was combined with an increased number of blood monocytes and a borderline significant increase in YKL-40 levels in the CSF of tobacco-free FEP patients. Plasma or CSF chemokines or blood monocytes did not correlate with the severity of symptoms or the level of functioning. CONCLUSION These data demonstrate activation of monocytes in FEP and strengthens the idea of an immune dysfunction of psychotic disorders. Further studies are required to perceive a role of YKL-40 and MCP-1 in the initiation and progression of schizophrenia.
Collapse
Affiliation(s)
- F Orhan
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - L Schwieler
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - H Fatouros-Bergman
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden
| | - A Malmqvist
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - S Cervenka
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden
| | - K Collste
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden
| | - L Flyckt
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden
| | - L Farde
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden.,PET Science Centre, Precision Medicine and Genomics, IMED Biotech Unit, AstraZeneca, Karolinska Institutet, Stockholm, Sweden
| | - C M Sellgren
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.,Department of Psychiatry, Harvard Medical School, Boston, MA, USA.,Center for Experimental Drugs and Diagnostics, Center for Human Genetic Research and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - F Piehl
- Department of Clinical Neuroscience, Neuroimmunology Unit, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | | | - G Engberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - S Erhardt
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
66
|
Deshpande SN, Nimgaonkar VL. Exploring the associations of herpes simplex virus infection and cognitive dysfunction in schizophrenia: Studies in India. Indian J Psychiatry 2018; 60:393-397. [PMID: 30581203 PMCID: PMC6278214 DOI: 10.4103/psychiatry.indianjpsychiatry_381_18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Cognitive dysfunctions being core features of schizophrenia (SZ), cause disability, increase burden and are refractory to treatment. Viral infections are not risk factors for SZ, but growing evidence indicates infection with some neurotropic viruses, particularly Herpes simplex virus type 1 (HSV -1) as a risk factor for cognitive dysfunction. STUDIES IN INDIA Three research studies in India are described. In the first, participants were evaluated for HSV-1 infection and cognitive functions (cases 198 and controls 100). In the second, patients and normal nonpsychotic control individuals were examined at baseline and followed up over 1-3 years (cases 138 and controls 88). In the third, a randomized, double-blind placebo-controlled antipsychotic adjunctive trial was conducted to examine the effect of anti-viral drug valacyclovir over 16 weeks on cognitive functioning (valacyclovir 30; placebo 32, treatment for 16 weeks). RESULTS OF INDIAN STUDIES Cross-sectional study: HSV-1 infection was associated with modest dysfunction, especially on attention (accuracy) and spatial processing (speed). LONGITUDINAL STUDY HSV-1 seropositive participants had lower scores at baseline on 6/16 measures, regardless of SZ diagnoses. At follow-up, there was a significant decline in HSV-1-positive participants for abstraction and mental flexibility and emotion discrimination. RANDOMIZED CONTROLLED TRIAL Significantly, greater improvement in accuracy index of emotion discrimination in the valacyclovir-treated versus placebo sample was found. CONCLUSIONS Indian studies are consistent with a causative role for HSV-1 in cognitive dysfunction regardless of SZ diagnosis; more rigorous studies of the causal hypothesis are needed, particularly larger randomized controlled trials.
Collapse
Affiliation(s)
| | - Vishwajit Laxmikant Nimgaonkar
- Department of Psychiatry and Genetics, School of Medicine, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
67
|
Abstract
Typical and atypical antipsychotics are the first-line treatments for schizophrenia, but these classes of drugs are not universally effective, and they can have serious side effects that impact compliance. Antipsychotic drugs generally target the dopamine pathways with some variation. As research of schizophrenia pathophysiology has shifted away from a strictly dopamine-centric focus, the development of new pharmacotherapies has waned. A field of inquiry with centuries-old roots is gaining traction in psychiatric research circles and may represent a new frontier for drug discovery in schizophrenia. At the forefront of this investigative effort is the immune system and its many components, pathways and phenotypes, which are now known to actively engage the brain. Studies in schizophrenia reveal an intricate association of environmentally-driven immune activation in concert with a disrupted genetic template. A consistent conduit through this gene-environmental milieu is the gut-brain axis, which when dysregulated can generate pathological autoimmunity. In this review, we present epidemiological and biochemical evidence in support of an autoimmune component in schizophrenia and depict gut processes and a dysbiotic microbiome as a source and perpetuator of autoimmune dysfunction in the brain. Within this framework, we review the role of infectious agents, inflammation, gut dysbioses and autoantibody propagation on CNS pathologies such as neurotransmitter receptor hypofunction and complement pathway-mediated synaptic pruning. We then review the new pharmacotherapeutic horizon and novel agents directed to impact these pathological conditions. At the core of this discourse is the understanding that schizophrenia is etiologically and pathophysiologically heterogeneous and thus its treatment requires individualized attention with disease state variants diagnosed with objective biomarkers.
Collapse
Affiliation(s)
| | | | - Robert H Yolken
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
68
|
Ajdacic-Gross V, Bechtiger L, Rodgers S, Müller M, Kawohl W, von Känel R, Mutsch M, Rössler W, Seifritz E, Castelao E, Strippoli MPF, Vandeleur C, Preisig M, Howell P. Subtypes of stuttering determined by latent class analysis in two Swiss epidemiological surveys. PLoS One 2018; 13:e0198450. [PMID: 30086147 PMCID: PMC6080750 DOI: 10.1371/journal.pone.0198450] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 05/18/2018] [Indexed: 02/03/2023] Open
Abstract
Aims Associations between stuttering in childhood and a broad spectrum of risk factors, associated factors and comorbidities were examined in two large epidemiological studies. Subtypes of stuttering were then identified based on latent class analysis (LCA). Methods Data were from two representative Swiss population samples: PsyCoLaus (N = 4,874, age 35–82 years) and the ZInEP Epidemiology Survey (N = 1,500, age 20–41 years). Associations between stuttering and sociodemographic characteristics, familial aggregation, comorbidity and psychosocial risk / associated factors were investigated in both samples. LCAs were conducted on selected items from people in both samples who reported having stuttered in childhood. Results Initial analyses linked early anxiety disorders, such as separation anxiety disorder and overanxious disorder, to stuttering (PsyCoLaus). ADHD was associated with stuttering in both datasets. In the analyses of risk / associated factors, dysfunctional parental relationships, inter-parental violence and further childhood adversities were mutual predictors of stuttering. Moreover, comorbidities were seen with hay fever, asthma, eczema and psoriasis (PsyCoLaus). Subsequent LCA identified an unspecific group of persons who self-reported that they stuttered and a group defined by associations with psychosocial adversities (ZINEP, PsyCoLaus) and atopic diseases (PsyCoLaus). Conclusions The two subtypes of developmental stuttering have different risk / associated factors and comorbidity patterns. Most of the factors are associated with vulnerability mechanisms that occur early in life and that have also been linked with other neurodevelopmental disorders. Both psychosocial and biological factors appear to be involved in the etiopathogenesis of stuttering.
Collapse
Affiliation(s)
- Vladeta Ajdacic-Gross
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Zurich, Switzerland
- ZInEP, The Zurich Program for Sustainable Development of Mental Health Services, University of Zurich, Zurich, Switzerland
- Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Zurich, Switzerland
- * E-mail: (VA); (PH)
| | - Laura Bechtiger
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Zurich, Switzerland
| | - Stephanie Rodgers
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Zurich, Switzerland
- ZInEP, The Zurich Program for Sustainable Development of Mental Health Services, University of Zurich, Zurich, Switzerland
| | - Mario Müller
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Zurich, Switzerland
- ZInEP, The Zurich Program for Sustainable Development of Mental Health Services, University of Zurich, Zurich, Switzerland
| | - Wolfram Kawohl
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Zurich, Switzerland
- ZInEP, The Zurich Program for Sustainable Development of Mental Health Services, University of Zurich, Zurich, Switzerland
| | - Roland von Känel
- Department of Consultation-Liaison Psychiatry and Psychosomatic Medicine, University Hospital Zurich, Zurich, Switzerland
| | - Margot Mutsch
- Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Zurich, Switzerland
| | - Wulf Rössler
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Zurich, Switzerland
- ZInEP, The Zurich Program for Sustainable Development of Mental Health Services, University of Zurich, Zurich, Switzerland
- Collegium Helveticum, University of Zurich and Swiss Federal Institute of Technology, Zurich, Switzerland
- Institute of Psychiatry, Laboratory of Neuroscience (LIM27), University of Sao Paulo, Sao Paulo, Brazil
| | - Erich Seifritz
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Zurich, Switzerland
| | - Enrique Castelao
- Department of Psychiatry, Lausanne University Hospital, Prilly, Switzerland
| | | | - Caroline Vandeleur
- Department of Psychiatry, Lausanne University Hospital, Prilly, Switzerland
| | - Martin Preisig
- Department of Psychiatry, Lausanne University Hospital, Prilly, Switzerland
| | - Peter Howell
- Department of Experimental Psychology, University College, London, United Kingdom
- * E-mail: (VA); (PH)
| |
Collapse
|
69
|
Abstract
PURPOSE OF REVIEW Viruses, particularly herpes simplex virus (HSV), may be a cause of Alzheimer's disease (AD). The evidence supporting the viral hypothesis suggests that antiviral treatment trials, which have not been conducted, are warranted. RECENT FINDINGS HSV1 (oral herpes) and HSV2 (genital herpes) can trigger amyloid aggregation, and their DNA is common in amyloid plaques. HSV1 reactivation is associated with tau hyperphosphorylation and possibly tau propagation. Anti-HSV drugs reduce Aβ and p-tau accumulation in infected mouse brains. Clinically, after the initial oral infection, herpes simplex virus-1 (HSV1) becomes latent in the trigeminal ganglion and recurrent reactivation may produce neuronal damage and AD pathology. Clinical studies show cognitive impairment in HSV seropositive patients, and antiviral drugs show strong efficacy against HSV. An antiviral treatment trial in AD is clearly warranted. A phase II treatment trial with valacyclovir, an anti-HSV drug, recently began with evaluation of clinical and biomarker outcomes.
Collapse
|
70
|
Avramopoulos D. Recent Advances in the Genetics of Schizophrenia. MOLECULAR NEUROPSYCHIATRY 2018; 4:35-51. [PMID: 29998117 PMCID: PMC6032037 DOI: 10.1159/000488679] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 03/21/2018] [Indexed: 12/27/2022]
Abstract
The last decade brought tremendous progress in the field of schizophrenia genetics. As a result of extensive collaborations and multiple technological advances, we now recognize many types of genetic variants that increase the risk. These include large copy number variants, rare coding inherited and de novο variants, and over 100 loci harboring common risk variants. While the type and contribution to the risk vary among genetic variants, there is concordance in the functions of genes they implicate, such as those whose RNA binds the fragile X-related protein FMRP and members of the activity-regulated cytoskeletal complex involved in learning and memory. Gene expression studies add important information on the biology of the disease and recapitulate the same functional gene groups. Studies of alternative phenotypes help us widen our understanding of the genetic architecture of mental function and dysfunction, how diseases overlap not only with each other but also with non-disease phenotypes. The challenge is to apply this new knowledge to prevention and treatment and help patients. The data generated so far and emerging technologies, including new methods in cell engineering, offer significant promise that in the next decade we will unlock the translational potential of these significant discoveries.
Collapse
Affiliation(s)
- Dimitrios Avramopoulos
- Institute of Genetic Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Psychiatry, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
71
|
Jézéquel J, Johansson EM, Leboyer M, Groc L. Pathogenicity of Antibodies against NMDA Receptor: Molecular Insights into Autoimmune Psychosis. Trends Neurosci 2018; 41:502-511. [PMID: 29807730 DOI: 10.1016/j.tins.2018.05.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 04/05/2018] [Accepted: 05/01/2018] [Indexed: 11/16/2022]
Abstract
Recent years have seen a flourishing literature on detection of circulating autoantibodies against neurotransmitter receptors in patients with neuropsychiatric disorders. These studies have generated hope for a better understanding of the underlying molecular dysfunctions and for appropriate therapeutic strategies. However, the detection of these autoantibodies in healthy subjects, and the lack of mechanistic insights have fostered debate about the pathogenic role of such autoantibodies. Here, we specifically discuss the biological evidence linking autoantibodies directed against the glutamatergic N-methyl-d-aspartate (NMDA) receptor (NMDAR-Abs) and psychosis, emphasising recent single-molecule imaging investigations that unveiled the impaired surface trafficking of NMDAR in the presence of NMDAR-Abs from psychotic patients. Although still in its infancy, the hypothesis that NMDAR-Abs from patients with psychosis play a pathogenic role is thus gaining support, opening avenues of fundamental and translational investigations.
Collapse
Affiliation(s)
- J Jézéquel
- Université de Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France; CNRS, IINS UMR 5297, Bordeaux, France
| | - E M Johansson
- Université de Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France; CNRS, IINS UMR 5297, Bordeaux, France
| | - M Leboyer
- University Paris Est Créteil, Psychiatry Department, Hopitaux Universitaires Henri Mondor, AP-HP, DHU PePSY, INSERM, U955, Créteil, France
| | - L Groc
- Université de Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France; CNRS, IINS UMR 5297, Bordeaux, France.
| |
Collapse
|
72
|
Zhong QY, Gelaye B, Fricchione GL, Avillach P, Karlson EW, Williams MA. Adverse obstetric and neonatal outcomes complicated by psychosis among pregnant women in the United States. BMC Pregnancy Childbirth 2018; 18:120. [PMID: 29720114 PMCID: PMC5930732 DOI: 10.1186/s12884-018-1750-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 04/19/2018] [Indexed: 12/30/2022] Open
Abstract
Background Adverse obstetric and neonatal outcomes among women with psychosis, particularly affective psychosis, has rarely been studied at the population level. We aimed to assess the risk of adverse obstetric and neonatal outcomes among women with psychosis (schizophrenia, affective psychosis, and other psychoses). Methods From the 2007 – 2012 National (Nationwide) Inpatient Sample, 23,507,597 delivery hospitalizations were identified. From the same hospitalization, International Classification of Diseases diagnosis codes were used to identify maternal psychosis and outcomes. Adjusted odds ratios (aOR) and 95% confidence intervals (CI) were obtained using logistic regression. Results The prevalence of psychosis at delivery was 698.76 per 100,000 hospitalizations. After adjusting for sociodemographic characteristics, smoking, alcohol/substance abuse, and pregnancy-related hypertension, women with psychosis were at a heightened risk for cesarean delivery (aOR = 1.26; 95% CI: 1.23 - 1.29), induced labor (aOR = 1.05; 95% CI: 1.02 - 1.09), antepartum hemorrhage (aOR = 1.22; 95% CI: 1.14 - 1.31), placental abruption (aOR = 1.22; 95% CI: 1.13 - 1.32), postpartum hemorrhage (aOR = 1.18; 95% CI: 1.10 - 1.27), premature delivery (aOR = 1.40; 95% CI: 1.36 - 1.46), stillbirth (aOR = 1.37; 95% CI: 1.23 - 1.53), premature rupture of membranes (aOR = 1.22; 95% CI: 1.15 - 1.29), fetal abnormalities (aOR = 1.49; 95% CI: 1.38 - 1.61), poor fetal growth (aOR = 1.26; 95% CI: 1.19 - 1.34), and fetal distress (aOR = 1.14; 95% CI: 1.10 - 1.18). Maternal death during hospitalizations (aOR = 1.00; 95% CI: 0.30 - 3.31) and excessive fetal growth (aOR = 1.06; 95% CI: 0.98 - 1.14) were not statistically significantly associated with psychosis. Conclusions Pregnant women with psychosis have elevated risk of several adverse obstetric and neonatal outcomes. Efforts to identify and manage pregnancies complicated by psychosis may contribute to improved outcomes. Electronic supplementary material The online version of this article (10.1186/s12884-018-1750-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Qiu-Yue Zhong
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, 677 Huntington Avenue, Room Kresge 502A, Boston, MA, 02115, USA.
| | - Bizu Gelaye
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, 677 Huntington Avenue, Room Kresge 502A, Boston, MA, 02115, USA
| | - Gregory L Fricchione
- Division of Psychiatry and Medicine, Pierce Division of Global Psychiatry, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Paul Avillach
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, 677 Huntington Avenue, Room Kresge 502A, Boston, MA, 02115, USA.,Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts, USA.,Children's Hospital Informatics Program, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Elizabeth W Karlson
- Division of Rheumatology, Allergy and Immunology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Michelle A Williams
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, 677 Huntington Avenue, Room Kresge 502A, Boston, MA, 02115, USA
| |
Collapse
|
73
|
Parks S, Avramopoulos D, Mulle J, McGrath J, Wang R, Goes FS, Conneely K, Ruczinski I, Yolken R, Pulver AE, Pearce BD. HLA typing using genome wide data reveals susceptibility types for infections in a psychiatric disease enriched sample. Brain Behav Immun 2018; 70:203-213. [PMID: 29574260 DOI: 10.1016/j.bbi.2018.03.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 02/27/2018] [Accepted: 03/03/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The infections Toxoplasma gondii (T. gondii), cytomegalovirus, and Herpes Simplex Virus Type 1 (HSV1) are common persistent infections that have been associated with schizophrenia and bipolar disorder. The major histocompatibility complex (MHC, termed HLA in humans) region has been implicated in these infections and these mental illnesses. The interplay of MHC genetics, mental illness, and infection has not been systematically examined in previous research. METHODS In a cohort of 1636 individuals, we used genome-wide association data to impute 7 HLA types (A, B, C, DRB1, DQA1, DQB1, DPB1), and combined this data with serology data for these infections. We used regression analysis to assess the association between HLA alleles, infections (individually and collectively), and mental disorder status (schizophrenia, bipolar disorder, controls). RESULTS After Bonferroni correction for multiple comparisons, HLA C∗07:01 was associated with increased HSV1 infection among mentally healthy controls (OR 3.4, p = 0.0007) but not in the schizophrenia or bipolar groups (P > 0.05). For the multiple infection outcome, HLA B∗ 38:01 and HLA C∗12:03 were protective in the healthy controls (OR ≈ 0.4) but did not have a statistically-significant effect in the schizophrenia or bipolar groups. T. gondii had several nominally-significant positive associations, including the haplotypes HLA DRB∗03:01 ∼ HLA DQA∗05:01 ∼ HLA DQB∗02:01 and HLA B∗08:01 ∼ HLA C∗07:01. CONCLUSIONS We identified HLA types that showed strong and significant associations with neurotropic infections. Since some of these associations depended on mental illness status, the engagement of HLA-related pathways may be altered in schizophrenia due to immunogenetic differences or exposure history.
Collapse
Affiliation(s)
- Samuel Parks
- Dept. of Epidemiology, Rollins School of Public Health, USA
| | - Dimitrios Avramopoulos
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jennifer Mulle
- Department of Human Genetics, Emory University, Atlanta, GA, USA
| | - John McGrath
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ruihua Wang
- McKusick Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Fernando S Goes
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Karen Conneely
- Department of Human Genetics, Emory University, Atlanta, GA, USA
| | - Ingo Ruczinski
- Bloomberg School of Public Heath, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert Yolken
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ann E Pulver
- Bloomberg School of Public Heath, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Brad D Pearce
- Dept. of Epidemiology, Rollins School of Public Health, USA.
| |
Collapse
|
74
|
McFarland R, Wang ZT, Jouroukhin Y, Li Y, Mychko O, Coppens I, Xiao J, Jones-Brando L, Yolken RH, Sibley LD, Pletnikov MV. AAH2 gene is not required for dopamine-dependent neurochemical and behavioral abnormalities produced by Toxoplasma infection in mouse. Behav Brain Res 2018; 347:193-200. [PMID: 29555339 DOI: 10.1016/j.bbr.2018.03.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 03/10/2018] [Accepted: 03/14/2018] [Indexed: 01/25/2023]
Abstract
Infection with the protozoan parasite, Toxoplasma gondii (T. gondii), has been associated with the increased risk for several psychiatric disorders. The exact mechanisms of a hypothesized contribution of T. gondii infection are poorly understood. The T. gondii genome contains two aromatic amino acid hydroxylase genes (AAH1 and AAH2) that encode proteins that can produce L-DOPA. One popular hypothesis posits that these encoded enzymes might influence dopamine (DA) production and hence DA synaptic transmission, leading to neurobehavioral abnormalities in the infected host. Prior studies have shown that deletion of these genes does not alter DA levels in the brain or exploratory activity in infected mice. However, possible effects of AAH gene deficiency on infection-induced brain and behavior alterations that are directly linked to DA synaptic transmission have not been evaluated. We found that chronic T. gondii infection of BALB/c mice leads to blunted response to amphetamine or cocaine and decreased expression of Dopamine Transporter (DAT) and Vesicular Monoamine Transporter 2 (VMAT2). Deletion of AAH2 had no effects on these changes in infected mice. Both wild type and Δaah2 strains produced comparable levels of neuroinflammation. Our findings demonstrate that AAH2 is not required for T. gondii infection-produced DA-dependent neurobehavioral abnormalities.
Collapse
Affiliation(s)
- Ross McFarland
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, 21205, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Zi Teng Wang
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Yan Jouroukhin
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Ye Li
- Stanley Neurovirology Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Olga Mychko
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Isabelle Coppens
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Jianchun Xiao
- Stanley Neurovirology Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Lorraine Jones-Brando
- Stanley Neurovirology Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Robert H Yolken
- Stanley Neurovirology Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - L David Sibley
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Mikhail V Pletnikov
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, 21205, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
75
|
Lindgren M, Torniainen-Holm M, Härkänen T, Dickerson F, Yolken RH, Suvisaari J. The association between toxoplasma and the psychosis continuum in a general population setting. Schizophr Res 2018; 193:329-335. [PMID: 28711477 DOI: 10.1016/j.schres.2017.06.052] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 06/27/2017] [Accepted: 06/27/2017] [Indexed: 11/18/2022]
Abstract
Toxoplasma gondii infection is associated with increased risk for psychosis. However, the possible association between T. gondii and psychotic-like symptoms in the general adult population is unknown. We investigated whether T. gondii is associated with psychotic-like symptoms and psychosis diagnoses using data from Health 2000, a large cross-sectional health survey of the Finnish general population aged 30 and above. Seropositivity to toxoplasma was defined as a cutoff of 50IU/ml of IgG antibodies. Lifetime psychotic-like symptoms were identified with section G of the Composite International Diagnostic Interview, Munich version (M-CIDI). Symptoms were considered clinically relevant if they caused distress or help-seeking or there were at least three of them. Lifetime psychotic disorders were screened from the sample and were diagnosed with DSM-IV using SCID-I interview and information from medical records. All data were available for 5906 participants. We adjusted for variables related to T. gondii seropositivity (age, gender, education, region of residence, cat ownership, and C-reactive protein measuring inflammation) in regression models. We found that T. gondii seropositivity was significantly associated with clinically relevant psychotic-like symptoms (OR 1.77, p=0.001) and with the number of psychotic-like symptoms (IRR=1.55, p=0.001). The association between toxoplasma and diagnosed psychotic disorders did not reach statistical significance (OR 1.45 for schizophrenia). In a large sample representing the whole Finnish adult population, we found that serological evidence of toxoplasma infection predicted psychotic-like symptoms, independent of demographic factors and levels of C-reactive protein. Toxoplasma infection may be a risk factor for manifestation of psychotic-like symptoms.
Collapse
Affiliation(s)
- Maija Lindgren
- National Institute for Health and Welfare (THL), Mental Health Unit, Finland.
| | - Minna Torniainen-Holm
- National Institute for Health and Welfare (THL), Mental Health Unit, Finland; Institute for Molecular Medicine Finland (FIMM), Helsinki, Finland
| | - Tommi Härkänen
- National Institute for Health and Welfare (THL), Health Monitoring Unit, Finland
| | - Faith Dickerson
- Stanley Research Program, Sheppard Pratt Health System, Baltimore, MD, USA
| | - Robert H Yolken
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, John Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jaana Suvisaari
- National Institute for Health and Welfare (THL), Mental Health Unit, Finland
| |
Collapse
|
76
|
Kakooza-Mwesige A, Mohammed AH, Kristensson K, Juliano SL, Lutwama JJ. Emerging Viral Infections in Sub-Saharan Africa and the Developing Nervous System: A Mini Review. Front Neurol 2018. [PMID: 29527187 PMCID: PMC5829034 DOI: 10.3389/fneur.2018.00082] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The global public health concern is heightened over the increasing number of emerging viruses, i.e., newly discovered or previously known that have expanded into new geographical zones. These viruses challenge the health-care systems in sub-Saharan Africa (SSA) countries from which several of them have originated and been transmitted by insects worldwide. Some of these viruses are neuroinvasive, but have been relatively neglected by neuroscientists. They may provide experiments by nature to give a time window for exposure to a new virus within sizeable, previously non-infected human populations, which, for instance, enables studies on potential long-term or late-onset effects on the developing nervous system. Here, we briefly summarize studies on the developing brain by West Nile, Zika, and Chikungunya viruses, which are mosquito-borne and have spread worldwide out of SSA. They can all be neuroinvasive, but their effects vary from malformations caused by prenatal infections to cognitive disturbances following perinatal or later infections. We also highlight Ebola virus, which can leave surviving children with psychiatric disturbances and cause persistent infections in the non-human primate brain. Greater awareness within the neuroscience community is needed to emphasize the menace evoked by these emerging viruses to the developing brain. In particular, frontline neuroscience research should include neuropediatric follow-up studies in the field on long-term or late-onset cognitive and behavior disturbances or neuropsychiatric disorders. Studies on pathogenetic mechanisms for viral-induced perturbations of brain maturation should be extended to the vulnerable periods when neurocircuit formations are at peaks during infancy and early childhood.
Collapse
Affiliation(s)
- Angelina Kakooza-Mwesige
- Department of Paediatrics and Child Health, Makerere University College of Health Sciences and Mulago Hospital, Kampala, Uganda
| | | | | | - Sharon L Juliano
- Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Julius J Lutwama
- Arbovirology Laboratory, Uganda Virus Research Institute, Entebbe, Uganda
| |
Collapse
|
77
|
Giannitelli M, Consoli A, Raffin M, Jardri R, Levinson DF, Cohen D, Laurent-Levinson C. An overview of medical risk factors for childhood psychosis: Implications for research and treatment. Schizophr Res 2018; 192:39-49. [PMID: 28526280 DOI: 10.1016/j.schres.2017.05.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 05/05/2017] [Accepted: 05/08/2017] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Psychotic disorders in childhood and early adolescence often progress to chronic schizophrenia, but in many cases there are diagnosable medical and genetic causes or risk factors. We reviewed our clinical experience and the relevant literature to identify these factors and to define their clinical features, appropriate work-up and treatment. METHOD We reviewed the results of comprehensive medical evaluations of 160 psychotic children and adolescents in our center. We also searched the Medline database (January 1994 to December 2015) with the following keywords and combinations: early onset schizophrenia, childhood onset schizophrenia, early onset psychosis, first episode psychosis, inborn errors of metabolism (IEM), genetic syndrome, copy number variants, autoimmune disorders, endocrine diseases, nutritional deficiencies, central nervous system infections, movement disorders, and epilepsy. RESULTS In our center, 12.5% of cases had medical disorders likely to be contributing to psychosis. Based on 66 relevant papers and our experience, we describe the clinical features of multiple genetic syndromes, IEM, and autoimmune, neurological, endocrinological and nutritional disorders that increase the risk of psychotic disorders in childhood and adolescence. We propose an algorithm for systematic laboratory evaluation, informed by clinical examination, emphasizing common and/or treatable factors. CONCLUSIONS In children and early adolescents with psychotic disorders, systematic medical work-up is warranted to identify medical and genetic factors. Not every rare cause can be worked up, thus careful clinical examinations are required to detect medical, neurological and genetic signs. Comprehensive medical evaluation can detect treatable diseases among cases of early-onset psychosis.
Collapse
Affiliation(s)
- Marianna Giannitelli
- Sorbonne Universités, UPMC Univ Paris 06, Assistance Publique-Hôpitaux de Paris, Groupe de Recherche Clinique n°15 (PSYDEV), Hôpital Pitié-Salpêtrière, 47-83, boulevard de l'Hôpital, 75013 Paris, France; Centre de référence des maladies rares à expression psychiatrique, Department of Child and Adolescent Psychiatry, Hôpital Pitié-Salpêtrière, 47-83, boulevard de l'Hôpital, 75013 Paris, France; CNRS UMR 7222, Institut des Systèmes Intelligents et Robotiques, Université Pierre et Marie Curie, 1 place Jussieu, 75005 Paris, France
| | - Angèle Consoli
- Sorbonne Universités, UPMC Univ Paris 06, Assistance Publique-Hôpitaux de Paris, Groupe de Recherche Clinique n°15 (PSYDEV), Hôpital Pitié-Salpêtrière, 47-83, boulevard de l'Hôpital, 75013 Paris, France; Centre de référence des maladies rares à expression psychiatrique, Department of Child and Adolescent Psychiatry, Hôpital Pitié-Salpêtrière, 47-83, boulevard de l'Hôpital, 75013 Paris, France
| | - Marie Raffin
- Sorbonne Universités, UPMC Univ Paris 06, Assistance Publique-Hôpitaux de Paris, Groupe de Recherche Clinique n°15 (PSYDEV), Hôpital Pitié-Salpêtrière, 47-83, boulevard de l'Hôpital, 75013 Paris, France
| | - Renaud Jardri
- University of Lille, SCALab, CNRS UMR-9193 & CHU Lille, CURE platform, Fontan Hospital, Lille, France
| | - Douglas F Levinson
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - David Cohen
- Sorbonne Universités, UPMC Univ Paris 06, Assistance Publique-Hôpitaux de Paris, Groupe de Recherche Clinique n°15 (PSYDEV), Hôpital Pitié-Salpêtrière, 47-83, boulevard de l'Hôpital, 75013 Paris, France; Centre de référence des maladies rares à expression psychiatrique, Department of Child and Adolescent Psychiatry, Hôpital Pitié-Salpêtrière, 47-83, boulevard de l'Hôpital, 75013 Paris, France; CNRS UMR 7222, Institut des Systèmes Intelligents et Robotiques, Université Pierre et Marie Curie, 1 place Jussieu, 75005 Paris, France
| | - Claudine Laurent-Levinson
- Sorbonne Universités, UPMC Univ Paris 06, Assistance Publique-Hôpitaux de Paris, Groupe de Recherche Clinique n°15 (PSYDEV), Hôpital Pitié-Salpêtrière, 47-83, boulevard de l'Hôpital, 75013 Paris, France; Centre de référence des maladies rares à expression psychiatrique, Department of Child and Adolescent Psychiatry, Hôpital Pitié-Salpêtrière, 47-83, boulevard de l'Hôpital, 75013 Paris, France; Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA.
| |
Collapse
|
78
|
Victor TA, Khalsa SS, Simmons WK, Feinstein JS, Savitz J, Aupperle RL, Yeh HW, Bodurka J, Paulus MP. Tulsa 1000: a naturalistic study protocol for multilevel assessment and outcome prediction in a large psychiatric sample. BMJ Open 2018; 8:e016620. [PMID: 29371263 PMCID: PMC5786129 DOI: 10.1136/bmjopen-2017-016620] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 11/07/2017] [Accepted: 11/09/2017] [Indexed: 12/31/2022] Open
Abstract
INTRODUCTION Although neuroscience has made tremendous progress towards understanding the basic neural circuitry underlying important processes such as attention, memory and emotion, little progress has been made in applying these insights to psychiatric populations to make clinically meaningful treatment predictions. The overall aim of the Tulsa 1000 (T-1000) study is to use the NIMH Research Domain Criteria framework in order to establish a robust and reliable dimensional set of variables that quantifies the positive and negative valence, cognition and arousal domains, including interoception, to generate clinically useful treatment predictions. METHODS AND ANALYSIS The T-1000 is a naturalistic study that will recruit, assess and longitudinally follow 1000 participants, including healthy controls and treatment-seeking individuals with mood, anxiety, substance use and eating disorders. Each participant will undergo interview, behavioural, biomarker and neuroimaging assessments over the course of 1 year. The study goal is to determine how disorders of affect, substance use and eating behaviour organise across different levels of analysis (molecules, genes, cells, neural circuits, physiology, behaviour and self-report) to predict symptom severity, treatment outcome and long-term prognosis. The data will be used to generate computational models based on Bayesian statistics. The final end point of this multilevel latent variable analysis will be standardised assessments that can be developed into clinical tools to help clinicians predict outcomes and select the best intervention for each individual, thereby reducing the burden of mental disorders, and taking psychiatry a step closer towards personalised medicine. ETHICS AND DISSEMINATION Ethical approval was obtained from Western Institutional Review Board screening protocol #20101611. The dissemination plan includes informing health professionals of results for clinical practice, submitting results to journals for peer-reviewed publication, presenting results at national and international conferences and making the dataset available to researchers and mental health professionals. TRIAL REGISTRATION NUMBER NCT02450240; Pre-results.
Collapse
Affiliation(s)
| | - Sahib S Khalsa
- Laureate Institute for Brain Research, Tulsa, Oklahoma, USA
- Oxley College of Health Sciences, University of Tulsa, Tulsa, Oklahoma, USA
| | - W Kyle Simmons
- Laureate Institute for Brain Research, Tulsa, Oklahoma, USA
- Oxley College of Health Sciences, University of Tulsa, Tulsa, Oklahoma, USA
| | - Justin S Feinstein
- Laureate Institute for Brain Research, Tulsa, Oklahoma, USA
- Oxley College of Health Sciences, University of Tulsa, Tulsa, Oklahoma, USA
| | - Jonathan Savitz
- Laureate Institute for Brain Research, Tulsa, Oklahoma, USA
- Oxley College of Health Sciences, University of Tulsa, Tulsa, Oklahoma, USA
| | - Robin L Aupperle
- Laureate Institute for Brain Research, Tulsa, Oklahoma, USA
- Oxley College of Health Sciences, University of Tulsa, Tulsa, Oklahoma, USA
| | - Hung-Wen Yeh
- Laureate Institute for Brain Research, Tulsa, Oklahoma, USA
| | - Jerzy Bodurka
- Laureate Institute for Brain Research, Tulsa, Oklahoma, USA
- Stephenson School of Biomedical Engineering, The University of Oklahoma, Tulsa, Oklahoma, USA
| | | |
Collapse
|
79
|
Notter T, Coughlin JM, Sawa A, Meyer U. Reconceptualization of translocator protein as a biomarker of neuroinflammation in psychiatry. Mol Psychiatry 2018; 23:36-47. [PMID: 29203847 DOI: 10.1038/mp.2017.232] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 09/05/2017] [Accepted: 10/02/2017] [Indexed: 02/06/2023]
Abstract
A great deal of interest in psychiatric research is currently centered upon the pathogenic role of inflammatory processes. Positron emission tomography (PET) using radiolabeled ligands selective for the 18 kDa translocator protein (TSPO) has become the most widely used technique to assess putative neuroimmune abnormalities in vivo. Originally used to detect discrete neurotoxic damages, TSPO has generally turned into a biomarker of 'neuroinflammation' or 'microglial activation'. Psychiatric research has mostly accepted these denotations of TSPO, even if they may be inadequate and misleading under many pathological conditions. A reliable and neurobiologically meaningful diagnosis of 'neuroinflammation' or 'microglial activation' is unlikely to be achieved by the sole use of TSPO PET imaging. It is also very likely that the pathological meanings of altered TSPO binding or expression are disease-specific, and therefore, not easily generalizable across different neuropathologies or inflammatory conditions. This difficulty is intricately linked to the varying (and still ill-defined) physiological functions and cellular expression patterns of TSPO in health and disease. While altered TSPO binding or expression may indeed mirror ongoing neuroinflammatory processes in some cases, it may reflect other pathophysiological processes such as abnormalities in cell metabolism, energy production and oxidative stress in others. Hence, the increasing popularity of TSPO PET imaging has paradoxically introduced substantial uncertainty regarding the nature and meaning of neuroinflammatory processes and microglial activation in psychiatry, and likely in other neuropathological conditions as well. The ambiguity of conceiving TSPO simply as a biomarker of 'neuroinflammation' or 'microglial activation' calls for alternative interpretations and complimentary approaches. Without the latter, the ongoing scientific efforts and excitement surrounding the role of the neuroimmune system in psychiatry may not turn into therapeutic hope for affected individuals.
Collapse
Affiliation(s)
- T Notter
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - J M Coughlin
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, MD, USA.,Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - A Sawa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - U Meyer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
80
|
Pollak TA, Drndarski S, Stone JM, David AS, McGuire P, Abbott NJ. The blood-brain barrier in psychosis. Lancet Psychiatry 2018; 5:79-92. [PMID: 28781208 DOI: 10.1016/s2215-0366(17)30293-6] [Citation(s) in RCA: 207] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 06/12/2017] [Accepted: 06/27/2017] [Indexed: 02/08/2023]
Abstract
Blood-brain barrier pathology is recognised as a central factor in the development of many neurological disorders, but much less is known about the role of the blood-brain barrier in psychiatric disorders. We review post-mortem, serum-biomarker, CSF-biomarker, and neuroimaging studies that have examined blood-brain barrier structure and function in schizophrenia and related psychoses. We consider how blood-brain barrier dysfunction could relate to glutamatergic and inflammatory abnormalities, which are increasingly understood to play a part in the pathogenesis of psychosis. Mechanisms by which the blood-brain barrier and its associated solute transporters moderate CNS availability of antipsychotic drugs are summarised. We conclude that the complex nature of blood-brain barrier dysfunction in psychosis might be relevant to many aspects of disrupted neuronal and synaptic function, increased permeability to inflammatory molecules, disrupted glutamate homoeostasis, impaired action of antipsychotics, and development of antipsychotic resistance. Future research should address the longitudinal course of blood-brain barrier alterations in psychosis, to determine whether blood-brain barrier dysfunction is a cause or consequence of the pathology associated with the disorder.
Collapse
Affiliation(s)
- Thomas A Pollak
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| | | | - James M Stone
- Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Anthony S David
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Philip McGuire
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - N Joan Abbott
- Institute of Pharmaceutical Science, King's College London, London, UK
| |
Collapse
|
81
|
Morais FB, Arantes TEFE, Muccioli C. Seroprevalence and Manifestations of Ocular Toxoplasmosis in Patients with Schizophrenia. Ocul Immunol Inflamm 2017; 27:134-137. [PMID: 29283732 DOI: 10.1080/09273948.2017.1408843] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
PURPOSE Recent studies have linked infectious agents such as Toxoplasma gondii to schizophrenia. We investigated the seroprevalence of T. gondii and conducted ophthalmologic examinations in schizophrenia patients and controls to identify lesions suggestive of ocular toxoplasmosis. METHODS During 2015 and 2016, 34 schizophrenia patients and 85 healthy controls underwent ophthalmologic examination and anti-T. gondii IgG and IgM antibody measurements by chemiluminescence. RESULTS Schizophrenia patients had a higher prevalence of anti-T. gondii IgG positivity than controls (91.18% [95% confidence interval (CI), 77.04%-96.95%] vs. 70.59% [95% CI, 60.18%-79.21%]; p = 0.017). Anti-T. gondii IgM antibodies (acute form) were not detected in any patient. One (3%) schizophrenic patient and two (2.4%) control patients presented fundoscopic scarring. CONCLUSION The seropositivity rate was significantly higher among schizophrenia patients than among controls (p = 0.017). There was no association between the presence of fundoscopic scarring and schizophrenia (p = 1.000).
Collapse
Affiliation(s)
- Fábio Barreto Morais
- a Department of Ophthalmology , Universidade Federal de São Paulo - UNIFESP , São Paulo , SP , Brazil and
| | | | - Cristina Muccioli
- a Department of Ophthalmology , Universidade Federal de São Paulo - UNIFESP , São Paulo , SP , Brazil and
| |
Collapse
|
82
|
Severance EG, Tveiten D, Lindström LH, Yolken RH, Reichelt KL. The Gut Microbiota and the Emergence of Autoimmunity: Relevance to Major Psychiatric Disorders. Curr Pharm Des 2017; 22:6076-6086. [PMID: 27634185 DOI: 10.2174/1381612822666160914183804] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 08/30/2016] [Accepted: 09/06/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND Autoimmune phenotypes are prevalent in major psychiatric disorders. Disequilibria of cellular processes occurring in the gastrointestinal (GI) tract likely contribute to immune dysfunction in psychiatric disorders. As the venue of a complex community of resident microbes, the gut in a homeostatic state equates with a functional digestive system, cellular barrier stability and properly regulated recognition of self and non-self antigens. When gut processes become disrupted as a result of environmental or genetic factors, autoimmunity may ensue. METHODS Here, we review the issues pertinent to autoimmunity and the microbiome in psychiatric disorders and show that many of the reported immune risk factors for the development of these brain disorders are in fact related and consistent with dysfunctions occurring in the gut. We review the few human microbiome studies that have been done in people with psychiatric disorders and supplement this information with mechanistic data gleaned from experimental rodent studies. RESULTS These investigations demonstrate changes in behavior and brain biochemistry directly attributable to alterations in the gut microbiome. We present a model by which autoantigens are produced by extrinsicallyderived food and microbial factors bound to intrinsic components of the gut including receptors present in the enteric nervous system. CONCLUSION This new focus on examining activities outside of the CNS for relevance to the etiology and pathophysiology of psychiatric disorders may require new modalities or a re-evaluation of pharmaceutical targets found in peripheral systems.
Collapse
Affiliation(s)
- Emily G Severance
- Stanley Division of Developmental Neurovirology; Department of Pediatrics; Johns Hopkins University School of Medicine; 600 North Wolfe Street; Blalock 1105; Baltimore, MD 21287, USA
| | | | | | | | | |
Collapse
|
83
|
Nimgaonkar VL, Prasad KM, Chowdari KV, Severance EG, Yolken RH. The complement system: a gateway to gene-environment interactions in schizophrenia pathogenesis. Mol Psychiatry 2017; 22:1554-1561. [PMID: 28761078 PMCID: PMC5656502 DOI: 10.1038/mp.2017.151] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 05/15/2017] [Accepted: 05/16/2017] [Indexed: 02/08/2023]
Abstract
The pathogenesis of schizophrenia is considered to be multi-factorial, with likely gene-environment interactions (GEI). Genetic and environmental risk factors are being identified with increasing frequency, yet their very number vastly increases the scope of possible GEI, making it difficult to identify them with certainty. Accumulating evidence suggests a dysregulated complement pathway among the pathogenic processes of schizophrenia. The complement pathway mediates innate and acquired immunity, and its activation drives the removal of damaged cells, autoantigens and environmentally derived antigens. Abnormalities in complement functions occur in many infectious and autoimmune disorders that have been linked to schizophrenia. Many older reports indicate altered serum complement activity in schizophrenia, though the data are inconclusive. Compellingly, recent genome-wide association studies suggest repeat polymorphisms incorporating the complement 4A (C4A) and 4B (C4B) genes as risk factors for schizophrenia. The C4A/C4B genetic associations have re-ignited interest not only in inflammation-related models for schizophrenia pathogenesis, but also in neurodevelopmental theories, because rodent models indicate a role for complement proteins in synaptic pruning and neurodevelopment. Thus, the complement system could be used as one of the 'staging posts' for a variety of focused studies of schizophrenia pathogenesis. They include GEI studies of the C4A/C4B repeat polymorphisms in relation to inflammation-related or infectious processes, animal model studies and tests of hypotheses linked to autoimmune diseases that can co-segregate with schizophrenia. If they can be replicated, such studies would vastly improve our understanding of pathogenic processes in schizophrenia through GEI analyses and open new avenues for therapy.
Collapse
Affiliation(s)
- Vishwajit L. Nimgaonkar
- Department of Psychiatry, University of Pittsburgh, School of Medicine, Pittsburgh, PA
- Department of Human Genetics, University of Pittsburgh, Graduate School of Public Health, Pittsburgh, PA
| | - Konasale M. Prasad
- Department of Psychiatry, University of Pittsburgh, School of Medicine, Pittsburgh, PA
| | - Kodavali V. Chowdari
- Department of Psychiatry, University of Pittsburgh, School of Medicine, Pittsburgh, PA
| | - Emily G. Severance
- Stanley Division of Neurovirology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Robert H. Yolken
- Stanley Division of Neurovirology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Md
| |
Collapse
|
84
|
Al‐Diwani AAJ, Pollak TA, Irani SR, Lennox BR. Psychosis: an autoimmune disease? Immunology 2017; 152:388-401. [PMID: 28704576 PMCID: PMC5629440 DOI: 10.1111/imm.12795] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 07/03/2017] [Accepted: 07/05/2017] [Indexed: 12/22/2022] Open
Abstract
Psychotic disorders are common and disabling. Overlaps in clinical course in addition to epidemiological and genetic associations raise the possibility that autoimmune mechanisms may underlie some psychoses, potentially offering novel therapeutic approaches. Several immune loci including the major histocompatibility complex and B-cell markers CD19 and CD20 achieve genome-wide significance in schizophrenia. Emerging evidence suggests a potential role via neurodevelopment in addition to classical immune pathways. Additionally, lymphocyte biology is increasingly investigated. Some reports note raised peripheral CD19+ and reduced CD3+ lymphocyte counts, with altered CD4 : CD8 ratios in acute psychosis. Also, post-mortem studies have found CD3+ and CD20+ lymphocyte infiltration in brain regions that are of functional relevance to psychosis. More specifically, the recent paradigm of neuronal surface antibody-mediated (NSAb) central nervous system disease provides an antigen-specific model linking adaptive autoimmunity to psychopathology. NSAbs bind extracellular epitopes of signalling molecules that are classically implicated in psychosis such as NMDA and GABA receptors. This interaction may cause circuit dysfunction leading to psychosis among other neurological features in patients with autoimmune encephalitis. The detection of these cases is crucial as autoimmune encephalitis is ameliorated by commonly available immunotherapies. Meanwhile, the prevalence and relevance of these antibodies in people with isolated psychotic disorders is an area of emerging scientific and clinical interest. Collaborative efforts to achieve larger sample sizes, comparison of assay platforms, and placebo-controlled randomized clinical trials are now needed to establish an autoimmune contribution to psychosis.
Collapse
Affiliation(s)
- Adam A. J. Al‐Diwani
- Department of PsychiatryWarneford HospitalUniversity of OxfordOxfordUK
- Autoimmune Neurology GroupNuffield Department of Clinical NeurosciencesJohn Radcliffe HospitalUniversity of OxfordOxfordUK
| | - Thomas A. Pollak
- Department of Psychosis StudiesInstitute of Psychiatry, Psychology and NeuroscienceKing's Health PartnersLondonUK
| | - Sarosh R. Irani
- Autoimmune Neurology GroupNuffield Department of Clinical NeurosciencesJohn Radcliffe HospitalUniversity of OxfordOxfordUK
| | - Belinda R. Lennox
- Department of PsychiatryWarneford HospitalUniversity of OxfordOxfordUK
| |
Collapse
|
85
|
Kelly JR, Minuto C, Cryan JF, Clarke G, Dinan TG. Cross Talk: The Microbiota and Neurodevelopmental Disorders. Front Neurosci 2017; 11:490. [PMID: 28966571 PMCID: PMC5605633 DOI: 10.3389/fnins.2017.00490] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 08/17/2017] [Indexed: 12/11/2022] Open
Abstract
Humans evolved within a microbial ecosystem resulting in an interlinked physiology. The gut microbiota can signal to the brain via the immune system, the vagus nerve or other host-microbe interactions facilitated by gut hormones, regulation of tryptophan metabolism and microbial metabolites such as short chain fatty acids (SCFA), to influence brain development, function and behavior. Emerging evidence suggests that the gut microbiota may play a role in shaping cognitive networks encompassing emotional and social domains in neurodevelopmental disorders. Drawing upon pre-clinical and clinical evidence, we review the potential role of the gut microbiota in the origins and development of social and emotional domains related to Autism spectrum disorders (ASD) and schizophrenia. Small preliminary clinical studies have demonstrated gut microbiota alterations in both ASD and schizophrenia compared to healthy controls. However, we await the further development of mechanistic insights, together with large scale longitudinal clinical trials, that encompass a systems level dimensional approach, to investigate whether promising pre-clinical and initial clinical findings lead to clinical relevance.
Collapse
Affiliation(s)
- John R Kelly
- Department of Psychiatry and Neurobehavioural Science, University College CorkCork, Ireland.,APC Microbiome Institute, University College CorkCork, Ireland
| | - Chiara Minuto
- Department of Psychiatry and Neurobehavioural Science, University College CorkCork, Ireland.,APC Microbiome Institute, University College CorkCork, Ireland
| | - John F Cryan
- APC Microbiome Institute, University College CorkCork, Ireland.,Department of Anatomy and Neuroscience, University College CorkCork, Ireland
| | - Gerard Clarke
- Department of Psychiatry and Neurobehavioural Science, University College CorkCork, Ireland.,APC Microbiome Institute, University College CorkCork, Ireland
| | - Timothy G Dinan
- Department of Psychiatry and Neurobehavioural Science, University College CorkCork, Ireland.,APC Microbiome Institute, University College CorkCork, Ireland
| |
Collapse
|
86
|
Lambrichts S, Van Oudenhove L, Sienaert P. Antibiotics and mania: A systematic review. J Affect Disord 2017; 219:149-156. [PMID: 28550767 DOI: 10.1016/j.jad.2017.05.029] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 05/09/2017] [Accepted: 05/19/2017] [Indexed: 11/25/2022]
Abstract
OBJECTIVES Mania can occur secondary to a medical condition and can be elicited by various pharmacological treatments, both in patients with or without a history of affective disorder. Antibiotic-induced mania or antibiomania is suggested to be a rare phenomenon. We reviewed the literature in order to collect published reports of antibiomania and to summarize new insights about its mechanism and management. METHODS We performed a MEDLINE-search and used manual cross-referencing for reports of antibiotic-induced mania and included cases in which a (hypo)manic episode was diagnosed in close temporal relationship with the prescription of an antibiotic. RESULTS 47 cases were published. Patients' ages ranged from 3 to 77 years (mean 40). Two-thirds of the cases were male. Twelve different anti-bacterial agents were implicated, with antitubercular agents, macrolides and quinolones being the most common causative groups. CONCLUSIONS Antibiotic treatment can be associated with (hypo)mania. The paucity of reported cases precludes statements regarding incidence or antibiotic-specific warnings. In the event of an antibiotic-induced mania, the suspicious drug should be discontinued and manic symptoms can be treated lege artis. The pathophysiological mechanism of antibiomania remains elusive.
Collapse
Affiliation(s)
- Simon Lambrichts
- KU Leuven, University Psychiatric Center KU Leuven, Herestraat 49, 3000 Leuven / Leuvensesteenweg 517, 3070 Kortenberg, Belgium
| | - Lukas Van Oudenhove
- Laboratory for Brain-Gut Axis Studies (LABGAS), Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Clinical and Experimental Medicine, University of Leuven, Leuven, Belgium; Consultation-Liaison Psychiatry, University Psychiatric Center KU Leuven, Campus Gasthuisberg, University of Leuven, Leuven, Belgium
| | - Pascal Sienaert
- KU Leuven, University Psychiatric Center KU Leuven, Herestraat 49, 3000 Leuven / Leuvensesteenweg 517, 3070 Kortenberg, Belgium; Academic Center for ECT and Neuromodulation (AcCENT), University Psychiatric Center KU Leuven, Leuvensesteenweg 517, 3070 Kortenberg, Belgium.
| |
Collapse
|
87
|
Kannan G, Gressitt KL, Yang S, Stallings CR, Katsafanas E, Schweinfurth LA, Savage CLG, Adamos MB, Sweeney KM, Origoni AE, Khushalani S, Bahn S, Leweke FM, Dickerson FB, Yolken RH, Pletnikov MV, Severance EG. Pathogen-mediated NMDA receptor autoimmunity and cellular barrier dysfunction in schizophrenia. Transl Psychiatry 2017; 7:e1186. [PMID: 28763062 PMCID: PMC5611729 DOI: 10.1038/tp.2017.162] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 06/09/2017] [Accepted: 06/13/2017] [Indexed: 12/15/2022] Open
Abstract
Autoantibodies that bind the N-methyl-D-aspartate receptor (NMDAR) may underlie glutamate receptor hypofunction and related cognitive impairment found in schizophrenia. Exposure to neurotropic pathogens can foster an autoimmune-prone environment and drive systemic inflammation leading to endothelial barrier defects. In mouse model cohorts, we demonstrate that infection with the protozoan parasite, Toxoplasma gondii, caused sustained elevations of IgG class antibodies to the NMDAR in conjunction with compromised blood-gut and blood-brain barriers. In human cohorts, NMDAR IgG and markers of barrier permeability were significantly associated with T. gondii exposure in schizophrenia compared with controls and independently of antipsychotic medication. Combined T. gondii and NMDAR antibody seropositivity in schizophrenia resulted in higher degrees of cognitive impairment as measured by tests of delayed memory. These data underscore the necessity of disentangling the heterogeneous pathophysiology of schizophrenia so that relevant subsets eligible for NMDAR-related treatment can be identified. Our data aid to reconcile conflicting reports regarding a role of pathological NMDAR autoantibodies in this disorder.
Collapse
Affiliation(s)
- G Kannan
- Department of Psychiatry, Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - K L Gressitt
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - S Yang
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - C R Stallings
- Sheppard Pratt Health System, Stanley Research Program, Baltimore, MD, USA
| | - E Katsafanas
- Sheppard Pratt Health System, Stanley Research Program, Baltimore, MD, USA
| | - L A Schweinfurth
- Sheppard Pratt Health System, Stanley Research Program, Baltimore, MD, USA
| | - C L G Savage
- Sheppard Pratt Health System, Stanley Research Program, Baltimore, MD, USA
| | - M B Adamos
- Sheppard Pratt Health System, Stanley Research Program, Baltimore, MD, USA
| | - K M Sweeney
- Sheppard Pratt Health System, Stanley Research Program, Baltimore, MD, USA
| | - A E Origoni
- Sheppard Pratt Health System, Stanley Research Program, Baltimore, MD, USA
| | - S Khushalani
- Sheppard Pratt Health System, Stanley Research Program, Baltimore, MD, USA
| | - S Bahn
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - F M Leweke
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Brain and Mind Centre, University of Sydney, Camperdown, NSW, Australia
| | - F B Dickerson
- Sheppard Pratt Health System, Stanley Research Program, Baltimore, MD, USA
| | - R H Yolken
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - M V Pletnikov
- Department of Psychiatry, Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Molecular Immunology and Microbiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - E G Severance
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
88
|
Uranova NA, Bonartsev PD, Androsova LV, Rakhmanova VI, Kaleda VG. Impaired monocyte activation in schizophrenia: ultrastructural abnormalities and increased IL-1β production. Eur Arch Psychiatry Clin Neurosci 2017; 267:417-426. [PMID: 28314932 DOI: 10.1007/s00406-017-0782-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 03/06/2017] [Indexed: 12/25/2022]
Abstract
Monocyte activation is consistently reported in patients with schizophrenia (SZ). We aimed to study the ultrastructure of monocytes and monocyte production of IL-1β in drug-free patients with SZ and controls. Monocytes from young (18-30 y.o.) healthy and SZ men in relapse were studied. Electron microscopy and morphometry were applied to estimate areas of monocytes, volume density (Vv), areas, and number of organelles. The production IL-1β by monocytes was estimated by the ELISA method. Group differences were examined using ANCOVA. Pearson's correlation coefficients were used to examine the effects of possible confounding variables. Correlation analyses were applied to detect the relationships between the parameters of monocytes measured and between the parameters measured and the IL-1β production. Area of nucleolus, Vv and area of mitochondria and lysosomes, and the number of lysosomes were significantly increased in patients as compared to controls. Area of mitochondria was correlated significantly with Vv and area of lysosomes, and the number of lysosomes was significantly correlated with area of monocyte and Vv of vacuoles only in the control group. The production of IL-1β by monocytes was higher in patients than in controls (p = 0.01) and was correlated with Vv of lysosomes (r = 0.68, p = 0.04) and area of lysosomes (r = 0.78, p = 0.013). The data provide new evidence for over activation of monocytes in SZ and disturbed metabolic relationships between lysosomes, mitochondria, and vacuoles.
Collapse
Affiliation(s)
- Natalya A Uranova
- Laboratory of Clinical Neuropathology, Mental Health Research Centre, Zagorodnoe Shosse 2, Moscow, 117152, Russia.
| | - P D Bonartsev
- Laboratory of Clinical Neuropathology, Mental Health Research Centre, Zagorodnoe Shosse 2, Moscow, 117152, Russia
| | - L V Androsova
- Laboratory of Neuroimmunology, Mental Health Research Centre, Zagorodnoe Shosse 2, Moscow, 117152, Russia
| | - V I Rakhmanova
- Laboratory of Clinical Neuropathology, Mental Health Research Centre, Zagorodnoe Shosse 2, Moscow, 117152, Russia
| | - V G Kaleda
- Department of Endogenous Psychoses, Mental Health Research Centre, Zagorodnoe Shosse 2, Moscow, 117152, Russia
| |
Collapse
|
89
|
Central Nervous System Infection with Borna Disease Virus Causes Kynurenine Pathway Dysregulation and Neurotoxic Quinolinic Acid Production. J Virol 2017; 91:JVI.00673-17. [PMID: 28446679 PMCID: PMC5487560 DOI: 10.1128/jvi.00673-17] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 04/19/2017] [Indexed: 12/27/2022] Open
Abstract
Central nervous system infection of neonatal and adult rats with Borna disease virus (BDV) results in neuronal destruction and behavioral abnormalities with differential immune-mediated involvement. Neuroactive metabolites generated from the kynurenine pathway of tryptophan degradation have been implicated in several human neurodegenerative disorders. Here, we report that brain expression of key enzymes in the kynurenine pathway are significantly, but differentially, altered in neonatal and adult rats with BDV infection. Gene expression analysis of rat brains following neonatal infection showed increased expression of kynurenine amino transferase II (KATII) and kynurenine-3-monooxygenase (KMO) enzymes. Additionally, indoleamine 2,3-dioxygenase (IDO) expression was only modestly increased in a brain region- and time-dependent manner in neonatally infected rats; however, its expression was highly increased in adult infected rats. The most dramatic impact on gene expression was seen for KMO, whose activity promotes the production of neurotoxic quinolinic acid. KMO expression was persistently elevated in brain regions of both newborn and adult BDV-infected rats, with increases reaching up to 86-fold. KMO protein levels were increased in neonatally infected rats and colocalized with neurons, the primary target cells of BDV infection. Furthermore, quinolinic acid was elevated in neonatally infected rat brains. We further demonstrate increased expression of KATII and KMO, but not IDO, in vitro in BDV-infected C6 astroglioma cells. Our results suggest that BDV directly impacts the kynurenine pathway, an effect that may be exacerbated by inflammatory responses in immunocompetent hosts. Thus, experimental models of BDV infection may provide new tools for discriminating virus-mediated from immune-mediated impacts on the kynurenine pathway and their relative contribution to neurodegeneration.IMPORTANCE BDV causes persistent, noncytopathic infection in vitro yet still elicits widespread neurodegeneration of infected neurons in both immunoincompetent and immunocompetent hosts. Here, we show that BDV infection induces expression of key enzymes of the kynurenine pathway in brains of newborn and adult infected rats and cultured astroglioma cells, shunting tryptophan degradation toward the production of neurotoxic quinolinic acid. Thus, our findings newly implicate this metabolic pathway in BDV-induced neurodegeneration. Given the importance of the kynurenine pathway in a wide range of human infections and neurodegenerative and neuropsychiatric disorders, animal models of BDV infection may serve as important tools for contrasting direct viral and indirect antiviral immune-mediated impacts on kynurenine pathway dysregulation and the ensuing neurodevelopmental and neuropathological consequences.
Collapse
|
90
|
Dickerson F, Wilcox HC, Adamos M, Katsafanas E, Khushalani S, Origoni A, Savage C, Schweinfurth L, Stallings C, Sweeney K, Yolken R. Suicide attempts and markers of immune response in individuals with serious mental illness. J Psychiatr Res 2017; 87:37-43. [PMID: 27988332 DOI: 10.1016/j.jpsychires.2016.11.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 11/02/2016] [Accepted: 11/07/2016] [Indexed: 12/11/2022]
Abstract
Previous studies have identified elevations in antibodies to Toxoplasma gondii in individuals with a history of suicide attempts but studies have not measured the association between suicide attempts and a panel of antibody markers. We assessed 162 patients receiving treatment for schizophrenia, bipolar disorder, or major depression on the Columbia Suicide Severity Rating Scale for suicide attempt history and other clinical measures. All participants had a blood sample drawn from which were measured antibodies to Toxoplasma gondii and other neurotropic infectious agents. A total of 72 (44%) of participants had a lifetime suicide attempt; these individuals had elevated levels of IgM class antibodies to Toxoplasma gondii and Cytomegalovirus (CMV). We also found an association between the levels of these antibodies and the number of suicide attempts. There was a particularly strong odds of a suicide attempt history in individuals who had elevated levels of IgM antibodies to both Toxoplasma gondii and to CMV suggesting an additive risk associated with the antibodies. These findings remained significant when adjusting for current cigarette smoking and history of drug/alcohol use which were also associated with suicide attempts. We did not find an association between a suicide attempt history and IgG class antibodies to Toxoplasma gondii, CMV, or IgM or IgG antibodies to the Epstein Barr Virus or other antigens tested. The identification of blood-based antibody markers should provide for more personalized methods for the assessment and treatment, and ultimately prevention, of suicide attempts in individuals with serious mental illnesses.
Collapse
Affiliation(s)
- Faith Dickerson
- Stanley Research Program, Sheppard Pratt Health System, Baltimore, MD, USA.
| | - Holly C Wilcox
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Maria Adamos
- Stanley Research Program, Sheppard Pratt Health System, Baltimore, MD, USA
| | - Emily Katsafanas
- Stanley Research Program, Sheppard Pratt Health System, Baltimore, MD, USA
| | - Sunil Khushalani
- Stanley Research Program, Sheppard Pratt Health System, Baltimore, MD, USA
| | - Andrea Origoni
- Stanley Research Program, Sheppard Pratt Health System, Baltimore, MD, USA
| | - Christina Savage
- Stanley Research Program, Sheppard Pratt Health System, Baltimore, MD, USA
| | - Lucy Schweinfurth
- Stanley Research Program, Sheppard Pratt Health System, Baltimore, MD, USA
| | - Cassie Stallings
- Stanley Research Program, Sheppard Pratt Health System, Baltimore, MD, USA
| | - Kevin Sweeney
- Stanley Research Program, Sheppard Pratt Health System, Baltimore, MD, USA
| | - Robert Yolken
- Stanley Neurovirology Laboratory, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
91
|
Perkovic MN, Erjavec GN, Strac DS, Uzun S, Kozumplik O, Pivac N. Theranostic Biomarkers for Schizophrenia. Int J Mol Sci 2017; 18:E733. [PMID: 28358316 PMCID: PMC5412319 DOI: 10.3390/ijms18040733] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 03/23/2017] [Accepted: 03/27/2017] [Indexed: 12/14/2022] Open
Abstract
Schizophrenia is a highly heritable, chronic, severe, disabling neurodevelopmental brain disorder with a heterogeneous genetic and neurobiological background, which is still poorly understood. To allow better diagnostic procedures and therapeutic strategies in schizophrenia patients, use of easy accessible biomarkers is suggested. The most frequently used biomarkers in schizophrenia are those associated with the neuroimmune and neuroendocrine system, metabolism, different neurotransmitter systems and neurotrophic factors. However, there are still no validated and reliable biomarkers in clinical use for schizophrenia. This review will address potential biomarkers in schizophrenia. It will discuss biomarkers in schizophrenia and propose the use of specific blood-based panels that will include a set of markers associated with immune processes, metabolic disorders, and neuroendocrine/neurotrophin/neurotransmitter alterations. The combination of different markers, or complex multi-marker panels, might help in the discrimination of patients with different underlying pathologies and in the better classification of the more homogenous groups. Therefore, the development of the diagnostic, prognostic and theranostic biomarkers is an urgent and an unmet need in psychiatry, with the aim of improving diagnosis, therapy monitoring, prediction of treatment outcome and focus on the personal medicine approach in order to improve the quality of life in patients with schizophrenia and decrease health costs worldwide.
Collapse
Affiliation(s)
| | | | - Dubravka Svob Strac
- Rudjer Boskovic Institute, Division of Molecular Medicine, 10000 Zagreb, Croatia.
| | - Suzana Uzun
- Clinic for Psychiatry Vrapce, 10090 Zagreb, Croatia.
| | | | - Nela Pivac
- Rudjer Boskovic Institute, Division of Molecular Medicine, 10000 Zagreb, Croatia.
| |
Collapse
|
92
|
Bipolar Disorder With Psychotic Features and Ocular Toxoplasmosis: A Possible Pathogenetic Role of the Parasite? J Nerv Ment Dis 2017; 205:192-195. [PMID: 27741079 DOI: 10.1097/nmd.0000000000000496] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Recent evidence suggests the involvement of Toxoplasma gondii infection in the emergence of psychotic and affective disorders. In this report, we describe the case of a young Brazilian woman affected by recurrent ocular toxoplasmosis and presenting with a manic episode with psychotic features in the context of a diagnosis of Bipolar Disorder (BD), type I. We observed a relationship between ocular manifestations and the clinical course of bipolar illness, confirmed by molecular analyses (nested-PCR), as well as by the high level of T. gondii specific IgG. This case report is the first showing the presence of circulating parasite DNA at the time of occurrence of psychiatric symptoms, thus providing further support for a possible role of the parasite in the pathogenesis of some cases of BD.
Collapse
|
93
|
Del Grande C, Galli L, Schiavi E, Dell'Osso L, Bruschi F. Is Toxoplasma gondii a Trigger of Bipolar Disorder? Pathogens 2017; 6:pathogens6010003. [PMID: 28075410 PMCID: PMC5371891 DOI: 10.3390/pathogens6010003] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 12/16/2016] [Accepted: 01/04/2017] [Indexed: 11/16/2022] Open
Abstract
Toxoplasma gondii, a ubiquitous intracellular parasite, has a strong tropism for the brain tissue, where it forms intracellular cysts within the neurons and glial cells, establishing a chronic infection. Although latent toxoplasmosis is generally assumed to be asymptomatic in immunocompetent individuals, it is now clear that it can induce behavioral manipulations in mice and infected humans. Moreover, a strong relation has emerged in recent years between toxoplasmosis and psychiatric disorders. The link between T. gondii and schizophrenia has been the most widely documented; however, a significant association with bipolar disorder (BD) and suicidal/aggressive behaviors has also been detected. T. gondii may play a role in the etiopathogenesis of psychiatric disorders affecting neurotransmitters, especially dopamine, that are implicated in the emergence of psychosis and behavioral Toxoplasma-induced abnormalities, and inducing brain inflammation by the direct stimulation of inflammatory cytokines in the central nervous system. Besides this, there is increasing evidence for a prominent role of immune dysregulation in psychosis and BD. The aim of this review is to describe recent evidence suggesting a link between Toxoplasma gondii and BD, focusing on the interaction between immune responses and this infectious agent in the etiopathogenesis of psychiatric symptoms.
Collapse
Affiliation(s)
- Claudia Del Grande
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, Via Roma 67, 56127 Pisa, Italy.
| | - Luca Galli
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy.
| | - Elisa Schiavi
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, Via Roma 67, 56127 Pisa, Italy.
| | - Liliana Dell'Osso
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, Via Roma 67, 56127 Pisa, Italy.
| | - Fabrizio Bruschi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy.
| |
Collapse
|
94
|
The kynurenine pathway and parasitic infections that affect CNS function. Neuropharmacology 2017; 112:389-398. [DOI: 10.1016/j.neuropharm.2016.02.029] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 02/19/2016] [Accepted: 02/23/2016] [Indexed: 12/14/2022]
|
95
|
The kynurenine pathway in schizophrenia and bipolar disorder. Neuropharmacology 2017; 112:297-306. [DOI: 10.1016/j.neuropharm.2016.05.020] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 05/26/2016] [Accepted: 05/27/2016] [Indexed: 11/20/2022]
|
96
|
Schwarcz R, Stone TW. The kynurenine pathway and the brain: Challenges, controversies and promises. Neuropharmacology 2017; 112:237-247. [PMID: 27511838 PMCID: PMC5803785 DOI: 10.1016/j.neuropharm.2016.08.003] [Citation(s) in RCA: 279] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 07/29/2016] [Accepted: 08/05/2016] [Indexed: 12/29/2022]
Abstract
Research on the neurobiology of the kynurenine pathway has suffered years of relative obscurity because tryptophan degradation, and its involvement in both physiology and major brain diseases, was viewed almost exclusively through the lens of the well-established metabolite serotonin. With increasing recognition that kynurenine and its metabolites can affect and even control a variety of classic neurotransmitter systems directly and indirectly, interest is expanding rapidly. Moreover, kynurenine pathway metabolism itself is modulated in conditions such as infection and stress, which are known to induce major changes in well-being and behaviour, so that kynurenines may be instrumental in the etiology of psychiatric and neurological disorders. It is therefore likely that the near future will not only witness the discovery of additional physiological and pathological roles for brain kynurenines, but also ever-increasing interest in drug development based on these roles. In particular, targeting the kynurenine pathway with new specific agents may make it possible to prevent disease by appropriate pharmacological or genetic manipulations. The following overview focuses on areas of kynurenine research which are either controversial, of major potential therapeutic interest, or just beginning to receive the degree of attention which will clarify their relevance to neurobiology and medicine. It also highlights technical issues so that investigators entering the field, and new research initiatives, are not misdirected by inappropriate experimental approaches or incorrect interpretations at this time of skyrocketing interest in the subject matter. This article is part of the Special Issue entitled 'The Kynurenine Pathway in Health and Disease'.
Collapse
Affiliation(s)
- Robert Schwarcz
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Trevor W Stone
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
97
|
Ajdacic-Gross V, Aleksandrowicz A, Rodgers S, Mutsch M, Tesic A, Müller M, Kawohl W, Rössler W, Seifritz E, Castelao E, Strippoli MPF, Vandeleur C, von Känel R, Paolicelli R, Landolt MA, Witthauer C, Lieb R, Preisig M. Infectious, atopic and inflammatory diseases, childhood adversities and familial aggregation are independently associated with the risk for mental disorders: Results from a large Swiss epidemiological study. World J Psychiatry 2016; 6:419-430. [PMID: 28078206 PMCID: PMC5183994 DOI: 10.5498/wjp.v6.i4.419] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 09/08/2016] [Accepted: 10/09/2016] [Indexed: 02/05/2023] Open
Abstract
AIM To examine the associations between mental disorders and infectious, atopic, inflammatory diseases while adjusting for other risk factors.
METHODS We used data from PsyCoLaus, a large Swiss Population Cohort Study (n = 3720; age range 35-66). Lifetime diagnoses of mental disorders were grouped into the following categories: Neurodevelopmental, anxiety (early and late onset), mood and substance disorders. They were regressed on infectious, atopic and other inflammatory diseases adjusting for sex, educational level, familial aggregation, childhood adversities and traumatic experiences in childhood. A multivariate logistic regression was applied to each group of disorders. In a complementary analysis interactions with sex were introduced via nested effects.
RESULTS Associations with infectious, atopic and other chronic inflammatory diseases were observable together with consistent effects of childhood adversities and familial aggregation, and less consistent effects of trauma in each group of mental disorders. Streptococcal infections were associated with neurodevelopmental disorders (men), and measles/mumps/rubella-infections with early and late anxiety disorders (women). Gastric inflammatory diseases took effect in mood disorders (both sexes) and in early disorders (men). Similarly, irritable bowel syndrome was prominent in a sex-specific way in mood disorders in women, and, moreover, was associated with early and late anxiety disorders. Atopic diseases were associated with late anxiety disorders. Acne (associations with mood disorders in men) and psoriasis (associations with early anxiety disorders in men and mood disorders in women) contributed sex-specific results. Urinary tract infections were associated with mood disorders and, in addition, in a sex-specific way with late anxiety disorders (men), and neurodevelopmental and early anxiety disorders (women).
CONCLUSION Infectious, atopic and inflammatory diseases are important risk factors for all groups of mental disorders. The sexual dimorphism of the associations is pronounced.
Collapse
|
98
|
Aripiprazole inhibits polyI:C-induced microglial activation possibly via TRPM7. Schizophr Res 2016; 178:35-43. [PMID: 27614570 DOI: 10.1016/j.schres.2016.08.022] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 08/20/2016] [Accepted: 08/20/2016] [Indexed: 12/13/2022]
Abstract
Viral infections during fetal and adolescent periods, as well as during the course of schizophrenia itself have been linked to the onset and/or relapse of a psychosis. We previously reported that the unique antipsychotic aripiprazole, a partial D2 agonist, inhibits the release of tumor necrosis factor (TNF)-α from interferon-γ-activated rodent microglial cells. Polyinosinic-polycytidylic acid (polyI:C) has recently been used as a standard model of viral infections, and recent in vitro studies have shown that microglia are activated by polyI:C. Aripiprazole has been reported to ameliorate behavioral abnormalities in polyI:C-induced mice. To clarify the anti-inflammatory properties of aripiprazole, we investigated the effects of aripiprazole on polyI:C-induced microglial activation in a cellular model of murine microglial cells and possible surrogate cells for human microglia. PolyI:C treatment of murine microglial cells activated the production of TNF-α and enhanced the p38 mitogen-activated protein kinase (MAPK) pathway, whereas aripiprazole inhibited these responses. In addition, polyI:C treatment of possible surrogate cells for human microglia markedly increased TNF-α mRNA expression in cells from three healthy volunteers. Aripiprazole inhibited this increase in cells from two individuals. PolyI:C consistently increased intracellular Ca2+ concentration ([Ca2+]i) in murine microglial cells by influx of extracellular Ca2+. We demonstrated that transient receptor potential in melastatin 7 (TRPM7) channels contributed to this polyI:C-induced increase in [Ca2+]i. Taken together, these data suggest that aripiprazole may be therapeutic for schizophrenia by reducing microglial inflammatory reactions, and TRPM7 may be a novel therapeutic target for schizophrenia. Further studies are needed to validate these findings.
Collapse
|
99
|
Neuropathology of mood disorders: do we see the stigmata of inflammation? Transl Psychiatry 2016; 6:e946. [PMID: 27824355 PMCID: PMC5314124 DOI: 10.1038/tp.2016.212] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 07/08/2016] [Accepted: 08/30/2016] [Indexed: 12/15/2022] Open
Abstract
A proportion of cases with mood disorders have elevated inflammatory markers in the blood that conceivably may result from stress, infection and/or autoimmunity. However, it is not yet clear whether depression is a neuroinflammatory disease. Multiple histopathological and molecular abnormalities have been found postmortem but the etiology of these abnormalities is unknown. Here, we take an immunological perspective of this literature. Increases in activated microglia or perivascular macrophages in suicide victims have been reported in the parenchyma. In contrast, astrocytic markers generally are downregulated in mood disorders. Impairment of astrocytic function likely compromises the reuptake of glutamate potentially leading to excitotoxicity. Inflammatory cytokines and microglia/macrophage-derived quinolinic acid (QA) downregulate the excitatory amino acid transporters responsible for this reuptake, while QA has the additional effect of inhibiting astroglial glutamine synthetase, which converts glutamate to glutamine. Given that oligodendroglia are particularly vulnerable to inflammation, it is noteworthy that reductions in numbers or density of oligodendrocyte cells are one of the most prominent findings in depression. Structural and/or functional changes to GABAergic interneurons also are salient in postmortem brain samples, and may conceivably be related to early inflammatory insults. Although the postmortem data are consistent with a neuroimmune etiology in a subgroup of depressed individuals, we do not argue that all depression-associated abnormalities are reflective of a neuroinflammatory process or even that all immunological activity in the brain is deleterious. Rather, we highlight the pervasive role of immune signaling pathways in brain function and provide an alternative perspective on the current postmortem literature.
Collapse
|
100
|
Benros ME, Trabjerg BB, Meier S, Mattheisen M, Mortensen PB, Mors O, Børglum AD, Hougaard DM, Nørgaard-Pedersen B, Nordentoft M, Agerbo E. Influence of Polygenic Risk Scores on the Association Between Infections and Schizophrenia. Biol Psychiatry 2016; 80:609-16. [PMID: 27364036 DOI: 10.1016/j.biopsych.2016.04.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 03/18/2016] [Accepted: 04/12/2016] [Indexed: 12/30/2022]
Abstract
BACKGROUND Several studies have suggested an important role of infections in the etiology of schizophrenia; however, shared genetic liability toward infections and schizophrenia could influence the association. We therefore investigated the possible effect of polygenic risk scores (PRSs) for schizophrenia on the association between infections and the risk of schizophrenia. METHODS We conducted a nested case-control study on a Danish population-based sample born after 1981 comprising of 1692 cases diagnosed with schizophrenia between 1994 and 2008 and 1724 matched controls. All individuals were linked utilizing nationwide population-based registers with virtually complete registration of all hospital contacts for infections. PRSs were calculated using discovery effect size estimates weights from an independent meta-analysis (34,600 cases and 45,968 control individuals). RESULTS A prior hospital contact with infection had occurred in 41% of the individuals with schizophrenia and increased the incidence rate ratio (IRR) of schizophrenia by 1.43 (95% confidence interval [CI] = 1.22-1.67). Adding PRS, which was robustly associated with schizophrenia (by an IRR of 1.46 [95% CI = 1.34-1.60] per standard deviation of the score), did not alter the association with infections and the increased risk of schizophrenia remained (IRR = 1.41; 95% CI = 1.20-1.66). Furthermore, there were no interactions between PRS and infections on the risk of developing schizophrenia (p = .554). Neither did PRS affect the risk of acquiring infections among patients with schizophrenia (odds ratio = 1.00; 95% CI = 0.89-1.12) nor among controls (odds ratio = 1.09; 95% CI: 0.96-1.24). CONCLUSIONS PRS and a history of infections have independent effects on the risk for schizophrenia, and the common genetic risk measured by PRS did not account for the association with infection in this sample.
Collapse
Affiliation(s)
- Michael E Benros
- Mental Health Centre Copenhagen, University of Copenhagen, Faculty of Health Sciences, Copenhagen; National Centre for Register-Based Research, Aarhus University, Aarhus; The Lundbeck Foundation Initiative for Integrative Psychiatric Research iPSYCH, Copenhagen.
| | - Betina B Trabjerg
- National Centre for Register-Based Research, Aarhus University, Aarhus; The Lundbeck Foundation Initiative for Integrative Psychiatric Research iPSYCH, Copenhagen
| | - Sandra Meier
- National Centre for Register-Based Research, Aarhus University, Aarhus; The Lundbeck Foundation Initiative for Integrative Psychiatric Research iPSYCH, Copenhagen
| | - Manuel Mattheisen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research iPSYCH, Copenhagen; Department of Biomedicine and Centre for Integrative Sequencing (iSEQ), Aarhus University, Aarhus
| | - Preben B Mortensen
- National Centre for Register-Based Research, Aarhus University, Aarhus; The Lundbeck Foundation Initiative for Integrative Psychiatric Research iPSYCH, Copenhagen
| | - Ole Mors
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research iPSYCH, Copenhagen; Research Department P, Aarhus University Hospital, Risskov
| | - Anders D Børglum
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research iPSYCH, Copenhagen; Department of Biomedicine and Centre for Integrative Sequencing (iSEQ), Aarhus University, Aarhus
| | - David M Hougaard
- Danish Centre for Neonatal Screening, Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Bent Nørgaard-Pedersen
- Danish Centre for Neonatal Screening, Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Merete Nordentoft
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research iPSYCH, Copenhagen
| | - Esben Agerbo
- National Centre for Register-Based Research, Aarhus University, Aarhus; The Lundbeck Foundation Initiative for Integrative Psychiatric Research iPSYCH, Copenhagen; CIRRAU-Centre for Integrated Register-Based Research, Aarhus University, Aarhus
| |
Collapse
|