1
|
Hofsink N, Groenink L, Plösch T. The fetal programming effect of maternal immune activation (MIA) on the offspring's immune system. Semin Immunopathol 2024; 46:14. [PMID: 39212791 PMCID: PMC11364800 DOI: 10.1007/s00281-024-01023-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
The first 1000 days of life is a critical period of development in which adverse circumstances can have long-term consequences for the child's health. Maternal immune activation is associated with increased risk of neurodevelopmental disorders in the child. Aberrant immune responses have been reported in individuals with neurodevelopmental disorders. Moreover, lasting effects of maternal immune activation on the offspring's immune system have been reported. Taken together, this indicates that the effect of maternal immune activation is not limited to the central nervous system. Here, we explore the impact of maternal immune activation on the immune system of the offspring. We first describe the development of the immune system and provide an overview of reported alterations in the cytokine profiles, immune cell profiles, immune cell function, and immune induction in pre-clinical models. Additionally, we highlight recent research on the impact of maternal COVID-19 exposure on the neonatal immune system and the potential health consequences for the child. Our review shows that maternal immune activation alters the offspring's immune system under certain conditions, but the reported effects are conflicting and inconsistent. In general, epigenetic modifications are considered the mechanism for fetal programming. The available data was insufficient to identify specific pathways that may contribute to immune programming. As a consequence of the COVID-19 pandemic, more research now focuses on the possible health effects of maternal immune activation on the offspring. Future research addressing the offspring's immune response to maternal immune activation can elucidate specific pathways that contribute to fetal immune programming and the long-term health effects for the offspring.
Collapse
Affiliation(s)
- Naomi Hofsink
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - Lucianne Groenink
- Department of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Torsten Plösch
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Carl von Ossietzky Universität Oldenburg School VI - School of Medicine and Health Sciences, Department of Paediatrics, Section of Neonatology, and Research Centre Neurosensory Science, Oldenburg, Germany
| |
Collapse
|
2
|
Meza-Sosa KF, Valle-Garcia D, González-Conchillos H, Blanco-Ayala T, Salazar A, Flores I, Gómez-Manzo S, González Esquivel DF, Pérez de la Cruz G, Pineda B, Pérez de la Cruz V. Molecular Mimicry between Toxoplasma gondii B-Cell Epitopes and Neurodevelopmental Proteins: An Immunoinformatic Approach. Biomolecules 2024; 14:933. [PMID: 39199321 PMCID: PMC11352964 DOI: 10.3390/biom14080933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/27/2024] [Accepted: 07/28/2024] [Indexed: 09/01/2024] Open
Abstract
Epidemiological studies and meta-analyses have shown a strong association between high seroprevalence of Toxoplasma gondii (T. gondii) and schizophrenia. Schizophrenic patients showed higher levels of anti-Toxoplasma immunoglobulins M and G (IgM and IgG) when compared to healthy controls. Previously, in a rat model, we demonstrated that the progeny of mothers immunized with T. gondii lysates before gestation had behavioral and social impairments during adulthood. Therefore, we suggested that T. gondii infection can trigger autoreactivity by molecularly mimicking host brain proteins. Here, we aimed to identify the occurrence of antigenic mimicry between T. gondii epitopes and host brain proteins. Using a bioinformatic approach, we predicted T. gondii RH-88 B cell epitopes and compared them to human cell-surface proteins involved in brain development and differentiation (BrainS). Five different algorithms for B-cell-epitope prediction were used and compared, resulting in 8584 T. gondii epitopes. We then compared T. gondii predicted epitopes to BrainS proteins by local sequence alignments using BLASTP. T. gondii immunogenic epitopes significantly overlapped with 42 BrainS proteins. Among these overlapping proteins essential for brain development and differentiation, we identified HSP90 and NOTCH receptors as the proteins most likely to be targeted by the maternally generated pathogenic antibodies due to their topological overlap at the extracellular region of their sequence. This analysis highlights the relevance of pregestational clinical surveillance and screening for potential pathogenic anti-T. gondii antibodies. It also identifies potential targets for the design of vaccines that could prevent behavioral and cognitive impairments associated with pre-gestational T. gondii exposure.
Collapse
Affiliation(s)
- Karla F. Meza-Sosa
- Neurochemistry and Behavior Laboratory, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico; (K.F.M.-S.); (T.B.-A.); (D.F.G.E.)
| | - David Valle-Garcia
- Neuroimmunology Department, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico; (D.V.-G.); (H.G.-C.); (A.S.); (I.F.)
| | - Hugo González-Conchillos
- Neuroimmunology Department, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico; (D.V.-G.); (H.G.-C.); (A.S.); (I.F.)
| | - Tonali Blanco-Ayala
- Neurochemistry and Behavior Laboratory, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico; (K.F.M.-S.); (T.B.-A.); (D.F.G.E.)
| | - Alelí Salazar
- Neuroimmunology Department, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico; (D.V.-G.); (H.G.-C.); (A.S.); (I.F.)
| | - Itamar Flores
- Neuroimmunology Department, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico; (D.V.-G.); (H.G.-C.); (A.S.); (I.F.)
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Manuel Carpio, Plutarco Elías Calles, Miguel Hidalgo, Mexico City 11350, Mexico
| | - Saúl Gómez-Manzo
- Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico;
| | - Dinora Fabiola González Esquivel
- Neurochemistry and Behavior Laboratory, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico; (K.F.M.-S.); (T.B.-A.); (D.F.G.E.)
| | - Gonzalo Pérez de la Cruz
- Department of Mathematics, Faculty of Sciences, Universidad Nacional Autónoma de Mexico (UNAM), Mexico City 04510, Mexico;
| | - Benjamín Pineda
- Neuroimmunology Department, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico; (D.V.-G.); (H.G.-C.); (A.S.); (I.F.)
| | - Verónica Pérez de la Cruz
- Neurochemistry and Behavior Laboratory, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico; (K.F.M.-S.); (T.B.-A.); (D.F.G.E.)
| |
Collapse
|
3
|
Zajkowska I, Niczyporuk P, Urbaniak A, Tomaszek N, Modzelewski S, Waszkiewicz N. Investigating the Impacts of Diet, Supplementation, Microbiota, Gut-Brain Axis on Schizophrenia: A Narrative Review. Nutrients 2024; 16:2228. [PMID: 39064675 PMCID: PMC11279812 DOI: 10.3390/nu16142228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/06/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Schizophrenia is a disease with a complex etiology that significantly impairs the functioning of patients. In recent years, there has been increasing focus on the importance of the gut microbiota in the context of the gut-brain axis. In our study, we analyzed data on the gut-brain axis in relation to schizophrenia, as well as the impacts of eating habits, the use of various supplements, and diets on schizophrenia. Additionally, the study investigated the impact of antipsychotics on the development of metabolic disorders, such as diabetes, dyslipidemia, and obesity. There may be significant clinical benefits to be gained from therapies supported by supplements such as omega-3 fatty acids, B vitamins, and probiotics. The results suggest the need for a holistic approach to the treatment of schizophrenia, incorporating both drug therapy and dietary interventions.
Collapse
Affiliation(s)
| | | | | | | | - Stefan Modzelewski
- Department of Psychiatry, Medical University of Bialystok, pl. Wołodyjowskiego 2, 15-272 Białystok, Poland; (I.Z.); (N.W.)
| | | |
Collapse
|
4
|
Gupta S, Dinesh S, Sharma S. Bridging the Mind and Gut: Uncovering the Intricacies of Neurotransmitters, Neuropeptides, and their Influence on Neuropsychiatric Disorders. Cent Nerv Syst Agents Med Chem 2024; 24:2-21. [PMID: 38265387 DOI: 10.2174/0118715249271548231115071021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/31/2023] [Accepted: 10/04/2023] [Indexed: 01/25/2024]
Abstract
BACKGROUND The gut-brain axis (GBA) is a bidirectional signaling channel that facilitates communication between the gastrointestinal tract and the brain. Recent research on the gut-brain axis demonstrates that this connection enables the brain to influence gut function, which in turn influences the brain and its cognitive functioning. It is well established that malfunctioning of this axis adversely affects both systems' ability to operate effectively. OBJECTIVE Dysfunctions in the GBA have been associated with disorders of gut motility and permeability, intestinal inflammation, indigestion, constipation, diarrhea, IBS, and IBD, as well as neuropsychiatric and neurodegenerative disorders like depression, anxiety, schizophrenia, autism, Alzheimer's, and Parkinson's disease. Multiple research initiatives have shown that the gut microbiota, in particular, plays a crucial role in the GBA by participating in the regulation of a number of key neurochemicals that are known to have significant effects on the mental and physical well-being of an individual. METHODS Several studies have investigated the relationship between neuropsychiatric disorders and imbalances or disturbances in the metabolism of neurochemicals, often leading to concomitant gastrointestinal issues and modifications in gut flora composition. The interaction between neurological diseases and gut microbiota has been a focal point within this research. The novel therapeutic interventions in neuropsychiatric conditions involving interventions such as probiotics, prebiotics, and dietary modifications are outlined in this review. RESULTS The findings of multiple studies carried out on mice show that modulating and monitoring gut microbiota can help treat symptoms of such diseases, which raises the possibility of the use of probiotics, prebiotics, and even dietary changes as part of a new treatment strategy for neuropsychiatric disorders and their symptoms. CONCLUSION The bidirectional communication between the gut and the brain through the gut-brain axis has revealed profound implications for both gastrointestinal and neurological health. Malfunctions in this axis have been connected to a range of disorders affecting gut function as well as cognitive and neuropsychiatric well-being. The emerging understanding of the role of gut microbiota in regulating key neurochemicals opens up possibilities for novel treatment approaches for conditions like depression, anxiety, and neurodegenerative diseases.
Collapse
Affiliation(s)
- Saumya Gupta
- Department of Bioinformatics, BioNome, Bengaluru, India
| | - Susha Dinesh
- Department of Bioinformatics, BioNome, Bengaluru, India
| | - Sameer Sharma
- Department of Bioinformatics, BioNome, Bengaluru, India
| |
Collapse
|
5
|
Bransfield RC, Mao C, Greenberg R. Microbes and Mental Illness: Past, Present, and Future. Healthcare (Basel) 2023; 12:83. [PMID: 38200989 PMCID: PMC10779437 DOI: 10.3390/healthcare12010083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/30/2023] [Accepted: 12/06/2023] [Indexed: 01/12/2024] Open
Abstract
A review of the association between microbes and mental illness is performed, including the history, relevant definitions, infectious agents associated with mental illnesses, complex interactive infections, total load theory, pathophysiology, psychoimmunology, psychoneuroimmunology, clinical presentations, early-life infections, clinical assessment, and treatment. Perspectives on the etiology of mental illness have evolved from demonic possession toward multisystem biologically based models that include gene expression, environmental triggers, immune mediators, and infectious diseases. Microbes are associated with a number of mental disorders, including autism, schizophrenia, bipolar disorder, depressive disorders, and anxiety disorders, as well as suicidality and aggressive or violent behaviors. Specific microbes that have been associated or potentially associated with at least one of these conditions include Aspergillus, Babesia, Bartonella, Borna disease virus, Borrelia burgdorferi (Lyme disease), Candida, Chlamydia, coronaviruses (e.g., SARS-CoV-2), Cryptococcus neoformans, cytomegalovirus, enteroviruses, Epstein-Barr virus, hepatitis C, herpes simplex virus, human endogenous retroviruses, human immunodeficiency virus, human herpesvirus-6 (HHV-6), human T-cell lymphotropic virus type 1, influenza viruses, measles virus, Mycoplasma, Plasmodium, rubella virus, Group A Streptococcus (PANDAS), Taenia solium, Toxoplasma gondii, Treponema pallidum (syphilis), Trypanosoma, and West Nile virus. Recognition of the microbe and mental illness association with the development of greater interdisciplinary research, education, and treatment options may prevent and reduce mental illness morbidity, disability, and mortality.
Collapse
Affiliation(s)
- Robert C. Bransfield
- Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
- Hackensack Meridian School of Medicine, Nutey, NJ 07110, USA
| | | | | |
Collapse
|
6
|
Dupont D, Robert M, Brenier-Pinchart M, Lefevre A, Wallon M, Pelloux H. Toxoplasma gondii, a plea for a thorough investigation of its oncogenic potential. Heliyon 2023; 9:e22147. [PMID: 38034818 PMCID: PMC10685377 DOI: 10.1016/j.heliyon.2023.e22147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 10/20/2023] [Accepted: 11/05/2023] [Indexed: 12/02/2023] Open
Abstract
It is estimated that 30 % of the world's population harbours the parasite Toxoplasma gondii, particularly in the brain. Beyond its implication in potentially severe opportunistic or congenital infections, this persistence has long been considered as without consequence. However, certain data in animals and humans suggest that this carriage may be linked to various neuropsychiatric or neurodegenerative disorders. The hypothesis of a potential cerebral oncogenicity of the parasite is also emerging. In this personal view, we will present the epidemiological arguments in favour of an association between toxoplasmosis and cerebral malignancy, before considering the points that could underlie a potential causal link. More specifically, we will focus on the brain as the preferred location for T. gondii persistence and the propensity of this parasite to interfere with the apoptosis and cell cycle signalling pathways of their host cell.
Collapse
Affiliation(s)
- D. Dupont
- Institut des Agents Infectieux, Parasitologie Mycologie, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, 69004, France
- Physiologie intégrée du système d’éveil, Centre de Recherche en Neurosciences de Lyon, INSERM U1028-CNRS UMR 5292, Faculté de Médecine, Université Claude Bernard Lyon 1, Bron, 69500, France
| | - M.G. Robert
- Service de Parasitologie-Mycologie, CHU Grenoble Alpes, Grenoble, 38000, France
- Université Grenoble Alpes, Institut pour l'Avancée des Biosciences (IAB), INSERM U1209-CNRS UMR 5309, Grenoble, 38000, France
| | - M.P. Brenier-Pinchart
- Service de Parasitologie-Mycologie, CHU Grenoble Alpes, Grenoble, 38000, France
- Université Grenoble Alpes, Institut pour l'Avancée des Biosciences (IAB), INSERM U1209-CNRS UMR 5309, Grenoble, 38000, France
| | - A. Lefevre
- Institut des Agents Infectieux, Parasitologie Mycologie, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, 69004, France
| | - M. Wallon
- Institut des Agents Infectieux, Parasitologie Mycologie, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, 69004, France
- Physiologie intégrée du système d’éveil, Centre de Recherche en Neurosciences de Lyon, INSERM U1028-CNRS UMR 5292, Faculté de Médecine, Université Claude Bernard Lyon 1, Bron, 69500, France
| | - H. Pelloux
- Service de Parasitologie-Mycologie, CHU Grenoble Alpes, Grenoble, 38000, France
- Université Grenoble Alpes, Institut pour l'Avancée des Biosciences (IAB), INSERM U1209-CNRS UMR 5309, Grenoble, 38000, France
| |
Collapse
|
7
|
Sager REH, Walker AK, Middleton FA, Robinson K, Webster MJ, Gentile K, Wong ML, Shannon Weickert C. Changes in cytokine and cytokine receptor levels during postnatal development of the human dorsolateral prefrontal cortex. Brain Behav Immun 2023; 111:186-201. [PMID: 36958512 DOI: 10.1016/j.bbi.2023.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 03/09/2023] [Accepted: 03/16/2023] [Indexed: 03/25/2023] Open
Abstract
In addition to their traditional roles in immune cell communication, cytokines regulate brain development. Cytokines are known to influence neural cell generation, differentiation, maturation, and survival. However, most work on the role of cytokines in brain development investigates rodents or focuses on prenatal events. Here, we investigate how mRNA and protein levels of key cytokines and cytokine receptors change during postnatal development of the human prefrontal cortex. We find that most cytokine transcripts investigated (IL1B, IL18, IL6, TNF, IL13) are lowest at birth and increase between 1.5 and 5 years old. After 5 years old, transcriptional patterns proceeded in one of two directions: decreased expression in teens and young adults (IL1B, p = 0.002; and IL18, p = 0.004) or increased mean expression with maturation, particularly in teenagers (IL6, p = 0.004; TNF, p = 0.002; IL13, p < 0.001). In contrast, cytokine proteins tended to remain elevated after peaking significantly around 3 years of age (IL1B, p = 0.012; IL18, p = 0.026; IL6, p = 0.039; TNF, p < 0.001), with TNF protein being highest in teenagers. An mRNA-only analysis of cytokine receptor transcripts found that early developmental increases in cytokines were paralleled by increases in their ligand-binding receptor subunits, such as IL1R1 (p = 0.033) and IL6R (p < 0.001) transcripts. In contrast, cytokine receptor-associated signaling subunits, IL1RAP and IL6ST, did not change significantly between age groups. Of the two TNF receptors, the 'pro-death' TNFRSF1A and 'pro-survival' TNFRSF1B, only TNFRSF1B was significantly changed (p = 0.028), increasing first in toddlers and again in young adults. Finally, the cytokine inhibitor, IL13, was elevated first in toddlers (p = 0.006) and again in young adults (p = 0.053). While the mean expression of interleukin-1 receptor antagonist (IL1RN) was highest in toddlers, this increase was not statistically significant. The fluctuations in cytokine expression reported here support a role for increases in specific cytokines at two different stages of human cortical development. The first is during the toddler/preschool period (IL1B, IL18, and IL13), and the other occurs at adolescence/young adult maturation (IL6, TNF and IL13).
Collapse
Affiliation(s)
- Rachel E H Sager
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Adam K Walker
- Laboratory of Immunopsychiatry, Neuroscience Research Australia, Sydney, NSW, Australia; Discipline of Psychiatry and Mental Health, University of New South Wales, Sydney, NSW, Australia; Monash Institute of Pharmaceutical Science, Monash University, Parkville, VIC, Australia
| | - Frank A Middleton
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Kate Robinson
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney, NSW, Australia
| | | | - Karen Gentile
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Ma-Li Wong
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, USA; Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Cynthia Shannon Weickert
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, USA; Discipline of Psychiatry and Mental Health, University of New South Wales, Sydney, NSW, Australia; Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney, NSW, Australia.
| |
Collapse
|
8
|
Burton BK, Andersen KK, Greve AN, Hemager N, Spang KS, Ellersgaard D, Christiani CJ, Gantriis D, Gregersen M, Søndergaard A, Jepsen JRM, Bliksted VF, Mors O, Plessen KJ, Nordentoft M, Thorup AAE. Sex differences across developmental domains among children with a familial risk of severe mental disorders. Psychol Med 2023; 53:3628-3643. [PMID: 35156599 DOI: 10.1017/s0033291722000265] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Sex differences in brain structure and neurodevelopment occur in non-clinical populations. We investigated whether sex had a similar effect on developmental domains amongst boys and girls with a familial risk of schizophrenia (FHR-SZ), bipolar disorder (FHR-BP), and controls. METHODS Through Danish registries, we identified 522 7-year-old children (242 girls) with FHR-SZ, FHR-BP, and controls. We assessed their performance within the domains of neurocognition, motor function, language, social cognition, social behavior, psychopathology, and home environment. RESULTS FHR-SZ boys compared with FHR-SZ girls had a higher proportion of disruptive behavior and attention-deficit hyperactivity disorder (ADHD) and exhibited lower performance in manual dexterity, balance, and emotion recognition. No sex differences were found between boys and girls within FHR-BP group. Compared with controls, both FHR-SZ boys and FHR-SZ girls showed impaired processing speed and working memory, had lower levels of global functioning, and were more likely to live in an inadequate home environment. Compared with control boys, FHR-SZ boys showed impaired manual dexterity, social behavior, and social responsiveness, and had a higher proportion of ADHD and disruptive behavior disorder diagnoses. Stress and adjustment disorders were more common in FHR-BP boys compared with control boys. We found no differences between FHR-BP girls and control girls. CONCLUSIONS Impairment within neurodevelopmental domains associated within FHR-SZ boys v. FHR-SZ girls was most evident among boys, whereas no sex differences were found within the FHR-BP group (FHR-BP boys v. FHR-BP girls). FHR-SZ boys exhibited the highest proportion of early developmental impairments.
Collapse
Affiliation(s)
- Birgitte Klee Burton
- Child and Adolescent Mental Health Centre, Mental Health Services Capital Region, Research Unit, Copenhagen University Hospital, Gentofte Hospitalsvej 3A, 1st floor, 2900 Hellerup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Aarhus, Denmark
| | - Klaus Kaae Andersen
- Danish Cancer Society Research Center, Statistics and Pharmacoepidemiology, Strandboulevarden 49, 2100 Copenhagen, Denmark
| | - Aja N Greve
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Aarhus, Denmark
- Psychosis Research Unit, Aarhus University Hospital Psychiatry, Palle Juul-Jensens Boulevard 175, 8200 Aarhus N, Denmark
| | - Nicoline Hemager
- Child and Adolescent Mental Health Centre, Mental Health Services Capital Region, Research Unit, Copenhagen University Hospital, Gentofte Hospitalsvej 3A, 1st floor, 2900 Hellerup, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Aarhus, Denmark
- Copenhagen Research Center for Mental Health - CORE, Mental Health Centre Copenhagen, Copenhagen University Hospital, Mental Health Services Capital Region, Gentofte Hospitalsvej 15, 4th floor, 2900 Hellerup, Denmark
| | - Katrine S Spang
- Child and Adolescent Mental Health Centre, Mental Health Services Capital Region, Research Unit, Copenhagen University Hospital, Gentofte Hospitalsvej 3A, 1st floor, 2900 Hellerup, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Aarhus, Denmark
| | - Ditte Ellersgaard
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Aarhus, Denmark
- Copenhagen Research Center for Mental Health - CORE, Mental Health Centre Copenhagen, Copenhagen University Hospital, Mental Health Services Capital Region, Gentofte Hospitalsvej 15, 4th floor, 2900 Hellerup, Denmark
| | - Camilla J Christiani
- Child and Adolescent Mental Health Centre, Mental Health Services Capital Region, Research Unit, Copenhagen University Hospital, Gentofte Hospitalsvej 3A, 1st floor, 2900 Hellerup, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Aarhus, Denmark
| | - Ditte Gantriis
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Aarhus, Denmark
- Psychosis Research Unit, Aarhus University Hospital Psychiatry, Palle Juul-Jensens Boulevard 175, 8200 Aarhus N, Denmark
| | - Maja Gregersen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Aarhus, Denmark
- Copenhagen Research Center for Mental Health - CORE, Mental Health Centre Copenhagen, Copenhagen University Hospital, Mental Health Services Capital Region, Gentofte Hospitalsvej 15, 4th floor, 2900 Hellerup, Denmark
| | - Anne Søndergaard
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Aarhus, Denmark
- Copenhagen Research Center for Mental Health - CORE, Mental Health Centre Copenhagen, Copenhagen University Hospital, Mental Health Services Capital Region, Gentofte Hospitalsvej 15, 4th floor, 2900 Hellerup, Denmark
| | - Jens Richardt M Jepsen
- Child and Adolescent Mental Health Centre, Mental Health Services Capital Region, Research Unit, Copenhagen University Hospital, Gentofte Hospitalsvej 3A, 1st floor, 2900 Hellerup, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Aarhus, Denmark
- Copenhagen Research Center for Mental Health - CORE, Mental Health Centre Copenhagen, Copenhagen University Hospital, Mental Health Services Capital Region, Gentofte Hospitalsvej 15, 4th floor, 2900 Hellerup, Denmark
- Centre for Neuropsychiatric Schizophrenia Research & Centre for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Copenhagen University Hospital, Psychiatric Hospital Centre Glostrup, Ndr. Ringvej 29-67, 2600 Glostrup, Denmark
| | - Vibeke Fuglsang Bliksted
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Aarhus, Denmark
- Psychosis Research Unit, Aarhus University Hospital Psychiatry, Palle Juul-Jensens Boulevard 175, 8200 Aarhus N, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Services, Aarhus University, Palle Juul-Jensens Boulevard 82, 8200 Aarhus N, Denmark
| | - Ole Mors
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Aarhus, Denmark
- Psychosis Research Unit, Aarhus University Hospital Psychiatry, Palle Juul-Jensens Boulevard 175, 8200 Aarhus N, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Services, Aarhus University, Palle Juul-Jensens Boulevard 82, 8200 Aarhus N, Denmark
| | - Kerstin Jessica Plessen
- Child and Adolescent Mental Health Centre, Mental Health Services Capital Region, Research Unit, Copenhagen University Hospital, Gentofte Hospitalsvej 3A, 1st floor, 2900 Hellerup, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Aarhus, Denmark
- Division of Child and Adolescent Psychiatry, Department of Psychiatry, University Medical Center, University of Lausanne, Avenue d'Echallens 9, CH-1004 Lausanne, Switzerland
| | - Merete Nordentoft
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Aarhus, Denmark
- Copenhagen Research Center for Mental Health - CORE, Mental Health Centre Copenhagen, Copenhagen University Hospital, Mental Health Services Capital Region, Gentofte Hospitalsvej 15, 4th floor, 2900 Hellerup, Denmark
| | - Anne A E Thorup
- Child and Adolescent Mental Health Centre, Mental Health Services Capital Region, Research Unit, Copenhagen University Hospital, Gentofte Hospitalsvej 3A, 1st floor, 2900 Hellerup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Aarhus, Denmark
| |
Collapse
|
9
|
Herrera-Imbroda J, Flores-López M, Ruiz-Sastre P, Gómez-Sánchez-Lafuente C, Bordallo-Aragón A, Rodríguez de Fonseca F, Mayoral-Cleríes F. The Inflammatory Signals Associated with Psychosis: Impact of Comorbid Drug Abuse. Biomedicines 2023; 11:biomedicines11020454. [PMID: 36830990 PMCID: PMC9953424 DOI: 10.3390/biomedicines11020454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/27/2023] [Accepted: 02/01/2023] [Indexed: 02/08/2023] Open
Abstract
Psychosis and substance use disorders are two diagnostic categories whose association has been studied for decades. In addition, both psychosis spectrum disorders and drug abuse have recently been linked to multiple pro-inflammatory changes in the central nervous system. We have carried out a narrative review of the literature through a holistic approach. We used PubMed as our search engine. We included in the review all relevant studies looking at pro-inflammatory changes in psychotic disorders and substance use disorders. We found that there are multiple studies that relate various pro-inflammatory lipids and proteins with psychosis and substance use disorders, with an overlap between the two. The main findings involve inflammatory mediators such as cytokines, chemokines, endocannabinoids, eicosanoids, lysophospholipds and/or bacterial products. Many of these findings are present in different phases of psychosis and in substance use disorders such as cannabis, cocaine, methamphetamines, alcohol and nicotine. Psychosis and substance use disorders may have a common origin in an abnormal neurodevelopment caused, among other factors, by a neuroinflammatory process. A possible convergent pathway is that which interrelates the transcriptional factors NFκB and PPARγ. This may have future clinical implications.
Collapse
Affiliation(s)
- Jesús Herrera-Imbroda
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
- Facultad de Medicina, Universidad de Málaga, Andalucía Tech, Campus de Teatinos s/n, 29071 Málaga, Spain
- Departamento de Farmacología y Pediatría, Universidad de Málaga, Andalucía Tech, Campus de Teatinos s/n, 29071 Málaga, Spain
| | - María Flores-López
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
- Facultad de Psicología, Universidad de Málaga, Andalucía Tech, Campus de Teatinos s/n, 29071 Málaga, Spain
| | - Paloma Ruiz-Sastre
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
- Facultad de Medicina, Universidad de Málaga, Andalucía Tech, Campus de Teatinos s/n, 29071 Málaga, Spain
- Correspondence: (P.R.-S.); (C.G.-S.-L.)
| | - Carlos Gómez-Sánchez-Lafuente
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
- Facultad de Psicología, Universidad de Málaga, Andalucía Tech, Campus de Teatinos s/n, 29071 Málaga, Spain
- Correspondence: (P.R.-S.); (C.G.-S.-L.)
| | - Antonio Bordallo-Aragón
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
| | - Fernando Rodríguez de Fonseca
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
| | - Fermín Mayoral-Cleríes
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
| |
Collapse
|
10
|
Lipopolysaccharide Exacerbates Ketamine-Induced Psychotic-Like Behavior, Oxidative Stress, and Neuroinflammation in Mice: Ameliorative Effect of Diosmin. J Mol Neurosci 2023; 73:129-142. [PMID: 36652038 DOI: 10.1007/s12031-022-02077-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 10/07/2022] [Indexed: 01/19/2023]
Abstract
Schizophrenia, a neuropsychiatric disorder has been associated with aberrant neurotransmission affecting behaviors, social preference, and cognition. Limitations in understanding its pathogenesis via the dopamine hypothesis have engendered other hypotheses such as the glutamate hypothesis. That antagonism of the N-methyl-D-aspartate receptor (NMDAR) elicits schizophrenia-like behaviors indistinguishable from the disorder in animal and human models. There are growing concerns that redox imbalance and neuro-immuno dysfunction may play role in aggravating the symptomologies of this disorder. This 14-day treatment study was designed to investigate the effect of diosmin on lipopolysaccharide (LPS) plus ketamine (NMDAR antagonist). Mice were divided into 4 groups (n = 6). Group 1 was administered 5% DMSO (10 mL/kg, i.p) while group 2-4 received LPS (0.1 mg/kg, i.p) daily for 14 days. Diosmin (50 mg/kg, i.p) and risperidone (0.5 mg/kg, i.p) were given to groups 3 and 4 respectively. Groups 2-4 were given KET (20 mg/kg, i.p.) daily from days 8-14. Behavioral tests were done 30 min after the last dose, and oxidative stress and neuroinflammatory maker were assayed. LPS plus ketamine-induced hyperlocomotion, stereotypy, decreased social preference, and memory impairment. Furthermore, LPS plus-ketamine-induced oxidative stress (reduced GSH, CAT, SOD, and increased MDA and nitrite levels) and marked pro-inflammatory cytokines TNF-α and IL-6 suggesting neuroinflammation. However, diosmin attenuated behavioral deficits and improved memory. Additionally, diosmin potentiated antioxidant level via increased GSH, CAT, and SOD while reducing MDA and nitrite levels. Finally, diosmin reduced TNF-α and IL-6 suggesting anti-neuro-immuno activity. Conclusively, diosmin attenuated LPS plus ketamine-induced behavioral deficits, oxidative stress, neuroinflammation, and improved memory.
Collapse
|
11
|
Shin C, Kim YK. Microbiota-Gut-Brain Axis: Pathophysiological Mechanism in Neuropsychiatric Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1411:17-37. [PMID: 36949304 DOI: 10.1007/978-981-19-7376-5_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
Gut microbiota influence human behavior. The immunological, metabolic, and endocrine systems are involved in bidirectional communication between the gut and the brain, which is regulated by microbes through the microbiota-derived neurochemicals and metabolites. Gut microbiota have certain effects on neurodevelopment and maturation of immunity. However, gut dysbiosis can lead to neuropsychiatric disorders. Animal research and clinical case-control studies have demonstrated that gut dysbiosis has an adverse effect on human behavior through a variety of mechanisms. Recent meta-analysis on clinical studies confirmed gut dysbiosis in several major neuropsychiatric disorders. Microbiota-targeted intervention has recently been in the spotlight and meta-analyses have confirmed its effectiveness. In this chapter, we summarize the evidence for the interactions between microbiota and brain-gut network, as well as the potential pathophysiological mechanisms involved.
Collapse
Affiliation(s)
- Cheolmin Shin
- Department of Psychiatry, College of Medicine, Korea University Ansan Hospital, Ansan, Republic of Korea
| | - Yong-Ku Kim
- Department of Psychiatry, College of Medicine, Korea University Ansan Hospital, Ansan, Republic of Korea.
| |
Collapse
|
12
|
Abstract
There is increasingly compelling evidence that microorganisms may play an etiological role in the emergence of mental illness in a subset of the population. Historically, most work has focused on the neurotrophic herpesviruses, herpes simplex virus type 1 (HSV-1), cytomegalovirus (CMV), and Epstein-Barr virus (EBV) as well as the protozoan, Toxoplasma gondii. In this chapter, we provide an umbrella review of this literature and additionally highlight prospective studies that allow more mechanistic conclusions to be drawn. Next, we focus on clinical trials of anti-microbial medications for the treatment of psychiatric disorders. We critically evaluate six trials that tested the impact of anti-herpes medications on inflammatory outcomes in the context of a medical disorder, nine clinical trials utilizing anti-herpetic medications for the treatment of myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) or schizophrenia, and four clinical trials utilizing anti-parasitic medications for the treatment of schizophrenia. We then turn our attention to evidence for a gut dysbiosis and altered microbiome in psychiatric disorders, and the potential therapeutic effects of probiotics, including an analysis of more than 10 randomized controlled trials of probiotics in the context of schizophrenia, bipolar disorder (BD), and major depressive disorder (MDD).
Collapse
|
13
|
Hall MB, Willis DE, Rodriguez EL, Schwarz JM. Maternal immune activation as an epidemiological risk factor for neurodevelopmental disorders: Considerations of timing, severity, individual differences, and sex in human and rodent studies. Front Neurosci 2023; 17:1135559. [PMID: 37123361 PMCID: PMC10133487 DOI: 10.3389/fnins.2023.1135559] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 03/13/2023] [Indexed: 05/02/2023] Open
Abstract
Epidemiological evidence suggests that one's risk of being diagnosed with a neurodevelopmental disorder (NDD)-such as autism, ADHD, or schizophrenia-increases significantly if their mother had a viral or bacterial infection during the first or second trimester of pregnancy. Despite this well-known data, little is known about how developing neural systems are perturbed by events such as early-life immune activation. One theory is that the maternal immune response disrupts neural processes important for typical fetal and postnatal development, which can subsequently result in specific and overlapping behavioral phenotypes in offspring, characteristic of NDDs. As such, rodent models of maternal immune activation (MIA) have been useful in elucidating neural mechanisms that may become dysregulated by MIA. This review will start with an up-to-date and in-depth, critical summary of epidemiological data in humans, examining the association between different types of MIA and NDD outcomes in offspring. Thereafter, we will summarize common rodent models of MIA and discuss their relevance to the human epidemiological data. Finally, we will highlight other factors that may interact with or impact MIA and its associated risk for NDDs, and emphasize the importance for researchers to consider these when designing future human and rodent studies. These points to consider include: the sex of the offspring, the developmental timing of the immune challenge, and other factors that may contribute to individual variability in neural and behavioral responses to MIA, such as genetics, parental age, the gut microbiome, prenatal stress, and placental buffering.
Collapse
|
14
|
Dubey H, Sharma RK, Krishnan S, Knickmeyer R. SARS-CoV-2 (COVID-19) as a possible risk factor for neurodevelopmental disorders. Front Neurosci 2022; 16:1021721. [PMID: 36590303 PMCID: PMC9800937 DOI: 10.3389/fnins.2022.1021721] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
Pregnant women constitute one of the most vulnerable populations to be affected by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, the cause of coronavirus disease 2019. SARS-CoV-2 infection during pregnancy could negatively impact fetal brain development via multiple mechanisms. Accumulating evidence indicates that mother to fetus transmission of SARS-CoV-2 does occur, albeit rarely. When it does occur, there is a potential for neuroinvasion via immune cells, retrograde axonal transport, and olfactory bulb and lymphatic pathways. In the absence of maternal to fetal transmission, there is still the potential for negative neurodevelopmental outcomes as a consequence of disrupted placental development and function leading to preeclampsia, preterm birth, and intrauterine growth restriction. In addition, maternal immune activation may lead to hypomyelination, microglial activation, white matter damage, and reduced neurogenesis in the developing fetus. Moreover, maternal immune activation can disrupt the maternal or fetal hypothalamic-pituitary-adrenal (HPA) axis leading to altered neurodevelopment. Finally, pro-inflammatory cytokines can potentially alter epigenetic processes within the developing brain. In this review, we address each of these potential mechanisms. We propose that SARS-CoV-2 could lead to neurodevelopmental disorders in a subset of pregnant women and that long-term studies are warranted.
Collapse
Affiliation(s)
- Harikesh Dubey
- Division of Neuroengineering, Institute for Quantitative Health Sciences and Engineering, Michigan State University, East Lansing, MI, United States
| | - Ravindra K. Sharma
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Suraj Krishnan
- Jacobi Medical Center, Albert Einstein College of Medicine, The Bronx, NY, United States
| | - Rebecca Knickmeyer
- Division of Neuroengineering, Institute for Quantitative Health Sciences and Engineering, Michigan State University, East Lansing, MI, United States,Department of Pediatrics and Human Development, Michigan State University, East Lansing, MI, United States,*Correspondence: Rebecca Knickmeyer,
| |
Collapse
|
15
|
Pregestational Exposure to T. gondii Produces Maternal Antibodies That Recognize Fetal Brain Mimotopes and Induces Neurochemical and Behavioral Dysfunction in the Offspring. Cells 2022; 11:cells11233819. [PMID: 36497079 PMCID: PMC9741080 DOI: 10.3390/cells11233819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/23/2022] [Accepted: 11/25/2022] [Indexed: 11/30/2022] Open
Abstract
The activation of the maternal immune system by a prenatal infection is considered a risk factor for developing psychiatric disorders in the offspring. Toxoplasma gondii is one of the pathogenic infections associated with schizophrenia. Recent studies have shown an association between high levels of IgG anti-T. gondii from mothers and their neonates, with a higher risk of developing schizophrenia. The absence of the parasite and the levels of IgGs found in the early stages of life suggest a transplacental transfer of the anti-T. gondii IgG antibodies, which could bind fetal brain structures by molecular mimicry and induce alterations in neurodevelopment. This study aimed to determine the maternal pathogenic antibodies formation that led to behavioral impairment on the progeny of rats immunized with T. gondii. Female rats were immunized prior to gestation with T. gondii lysate (3 times/once per week). The anti-T. gondii IgG levels were determined in the serum of pregestational exposed females' previous mating. After this, locomotor activity, cognitive and social tests were performed. Cortical neurotransmitter levels for dopamine and glutamate were evaluated at 60 PND in the progeny of rats immunized before gestation (Pregestational group). The maternal pathogenic antibodies were evidenced by their binding to fetal brain mimotopes in the Pregestational group and the reactivity of the serum containing anti-T. gondii IgG was tested in control fetal brains (non-immunized). These results showed that the Pregestational group presented impairment in short and long-term memory, hypoactivity and alteration in social behavior, which was also associated with a decrease in cortical glutamate and dopamine levels. We also found the IgG antibodies bound to brain mimotopes in fetuses from females immunized with T. gondii, as well as observing a strong reactivity of the serum females immunized for fetal brain structures of fetuses from unimmunized mothers. Our results suggest that the exposure to T. gondii before gestation produced maternal pathogenic antibodies that can recognize fetal brain mimotopes and lead to neurochemical and behavioral alterations in the offspring.
Collapse
|
16
|
Bahreini MS, Sami Jahromi S, Radfar AH, Salemi AM, Dastan N, Asgari Q. The Relationship of Latent Toxoplasmosis and Cigarette Smoking: Seroprevalence, Risk Factor, and Case-Control Study in Fars Province, Southern Iran. Pathogens 2022; 11:1274. [PMID: 36365025 PMCID: PMC9696781 DOI: 10.3390/pathogens11111274] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/06/2022] [Accepted: 09/15/2022] [Indexed: 12/20/2023] Open
Abstract
Toxoplasmosis is a parasitic disease with worldwide prevalence. Despite the relatively similar effects of toxoplasmosis and smoking on alteration in neurotransmitters, especially dopamine, little is known about the relation of Toxoplasma gondii infection and addiction to cigarette smoking. Therefore, the main objective of this study was to assess the relationship between latent toxoplasmosis and smoking. Through a case-control study, 216 regular cigarette smokers and 324 nonsmoker age- and gender-matched subjects were evaluated for anti-T.gondii IgG antibodies with enzyme-linked immunosorbent assay (ELISA). During the sampling, a structured questionnaire was used to obtain the demographic information of participants and the risk factors of acquired Toxoplasma. The median ages of case and control groups were 51.04 ± 18.1 (22-97 years) and 51.03 ± 16.5 (21-89 years), respectively (p = 0.99). Anti-T.gondii IgG antibodies were detected in 44 (20.37%) cases and in 135 (41.67%) controls. There was a statistically significant difference for the positivity rate between the smokers and the control group (OR = 0.35; 95%CI: 0.19-0.65; and p = 0.001). The overall prevalence was 33.14%. This study indicated the inverse association between seropositivity to Toxoplasma infection and cigarette smoking. This relationship could be due to the changes that latent toxoplasmosis has on the neurotransmitters, especially dopamine, which needs more research.
Collapse
Affiliation(s)
- Mohammad Saleh Bahreini
- Department of Parasitology and Mycology, School of Medicine, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
| | - Sareh Sami Jahromi
- Department of Pathology, School of Medicine, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
| | - Amir Hossein Radfar
- Department of Parasitology and Mycology, School of Medicine, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
| | - Amir Masoud Salemi
- Department of Parasitology and Mycology, School of Medicine, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
| | - Naghmeh Dastan
- Department of Parasitology and Mycology, School of Medicine, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
| | - Qasem Asgari
- Department of Parasitology and Mycology, School of Medicine, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
| |
Collapse
|
17
|
Gupta L, Hoffman KW. Exploring the intersection of the microbiome and the developing brain: Impacts on schizophrenia risk. Schizophr Res 2022; 247:92-100. [PMID: 34483026 DOI: 10.1016/j.schres.2021.08.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 08/04/2021] [Accepted: 08/08/2021] [Indexed: 12/21/2022]
Abstract
Recent findings show that the perinatal maternal and infant microbiomes have profound potential to impact long term health outcomes. Of particular interest are the ways in which the microbiome influences the developing brain during one of its most critical windows. Schizophrenia and psychosis risk are strongly connected to disruptions in perinatal neurodevelopment. In this review we present an overview of critical aspects in development of both the microbiome and brain, discuss their overlap, and consider what role the microbiome plays in schizophrenia risk during the perinatal window. Considering this, we discuss ways in which expecting and new mothers may reduce offspring schizophrenia risk.
Collapse
Affiliation(s)
- Lipi Gupta
- The University of Pennsylvania, Department of Psychiatry, Philadelphia, PA, United States
| | - Kevin W Hoffman
- The University of Pennsylvania, Department of Psychiatry, Philadelphia, PA, United States.
| |
Collapse
|
18
|
Haddad FL, Patel SV, Doornaert EE, De Oliveira C, Allman BL, Baines KJ, Renaud SJ, Schmid S. Interleukin 15 modulates the effects of poly I:C maternal immune activation on offspring behaviour. Brain Behav Immun Health 2022; 23:100473. [PMID: 35668725 PMCID: PMC9166394 DOI: 10.1016/j.bbih.2022.100473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 05/14/2022] [Indexed: 01/21/2023] Open
Abstract
Maternal infections during pregnancy are linked with an increased risk for disorders like Autism Spectrum Disorder and schizophrenia in the offspring. Although precise mechanisms are still unclear, clinical and preclinical evidence suggest a strong role for maternal immune activation (MIA) in the neurodevelopmental disruptions caused by maternal infection. Previously, studies using the Polyinosinic:Polycytidylic (Poly I:C) MIA preclinical model showed that cytokines like Interleukin 6 (Il6) are important mediators of MIA's effects. In this study, we hypothesized that Il15 may similarly act as a mediator of Poly I:C MIA, given its role in the antiviral immune response. To test this hypothesis, we induced Poly I:C MIA at gestational day 9.5 in wildtype (WT) and Il15−/− rat dams and tested their offspring in adolescence and adulthood. Poly I:C MIA and Il15 knockout produced both independent and synergistic effects on offspring behaviour. Poly I:C MIA decreased startle reactivity in adult WT offspring but resulted in increased adolescent anxiety and decreased adult locomotor activity in Il15−/− offspring. In addition, Poly I:C MIA led to genotype-independent effects on locomotor activity and prepulse inhibition. Finally, we showed that Il15−/− offspring exhibit distinct phenotypes that were unrelated to Poly I:C MIA including altered startle reactivity, locomotion and signal transduction in the auditory brainstem. Overall, our findings indicate that the lack of Il15 can leave offspring either more or less susceptible to Poly I:C MIA, depending on the phenotype in question. Future studies should examine the contribution of fetal versus maternal Il15 in MIA to determine the precise developmental mechanisms underlying these changes. Poly I:C MIA decreases startle reactivity in adult WT but not Il15−/− offspring. Il15−/− offspring exposed to Poly I:C MIA show altered PPI and open field exploration. Il15−/− rats exhibit distinct behavioural phenotypes independent from MIA.
Collapse
|
19
|
Huang Z, Kang M, Li G, Xiong P, Chen H, Kang L, Li S, Lu C, Li Q, Bai M. Predictive effect of Bayes discrimination in the level of serum protein factors and cognitive dysfunction in schizophrenia. J Psychiatr Res 2022; 151:539-545. [PMID: 35636029 DOI: 10.1016/j.jpsychires.2022.05.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 03/23/2022] [Accepted: 05/09/2022] [Indexed: 01/19/2023]
Abstract
Finding molecular biomarkers that can be related to the degree of cognitive dysfunction in schizophrenia remains a challenge. The levels of 6 Serum Protein Factors (NGF, BDNF, IL-6, TNF-α, S100β, GFAP) in peripheral blood of patients with schizophrenia were measured. The cognitive function of patients with schizophrenia was assessed by MATRICS Consensus Cognitive Battery (MCCB), a systematic assessment tool of international gold standard for cognitive function assessment of schizophrenia. To explore the correlation between these 6 biomarkers and the degree of cognitive dysfunction in schizophrenia,78 schizophrenic patients and 71 healthy controls were included in the study. The serum concentrations of BDNF and GFAP were lower in the patient group, but the concentrations of IL-6, TNF-α and S100β were higher. The speed of information processing, word learning, reasoning and problem solving, visual learning T-score of the patient group were lower than the control group. Bayes discriminant function model has a high correct discriminant rate for the severity of cognitive dysfunction in schizophrenia. The level of serum protein factor and clinical symptom score of schizophrenia may forecast the degree of cognitive dysfunction, which is expected to be a potential biomarker to identify the degree of cognitive dysfunction of schizophrenia, and provide objective basis for the clinical diagnosis and treatment of patients with schizophrenia.
Collapse
Affiliation(s)
- Zhengyuan Huang
- Department of Psychiatry, The First Affiliated Hospital of Kunming Medical University, #295 Xichang, Road, Kunming, Yunnan, 650032, China
| | - Minmin Kang
- Department of Psychiatry, The First Affiliated Hospital of Kunming Medical University, #295 Xichang, Road, Kunming, Yunnan, 650032, China; Department of Psychiatry, The Affiliated Hospital of Hubei Minzu University, #39 Xueyuan Road, Enshi, Hubei,445000, China
| | - Guangyu Li
- Department of Psychiatry, The First Affiliated Hospital of Kunming Medical University, #295 Xichang, Road, Kunming, Yunnan, 650032, China
| | - Peng Xiong
- Department of Psychiatry, The First Affiliated Hospital of Kunming Medical University, #295 Xichang, Road, Kunming, Yunnan, 650032, China; Yunnan Clinical Research Center for Mental Health, Kunming, Yunnan, 650032, China.
| | - Hongxu Chen
- Department of Psychiatry, The First Affiliated Hospital of Kunming Medical University, #295 Xichang, Road, Kunming, Yunnan, 650032, China
| | - Lin Kang
- Department of Psychiatry, The First Affiliated Hospital of Kunming Medical University, #295 Xichang, Road, Kunming, Yunnan, 650032, China
| | - Shan Li
- Department of Psychiatry, The First Affiliated Hospital of Kunming Medical University, #295 Xichang, Road, Kunming, Yunnan, 650032, China
| | - Cailian Lu
- Department of Psychiatry, The First Affiliated Hospital of Kunming Medical University, #295 Xichang, Road, Kunming, Yunnan, 650032, China
| | - Qianqian Li
- Department of Psychiatry, The First Affiliated Hospital of Kunming Medical University, #295 Xichang, Road, Kunming, Yunnan, 650032, China
| | - Meiyan Bai
- Department of Psychiatry, The First Affiliated Hospital of Kunming Medical University, #295 Xichang, Road, Kunming, Yunnan, 650032, China
| |
Collapse
|
20
|
Structural and Functional Deviations of the Hippocampus in Schizophrenia and Schizophrenia Animal Models. Int J Mol Sci 2022; 23:ijms23105482. [PMID: 35628292 PMCID: PMC9143100 DOI: 10.3390/ijms23105482] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/09/2022] [Accepted: 05/11/2022] [Indexed: 01/04/2023] Open
Abstract
Schizophrenia is a grave neuropsychiatric disease which frequently onsets between the end of adolescence and the beginning of adulthood. It is characterized by a variety of neuropsychiatric abnormalities which are categorized into positive, negative and cognitive symptoms. Most therapeutical strategies address the positive symptoms by antagonizing D2-dopamine-receptors (DR). However, negative and cognitive symptoms persist and highly impair the life quality of patients due to their disabling effects. Interestingly, hippocampal deviations are a hallmark of schizophrenia and can be observed in early as well as advanced phases of the disease progression. These alterations are commonly accompanied by a rise in neuronal activity. Therefore, hippocampal formation plays an important role in the manifestation of schizophrenia. Furthermore, studies with animal models revealed a link between environmental risk factors and morphological as well as electrophysiological abnormalities in the hippocampus. Here, we review recent findings on structural and functional hippocampal abnormalities in schizophrenic patients and in schizophrenia animal models, and we give an overview on current experimental approaches that especially target the hippocampus. A better understanding of hippocampal aberrations in schizophrenia might clarify their impact on the manifestation and on the outcome of this severe disease.
Collapse
|
21
|
Ryan AM, Bauman MD. Primate Models as a Translational Tool for Understanding Prenatal Origins of Neurodevelopmental Disorders Associated With Maternal Infection. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2022; 7:510-523. [PMID: 35276404 PMCID: PMC8902899 DOI: 10.1016/j.bpsc.2022.02.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 01/13/2022] [Accepted: 02/24/2022] [Indexed: 02/06/2023]
Abstract
Pregnant women represent a uniquely vulnerable population during an infectious disease outbreak, such as the COVID-19 pandemic. Although we are at the early stages of understanding the specific impact of SARS-CoV-2 exposure during pregnancy, mounting epidemiological evidence strongly supports a link between exposure to a variety of maternal infections and an increased risk for offspring neurodevelopmental disorders. Inflammatory biomarkers identified from archived or prospectively collected maternal biospecimens suggest that the maternal immune response is the critical link between infection during pregnancy and altered offspring neurodevelopment. This maternal immune activation (MIA) hypothesis has been tested in animal models by artificially activating the immune system during pregnancy and evaluating the neurodevelopmental consequences in MIA-exposed offspring. Although the vast majority of MIA model research is carried out in rodents, the nonhuman primate model has emerged in recent years as an important translational tool. In this review, we briefly summarize human epidemiological studies that have prompted the development of translationally relevant MIA models. We then highlight notable similarities between humans and nonhuman primates, including placental structure, pregnancy physiology, gestational timelines, and offspring neurodevelopmental stages, that provide an opportunity to explore the MIA hypothesis in species more closely related to humans. Finally, we provide a comprehensive review of neurodevelopmental alterations reported in current nonhuman primate models of maternal infection and discuss future directions for this promising area of research.
Collapse
Affiliation(s)
- Amy M Ryan
- Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis, Davis, California; California National Primate Research Center, University of California Davis, Davis, California
| | - Melissa D Bauman
- Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis, Davis, California; California National Primate Research Center, University of California Davis, Davis, California.
| |
Collapse
|
22
|
Elgueta D, Murgas P, Riquelme E, Yang G, Cancino GI. Consequences of Viral Infection and Cytokine Production During Pregnancy on Brain Development in Offspring. Front Immunol 2022; 13:816619. [PMID: 35464419 PMCID: PMC9021386 DOI: 10.3389/fimmu.2022.816619] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 03/02/2022] [Indexed: 12/12/2022] Open
Abstract
Infections during pregnancy can seriously damage fetal neurodevelopment by aberrantly activating the maternal immune system, directly impacting fetal neural cells. Increasing evidence suggests that these adverse impacts involve alterations in neural stem cell biology with long-term consequences for offspring, including neurodevelopmental disorders such as autism spectrum disorder, schizophrenia, and cognitive impairment. Here we review how maternal infection with viruses such as Influenza A, Cytomegalovirus, and Zika during pregnancy can affect the brain development of offspring by promoting the release of maternal pro-inflammatory cytokines, triggering neuroinflammation of the fetal brain, and/or directly infecting fetal neural cells. In addition, we review insights into how these infections impact human brain development from studies with animal models and brain organoids. Finally, we discuss how maternal infection with SARS-CoV-2 may have consequences for neurodevelopment of the offspring.
Collapse
Affiliation(s)
- Daniela Elgueta
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Paola Murgas
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago, Chile.,Escuela de Tecnología Médica, Facultad de Ciencias, Universidad Mayor, Santiago, Chile.,Escuela de Biotecnología, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Erick Riquelme
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago, Chile.,Escuela de Tecnología Médica, Facultad de Ciencias, Universidad Mayor, Santiago, Chile.,Escuela de Biotecnología, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Guang Yang
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Alberta Children's Hospital Research Institute, Calgary, AB, Canada
| | - Gonzalo I Cancino
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago, Chile.,Escuela de Biotecnología, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| |
Collapse
|
23
|
Toxoplasma gondii Infection in Patients with Psychiatric Disorders from Western Romania. Medicina (B Aires) 2022; 58:medicina58020208. [PMID: 35208532 PMCID: PMC8879735 DOI: 10.3390/medicina58020208] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/22/2022] [Accepted: 01/27/2022] [Indexed: 11/17/2022] Open
Abstract
Background and Objectives: High rates of infection with Toxoplasma gondii were found in psychiatric patients globally. In this study, we assessed for the first time the prevalence of T. gondii infection in psychiatric patients and healthy individuals with no known psychiatric disorders in Western Romania. Materials and Methods: The presence of specific IgG anti-T. gondii antibodies was evaluated in 308 psychiatric patients and 296 control subjects using a chemiluminescence assay. Results: Overall, the seroprevalence of IgG antibodies was higher in psychiatric patients (67.86%; 209/308), compared with the control group (54.05%; 160/296) (p < 0.001). Our results revealed a significantly higher prevalence of T. gondii antibodies among patients with schizophrenia (69.77%), organic (personality and behaviour) disorders (76.74%), and mental disorders concerning alcohol abuse (84.62%), compared with the control group (p = 0.009, p = 0.005, p = 0.043, respectively). Conclusions: This study provides new and important information on the seroprevalence of T. gondii in Romanian psychiatric patients and may serve for further scientific research regarding the status of T. gondii infection in patients with psychiatric disorders.
Collapse
|
24
|
Ginsenoside Rh2 reduces depression in offspring of mice with maternal toxoplasma infection during pregnancy by inhibiting microglial activation via the HMGB1/TLR4/NF-κB signaling pathway. J Ginseng Res 2022; 46:62-70. [PMID: 35035240 PMCID: PMC8753429 DOI: 10.1016/j.jgr.2021.04.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 04/09/2021] [Accepted: 04/09/2021] [Indexed: 02/08/2023] Open
Abstract
Background Maternal Toxoplasma gondii (T. gondii) infection during pregnancy has been associated with various mental illnesses in the offspring. Ginsenoside Rh2 (GRh2) is a major bioactive compound obtained from ginseng that has an anti-T. gondii effect and attenuates microglial activation through toll-like receptor 4 (TLR4)/nuclear factor-kappa B (NF-κB) signaling pathway. GRh2 also alleviated tumor-associated or lipopolysaccharide-induced depression. However, the effects and potential mechanisms of GRh2 on depression-like behavior in mouse offspring caused by maternal T. gondii infection during pregnancy have not been investigated. Methods We examined GRh2 effects on the depression-like behavior in mouse offspring, caused by maternal T. gondii infection during pregnancy, by measuring depression-like behaviors and assaying parameters at the neuronal and molecular level. Results We showed that GRh2 significantly improved behavioral measures: sucrose consumption, forced swim time and tail suspended immobility time of their offspring. These corresponded with increased tissue concentrations of 5-hydroxytryptamine and dopamine, and attenuated indoleamine 2,3-dioxygenase or enhanced tyrosine hydroxylase expression in the prefrontal cortex. GRh2 ameliorated neuronal damage in the prefrontal cortex. Molecular docking results revealed that GRh2 binds strongly to both TLR4 and high mobility group box 1 (HMGB1). Conclusion This study demonstrated that GRh2 ameliorated the depression-like behavior in mouse offspring of maternal T. gondii infection during pregnancy by attenuating the excessive activation of microglia and neuroinflammation through the HMGB1/TLR4/NF-κB signaling pathway. It suggests that GRh2 could be considered a potential therapy in preventing and treating psychiatric disorders in the offspring mice of mothers with prenatal exposure to T. gondii infection.
Collapse
|
25
|
Abstract
Depression and psychosis have a developmental component to their origin. Epidemiologic evidence, which we synthesize in this nonsystematic review, suggests that early-life infection, inflammation, and metabolic alterations could play a role in the etiology of these psychiatric disorders. The risk of depression and psychosis is associated with prenatal maternal and childhood infections, which could be mediated by impaired neurodevelopment. Evidence suggests linear dose-response associations between elevated concentrations of circulating inflammatory markers in childhood, particularly the inflammatory cytokine interleukin 6, and the risk for depression and psychosis subsequently in early adulthood. Childhood inflammatory markers are also associated with persistence of depressive symptoms subsequently in adolescence and early adulthood. Developmental trajectories reflecting persistently high insulin levels during childhood and adolescence are associated with a higher risk of psychosis in adulthood, whereas increased adiposity during and after puberty is associated with the risk of depression. Together, these findings suggest that higher levels of infection, inflammation, and metabolic alterations commonly seen in people with depression and psychosis could be a cause for, rather than simply a consequence of, these disorders. Therefore, early-life immuno-metabolic alterations, as well as factors influencing these alterations such as adversity or maltreatment, could represent targets for prevention of these psychiatric disorders. Inflammation could also be an important treatment target for depression and psychosis. The field requires further research to examine sensitive periods when exposure to such immuno-metabolic alterations is most harmful. Interventional studies are also needed to test the potential usefulness of targeting early-life immuno-metabolic alterations for preventing adult depression and psychosis.
Collapse
|
26
|
Handunnetthi L, Saatci D, Hamley JC, Knight JC. Maternal immune activation downregulates schizophrenia genes in the foetal mouse brain. Brain Commun 2021; 3:fcab275. [PMID: 34859219 PMCID: PMC8633770 DOI: 10.1093/braincomms/fcab275] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 10/04/2021] [Accepted: 10/12/2021] [Indexed: 12/02/2022] Open
Abstract
Susceptibility to schizophrenia is mediated by genetic and environmental risk factors. Maternal immune activation by infections during pregnancy is hypothesized to be a key environmental risk factor. However, little is known about how maternal immune activation contributes to schizophrenia pathogenesis. In this study, we investigated if maternal immune activation influences the expression of genes associated with schizophrenia in foetal mouse brains. We found that two sets of schizophrenia genes were downregulated more than expected by chance in the foetal mouse brain following maternal immune activation, namely those genes associated with schizophrenia through genome-wide association study (fold change = 1.93, false discovery rate = 4 × 10-4) and downregulated genes in adult schizophrenia brains (fold change = 1.51, false discovery rate = 4 × 10-10). We found that these genes mapped to key biological processes, such as neuronal cell adhesion. We also identified cortical excitatory neurons and inhibitory interneurons as the most vulnerable cell types to the deleterious effects of this interaction. Subsequently, we used gene expression information from herpes simplex virus 1 infection of neuronal precursor cells as orthogonal evidence to support our findings and to demonstrate that schizophrenia-associated cell adhesion genes, PCDHA2, PCDHA3 and PCDHA5, were downregulated following herpes simplex virus 1 infection. Collectively, our results provide novel evidence for a link between genetic and environmental risk factors in schizophrenia pathogenesis. These findings carry important implications for early preventative strategies in schizophrenia.
Collapse
Affiliation(s)
- Lahiru Handunnetthi
- Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 9DU, UK
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
| | - Defne Saatci
- Nuffield Department of Primary Care Health Sciences, Radcliffe Observatory Quarter, University of Oxford, Oxford OX2 6GG, UK
| | - Joseph C Hamley
- Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 9DU, UK
| | - Julian C Knight
- Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 9DU, UK
| |
Collapse
|
27
|
Yolken RH. Maternal Inflammation in Pregnancy: Setting the Pattern for Brain Development during Infancy and Beyond. J Pediatr 2021; 238:13-15. [PMID: 34126122 DOI: 10.1016/j.jpeds.2021.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 06/06/2021] [Indexed: 11/30/2022]
Affiliation(s)
- Robert H Yolken
- Stanley Division of Neurovirology, Johns Hopkins School of Medicine, Baltimore, Maryland.
| |
Collapse
|
28
|
Virus MA, Ehrhorn EG, Lui LM, Davis PH. Neurological and Neurobehavioral Disorders Associated with Toxoplasma gondii Infection in Humans. J Parasitol Res 2021; 2021:6634807. [PMID: 34712493 PMCID: PMC8548174 DOI: 10.1155/2021/6634807] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 09/15/2021] [Indexed: 01/17/2023] Open
Abstract
The intracellular parasite Toxoplasma gondii is estimated to infect up to 30% of the world population, leading to lifelong chronic infection of the brain and muscle tissue. Although most latent T. gondii infections in humans have traditionally been considered asymptomatic, studies in rodents suggest phenotypic neurological changes are possible. Consequently, several studies have examined the link between T. gondii infection and diseases such as schizophrenia, epilepsy, depression, bipolar disorder, dysphoria, Alzheimer's disease, Parkinson's disease, and obsessive-compulsive disorder (OCD). To date, there is varying evidence of the relationship of T. gondii to these human neurological or neurobehavioral disorders. A thorough review of T. gondii literature was conducted to highlight and summarize current findings. We found that schizophrenia was most frequently linked to T. gondii infection, while sleep disruption showed no linkage to T. gondii infection, and other conditions having mixed support for a link to T. gondii. However, infection as a cause of human neurobehavioral disease has yet to be firmly established.
Collapse
Affiliation(s)
- Maxwell A. Virus
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, USA
| | - Evie G. Ehrhorn
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, USA
| | - LeeAnna M. Lui
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, USA
| | - Paul H. Davis
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, USA
| |
Collapse
|
29
|
Hameete BC, Fernández-Calleja JM, de Groot MW, Oppewal TR, Tiemessen MM, Hogenkamp A, de Vries RB, Groenink L. The poly(I:C)-induced maternal immune activation model; a systematic review and meta-analysis of cytokine levels in the offspring. Brain Behav Immun Health 2021; 11:100192. [PMID: 34589729 PMCID: PMC8474626 DOI: 10.1016/j.bbih.2020.100192] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 12/19/2022] Open
Abstract
The maternal polyinosinic:polycytidylic acid (poly(I:C)) animal model is frequently used to study how maternal immune activation may impact neuro development in the offspring. Here, we present the first systematic review and meta-analysis on the effects of maternal poly(I:C) injection on immune mediators in the offspring and provide an openly accessible systematic map of the data including methodological characteristics. Pubmed and EMBASE were searched for relevant publications, yielding 45 unique papers that met inclusion criteria. We extracted data on immune outcomes and methodological characteristics, and assessed the risk of bias. The descriptive summary showed that most studies reported an absence of effect, with an equal number of studies reporting an increase or decrease in the immune mediator being studied. Meta-analysis showed increased IL-6 concentrations in the offspring of poly(I:C) exposed mothers. This effect appeared larger prenatally than post-weaning. Furthermore, poly(I:C) administration during mid-gestation was associated with higher IL-6 concentrations in the offspring. Maternal poly(I:C) induced changes in IL-1β, Il-10 and TNF-α concentrations were small and could not be associated with age of offspring, gestational period or sampling location. Finally, quality of reporting of potential measures to minimize bias was low, which stresses the importance of adherence to publication guidelines. Since neurodevelopmental disorders in humans tend to be associated with lifelong changes in cytokine concentrations, the absence of these effects as identified in this systematic review may suggest that combining the model with other etiological factors in future studies may provide further insight in the mechanisms through which maternal immune activation affects neurodevelopment. Long-term effects of maternal poly(I:C) on immune mediators in the offspring appear limited. Prenatal measurements and mid gestation poly(I:C) injection are associated with increases in IL-6 concentrations. Variety in methodological conduct hampers identification of key elements that affect cytokine concentrations. The quality of reporting of potential measures to minimize bias is poor.
Collapse
Affiliation(s)
- Bart C. Hameete
- Department of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, the Netherlands
| | - José M.S. Fernández-Calleja
- Department of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, the Netherlands
| | - Martje W.G.D.M. de Groot
- Department of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, the Netherlands
| | - Titia Rixt Oppewal
- University College Utrecht (UCU), Campusplein 1, Utrecht, 3584 ED, the Netherlands
- Stratingh Institute for Chemistry, University of Groningen, Nijenborgh 4, Groningen, 9747 AG, the Netherlands
| | - Machteld M. Tiemessen
- Research & Innovation, GCoE Immunology, Danone Nutricia Research, Uppsalalaan 12, Utrecht, 3584 CT, the Netherlands
| | - Astrid Hogenkamp
- Department of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, the Netherlands
| | - Rob B.M. de Vries
- SYstematic Review Center for Laboratory (Animal) Experimentation, Department for Health Evidence, Radboud University Medical Center, Geert Grooteplein zuid 10, Nijmegen, 6525 GA, the Netherlands
| | - Lucianne Groenink
- Department of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, the Netherlands
- Corresponding author.
| |
Collapse
|
30
|
Klein-Petersen AW, Köhler-Forsberg O, Benros ME. Infections, antibiotic treatment and the Microbiome in relation to schizophrenia. Schizophr Res 2021; 234:71-77. [PMID: 31859119 DOI: 10.1016/j.schres.2019.11.033] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 11/19/2019] [Accepted: 11/22/2019] [Indexed: 12/20/2022]
Abstract
Schizophrenia is a heterogeneous disorder with several potential pathophysiological mechanisms, including immune activation. Infections have been identified as a significant contributing risk factor for schizophrenia; this association is reviewed together with the potential impact of antibiotic treatment and alterations of the intestinal microbiota. Both infections and the treatment with antibiotics may alter the composition of the gut microbiota, causing dysbiosis, which in animal studies has been associated with alterations of behavior. Of the few studies that have been conducted on humans, some have suggested alterations in the microbial composition of individuals with schizophrenia compared to healthy controls, albeit with conflicting results. Recently, increased attention has emerged regarding potential adverse effects from antibiotics, as a number of these have been associated with an increased risk of psychotic episodes. Particularly, the fluoroquinolones have been associated with neurotoxic adverse events. The association between schizophrenia and infections, antibiotic treatment and dysbiosis, may be an epiphenomenon, which could be explained by other confounding factors. However, these associations could be causal and could therefore be important risk factors in a subgroup of patients. Large-scale well-matched longitudinal studies are needed with measurements of immune markers from multiple biological samples, ranging from material close to the brain, as cerebrospinal fluid and brain-scans targeting neuroinflammation, to analysis of blood and intestinal microbiota. This would help to obtain more definite results on the association between infections, immune components and microbiota alterations in relation to schizophrenia.
Collapse
Affiliation(s)
| | - Ole Köhler-Forsberg
- Mental Health Centre Copenhagen, Copenhagen University Hospital, Denmark; Psychosis Research Unit, Aarhus University Hospital - Psychiatry, Denmark; Department of Clinical Medicine, Aarhus University, Denmark
| | - Michael E Benros
- Mental Health Centre Copenhagen, Copenhagen University Hospital, Denmark.
| |
Collapse
|
31
|
Maternal infection in gestation increases the risk of non-affective psychosis in offspring: a meta-analysis. J Psychiatr Res 2021; 139:125-131. [PMID: 34058651 DOI: 10.1016/j.jpsychires.2021.05.039] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/25/2021] [Accepted: 05/20/2021] [Indexed: 01/07/2023]
Abstract
Maternal infection is thought to increase the risk of non-affective psychosis including schizophrenia. However, observational studies have produced conflicting results and little is known about the importance of timing of infection in mediating subsequent risk. In this study, we carried out a meta-analysis of observational studies to investigate the risk of maternal infection and subsequent risk of non-affective psychosis. Using seven cohort studies, we found that maternal infection during gestation increased the risk of non-affective psychosis [relative risk (RR): 1.28 (95% CI:1.05-1.57, p = 0.02, I2 = 36%)]. A subgroup analysis identified that there was greater risk for schizophrenia alone [RR: 1.65 (95% CI:1.23-2.22, p = 0.0008, I2 = 0%)]. In addition, infection during the second trimester resulted in increased risk [RR: 1.63 (95% CI:1.07-2.48, p = 0.02, I2 = 7%)], whilst risk during the first and third trimesters did not meet statistical significance. This study highlights maternal infection in gestation as an important environmental risk factor for non-affective psychosis and our findings carry important implications for future disease prevention strategies.
Collapse
|
32
|
St Clair D, Lang B. Schizophrenia: a classic battle ground of nature versus nurture debate. Sci Bull (Beijing) 2021; 66:1037-1046. [PMID: 36654248 DOI: 10.1016/j.scib.2021.01.032] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/29/2020] [Accepted: 10/13/2020] [Indexed: 01/20/2023]
Abstract
Much has been learned about the etiology and pathogenesis of schizophrenia since the term was first used by Eugene Bleuler over a century ago to describe one of the most important forms of major mental illness to affect mankind. Both nature and nurture feature prominently in our understanding of the genesis of the overall risk of developing schizophrenia. We now have a firm grasp of the broad structure of the genetic architecture and several key environmental risk factors have been identified and delineated. However, much of the heritability of schizophrenia remains unexplained and the reported environmental risk factors do not explain all the variances not attributable to genetic risk factors. The biggest problem at present is that our understanding of the causal mechanisms involved is still in its infancy. In this review, we describe the extent and limits of our knowledge of the specific genetic/constitutional and non-genetic/environmental factors that contribute to the overall risk of schizophrenia. We suggest novel methods may be required to understand the almost certainly immensely complex multi-level causal mechanisms that contribute to the generation of the schizophrenia phenotype.
Collapse
Affiliation(s)
- David St Clair
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, National Clinical Research Center for Mental Disorders, Changsha 410011, China; Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK; Bio-X Life Science Research Center, Shanghai Jiao Tong University, Shanghai 200030, China.
| | - Bing Lang
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, National Clinical Research Center for Mental Disorders, Changsha 410011, China; Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK.
| |
Collapse
|
33
|
Maternal infection exposure and the risk of psychosis in the offspring: A systematic review and meta-analysis. J Psychiatr Res 2021; 135:28-36. [PMID: 33445058 DOI: 10.1016/j.jpsychires.2020.12.065] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/20/2020] [Accepted: 12/29/2020] [Indexed: 12/27/2022]
Abstract
The relationship between maternal infection exposure and the risk of psychosis in the offspring is inconsistent. We systematically assessed this relationship. Unrestricted searches of the PubMed and Embase databases were conducted, with an end date of February 1, 2020, to identify relevant studies that met predetermined inclusion criteria. Random-effects models were adopted to estimate the overall relative risk. Twenty-three observational studies were included in the analysis. The results showed that mothers who had a history of infection during pregnancy experienced a significantly increased risk of developing psychosis in offspring (OR = 1.25, 95% confidence interval (CI): 1.1-1.41; P = 0.001). Sensitivity and subgroup analyses yielded consistent results. For specific pathogens, the risk of developing psychosis in offspring was increased among mothers with herpes simplex virus 2 (HSV-2) exposure (OR, 1.32; 95% CI, 1.09-1.6; P = 0.004). However, other maternal-specific pathogen exposures were not significantly associated with the risk of psychosis in offspring. No evidence of publication bias was observed. Although evidence of heterogeneity should be carefully evaluated, our findings suggest that maternal infection exposure may be associated with a greater risk of psychosis in the offspring.
Collapse
|
34
|
Paquin V, Lapierre M, Veru F, King S. Early Environmental Upheaval and the Risk for Schizophrenia. Annu Rev Clin Psychol 2021; 17:285-311. [PMID: 33544627 DOI: 10.1146/annurev-clinpsy-081219-103805] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Why does prenatal exposure to wars, natural disasters, urbanicity, or winter increase the risk for schizophrenia? Research from the last two decades has provided rich insight about the underlying chains of causation at play during environmental upheaval, from conception to early infancy. In this review, we appraise the evidence linking schizophrenia spectrum disorder to prenatal maternal stress, obstetric complications, early infections, and maternal nutrition and other lifestyle factors. We discuss putative mechanisms, including the maternal stress system, perinatal hypoxia, and maternal-offspring immune activation. We propose that gene-environment interactions, timing during development, and sex differentiate the neuropsychiatric outcomes. Future research should pursue the translation of animal studies to humans and the longitudinal associations between early exposures, intermediate phenotypes, and psychiatric disorders. Finally, to paint a comprehensive model of risk and to harness targets for prevention, we argue that risk factors should be situated within the individual's personal ecosystem.
Collapse
Affiliation(s)
- Vincent Paquin
- Department of Psychiatry, McGill University, Montréal, Québec H3A 1A1, Canada; .,Douglas Research Centre, Montréal, Québec H4H 1R3, Canada
| | - Mylène Lapierre
- Douglas Research Centre, Montréal, Québec H4H 1R3, Canada.,Department of Psychology, Université de Montréal, Montréal, Québec H2V 2S9, Canada
| | - Franz Veru
- Department of Psychiatry, McGill University, Montréal, Québec H3A 1A1, Canada; .,Douglas Research Centre, Montréal, Québec H4H 1R3, Canada
| | - Suzanne King
- Department of Psychiatry, McGill University, Montréal, Québec H3A 1A1, Canada; .,Douglas Research Centre, Montréal, Québec H4H 1R3, Canada.,Department of Psychology, Université de Montréal, Montréal, Québec H2V 2S9, Canada
| |
Collapse
|
35
|
Choudhury Z, Lennox B. Maternal Immune Activation and Schizophrenia-Evidence for an Immune Priming Disorder. Front Psychiatry 2021; 12:585742. [PMID: 33679468 PMCID: PMC7925413 DOI: 10.3389/fpsyt.2021.585742] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 01/25/2021] [Indexed: 12/13/2022] Open
Abstract
Schizophrenia is a complex neurodevelopmental disorder affecting around 19. 8 million people worldwide. The etiology of the disorder is due to many interacting genetic and environmental factors, with no one element causing the full spectrum of disease symptoms. Amongst these factors, maternal immune activation (MIA) acting during specific gestational timings has been implicated in increasing schizophrenia risk in offspring. Epidemiological studies have provided the rationale for this link with prevalence of maternal infection correlating to increased risk, but these studies have been unable to prove causality due to lack of control of confounding factors like genetic susceptibility and inability to identify specific cellular and molecular mechanisms. Animal models have proved significantly more useful in establishing the extent to which MIA can predispose an individual to schizophrenia, displaying how maternal infection alone can directly result in behavioral abnormalities in rodent offspring. Alongside information from genome wide association studies (GWAS), animal models have been able to identify the role of complement proteins, particularly C4, and display how alterations in this system can cause development of schizophrenia-associated neuropathology and behavior. This article will review the current literature in order to assess whether schizophrenia can, therefore, be viewed as an immune priming disorder.
Collapse
Affiliation(s)
- Zahra Choudhury
- The Queens College, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Belinda Lennox
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
36
|
Postolache TT, Wadhawan A, Rujescu D, Hoisington AJ, Dagdag A, Baca-Garcia E, Lowry CA, Okusaga OO, Brenner LA. Toxoplasma gondii, Suicidal Behavior, and Intermediate Phenotypes for Suicidal Behavior. Front Psychiatry 2021; 12:665682. [PMID: 34177652 PMCID: PMC8226025 DOI: 10.3389/fpsyt.2021.665682] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/30/2021] [Indexed: 12/27/2022] Open
Abstract
Within the general literature on infections and suicidal behavior, studies on Toxoplasma gondii (T. gondii) occupy a central position. This is related to the parasite's neurotropism, high prevalence of chronic infection, as well as specific and non-specific behavioral alterations in rodents that lead to increased risk taking, which are recapitulated in humans by T. gondii's associations with suicidal behavior, as well as trait impulsivity and aggression, mental illness and traffic accidents. This paper is a detailed review of the associations between T. gondii serology and suicidal behavior, a field of study that started 15 years ago with our publication of associations between T. gondii IgG serology and suicidal behavior in persons with mood disorders. This "legacy" article presents, chronologically, our primary studies in individuals with mood disorders and schizophrenia in Germany, recent attempters in Sweden, and in a large cohort of mothers in Denmark. Then, it reviews findings from all three meta-analyses published to date, confirming our reported associations and overall consistent in effect size [ranging between 39 and 57% elevation of odds of suicide attempt in T. gondii immunoglobulin (IgG) positives]. Finally, the article introduces certain links between T. gondii and biomarkers previously associated with suicidal behavior (kynurenines, phenylalanine/tyrosine), intermediate phenotypes of suicidal behavior (impulsivity, aggression) and state-dependent suicide risk factors (hopelessness/dysphoria, sleep impairment). In sum, an abundance of evidence supports a positive link between suicide attempts (but not suicidal ideation) and T. gondii IgG (but not IgM) seropositivity and serointensity. Trait impulsivity and aggression, endophenotypes of suicidal behavior have also been positively associated with T. gondii seropositivity in both the psychiatrically healthy as well as in patients with Intermittent Explosive Disorder. Yet, causality has not been demonstrated. Thus, randomized interventional studies are necessary to advance causal inferences and, if causality is confirmed, to provide hope that an etiological treatment for a distinct subgroup of individuals at an increased risk for suicide could emerge.
Collapse
Affiliation(s)
- Teodor T Postolache
- Department of Psychiatry, Mood and Anxiety Program, University of Maryland School of Medicine, Baltimore, MD, United States.,Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, CO, United States.,Mental Illness Research, Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 5, VA Capitol Health Care Network, Baltimore, MD, United States
| | - Abhishek Wadhawan
- Department of Psychiatry, Mood and Anxiety Program, University of Maryland School of Medicine, Baltimore, MD, United States.,Department of Psychiatry, Saint Elizabeth's Hospital, Washington, DC, United States
| | - Dan Rujescu
- Department of Psychiatry, Psychotherapy and Psychosomatics, University of Halle, Halle, Germany
| | - Andrew J Hoisington
- Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, CO, United States.,Department of Systems Engineering and Management, Air Force Institute of Technology, Dayton, OH, United States.,Department of Physical Medicine & Rehabilitation, University of Colorado, Anschutz Medical Campus, Aurora, CO, United States
| | - Aline Dagdag
- Department of Psychiatry, Mood and Anxiety Program, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Enrique Baca-Garcia
- Department of Psychiatry, Jimenez Diaz Foundation Hospital, Madrid, Spain.,Department of Psychiatry, Madrid Autonomous University, Madrid, Spain.,Department of Psychiatry, Rey Juan Carlos University Hospital, Móstoles, Spain.,Department of Psychiatry, General Hospital of Villalba, Madrid, Spain.,Department of Psychiatry, Infanta Elena University Hospital, Valdemoro, Spain.,Universidad Catolica del Maule, Talca, Chile.,Department of Psychiatry, Centre Hospitalier Universitaire de Nîmes, Nîmes, France
| | - Christopher A Lowry
- Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, CO, United States.,Department of Physical Medicine & Rehabilitation, University of Colorado, Anschutz Medical Campus, Aurora, CO, United States.,Department of Integrative Physiology, Center for Neuroscience, Center for Microbial Exploration, University of Colorado Boulder, Boulder, CO, United States
| | - Olaoluwa O Okusaga
- Department of Psychiatry, Mood and Anxiety Program, University of Maryland School of Medicine, Baltimore, MD, United States.,Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, United States.,Michael E DeBakey VA Medical Center, Houston, TX, United States
| | - Lisa A Brenner
- Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, CO, United States.,Department of Physical Medicine & Rehabilitation, University of Colorado, Anschutz Medical Campus, Aurora, CO, United States.,Department of Psychiatry & Neurology, University of Colorado, Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
37
|
Maternal immune activation induces sustained changes in fetal microglia motility. Sci Rep 2020; 10:21378. [PMID: 33288794 PMCID: PMC7721716 DOI: 10.1038/s41598-020-78294-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 11/20/2020] [Indexed: 12/17/2022] Open
Abstract
Maternal infection or inflammation causes abnormalities in brain development associated with subsequent cognitive impairment and in an increased susceptibility to schizophrenia and autism spectrum disorders. Maternal immune activation (MIA) and increases in serum cytokine levels mediates this association via effects on the fetal brain, and microglia can respond to maternal immune status, but consensus on how microglia may respond is lacking and no-one has yet examined if microglial process motility is impaired. In this study we investigated how MIA induced at two different gestational ages affected microglial properties at different developmental stages. Immune activation in mid-pregnancy increased IL-6 expression in embryonic microglia, but failed to cause any marked changes in morphology either at E18 or postnatally. In contrast MIA, particularly when induced earlier (at E12), caused sustained alterations in the patterns of microglial process motility and behavioral deficits. Our research has identified an important microglial property that is altered by MIA and which may contribute to the underlying pathophysiological mechanisms linking maternal immune status to subsequent risks for cognitive disease.
Collapse
|
38
|
Chaplin AB, Jones PB, Khandaker GM. Association between common early-childhood infection and subsequent depressive symptoms and psychotic experiences in adolescence: a population-based longitudinal birth cohort study. Psychol Med 2020; 52:1-11. [PMID: 33183379 PMCID: PMC9386436 DOI: 10.1017/s0033291720004080] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 09/09/2020] [Accepted: 10/14/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Childhood infections are associated with adult psychosis and depression, but studies of psychotic experiences (PEs) and depressive symptoms in childhood, adolescence, and early-adulthood are scarce. Previous studies have typically examined severe infections, but studies of common infections are also scarce. METHODS Using data from the Avon Longitudinal Study of Parents and Children (ALSPAC) birth cohort, we examined associations of the number of infections in childhood from age 1.5 to 7.5 years with depressive symptom scores at age 10, 13, 14, 17, 18, and 19 years, and with PEs at 12 and 18 years. We performed additional analysis using infection burden ('low' = 0-4 infections, 'medium' = 5-6, 'high' = 7-9, or 'very high' = 10-22 infections) as the exposure. RESULTS The risk set comprised 11 786 individuals with childhood infection data. Number of childhood infections was associated with depressive symptoms from age 10 (adjusted beta = 0.14; standard error (s.e.) = 0.04; p = <0.01) to 17 years (adjusted beta = 0.17; s.e. = 0.08; p = 0.04), and with PEs at age 12 (suspected/definite PEs: adjusted odds ratio (OR) = 1.18; 95% confidence interval (CI) = 1.09-1.27). These effect sizes were larger when the exposure was defined as very high infection burden (depressive symptoms age 17: adjusted beta = 0.79; s.e. = 0.29; p = 0.01; suspected/definite PEs at age 12: adjusted OR = 1.60; 95% CI = 1.25-2.05). Childhood infections were not associated with depressive/psychotic outcomes at age 18 or 19. CONCLUSIONS Common early-childhood infections are associated with depressive symptoms up to mid-adolescence and with PEs subsequently in childhood, but not with these outcomes in early-adulthood. These findings require replication including larger samples with outcomes in adulthood.
Collapse
Affiliation(s)
- Anna B. Chaplin
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Peter B. Jones
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Cambridgeshire and Peterborough NHS Foundation Trust, Cambridge, UK
| | - Golam M. Khandaker
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Cambridgeshire and Peterborough NHS Foundation Trust, Cambridge, UK
| |
Collapse
|
39
|
Fernandes SM, Dias AR, Miranda-Scippa Â. Association between exposure to toxoplasmosis and major psychiatric disorders: a systematic review. BRAZILIAN JOURNAL OF PSYCHIATRY 2020; 43:438-445. [PMID: 32965430 PMCID: PMC8352736 DOI: 10.1590/1516-4446-2020-0904] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 05/09/2020] [Indexed: 11/25/2022]
Abstract
Objective: To assess the association between exposure to toxoplasmosis and major psychiatric disorders through a systematic review of the literature. Methods: The literature review was performed in the MEDLINE, SciELO, and PsycINFO databases. To evaluate the quality of the studies included in the review, the Newcastle-Ottawa Scale was used. Results: Thirty-one studies were included, and the majority found an association between exposure to toxoplasmosis and schizophrenia or bipolar disorder (58.3 and 54.5% of the included papers, respectively), but not major depressive disorder. We found no significant difference in mean quality scores between studies that corroborated and contradicted the association hypothesis for either schizophrenia or bipolar disorder. All included papers were considered at least satisfactory according to the Newcastle-Ottawa Scale (total scores ≥ 6 out of 9). Conclusion: Although there was no association between exposure to toxoplasmosis and major depressive disorder, the results indicate an association with both bipolar disorder and schizophrenia, despite their heterogeneity. Further studies should be performed with more specific variables so that the nature of these relationships can be elucidated.
Collapse
Affiliation(s)
- Santiago M Fernandes
- Faculdade de Medicina da Bahia, Universidade Federal da Bahia (UFBA), Salvador, BA, Brazil.,Centro de Estudos de Transtornos de Humor e Ansiedade (CETHA), Complexo Hospitalar Universitário Professor Edgard Santos, UFBA, Salvador, BA, Brazil
| | - Alan R Dias
- Faculdade de Medicina da Bahia, Universidade Federal da Bahia (UFBA), Salvador, BA, Brazil.,Centro de Estudos de Transtornos de Humor e Ansiedade (CETHA), Complexo Hospitalar Universitário Professor Edgard Santos, UFBA, Salvador, BA, Brazil
| | - Ângela Miranda-Scippa
- Centro de Estudos de Transtornos de Humor e Ansiedade (CETHA), Complexo Hospitalar Universitário Professor Edgard Santos, UFBA, Salvador, BA, Brazil.,Programa de Pós-Graduação em Medicina e Saúde, UFBA, Salvador, BA, Brazil.,Departamento de Neurociências e Saúde Mental, Faculdade de Medicina da Bahia, UFBA, Salvador, BA, Brazil
| |
Collapse
|
40
|
Wegrzyn D, Manitz MP, Kostka M, Freund N, Juckel G, Faissner A. Poly I:C-induced maternal immune challenge reduces perineuronal net area and raises spontaneous network activity of hippocampal neurons in vitro. Eur J Neurosci 2020; 53:3920-3941. [PMID: 32757397 DOI: 10.1111/ejn.14934] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 07/08/2020] [Accepted: 07/23/2020] [Indexed: 12/17/2022]
Abstract
Activation of the maternal immune system (MIA) during gestation is linked to neuropsychiatric diseases like schizophrenia. While many studies address behavioural aspects, less is known about underlying cellular mechanisms. In the following study, BALB/c mice received intraperitoneal injections of polyinosinic-polycytidylic acid (Poly I:C) (20 µg/ml) or saline (0.9%) at gestation day (GD) 9.5 before hippocampal neurons were isolated and cultured from embryonic mice for further analysis. Interestingly, strongest effects were observed when the perineuronal net (PNN) wearing subpopulation of neurons was analysed. Here, a significant reduction of aggrecan staining intensity, area and soma size could be detected. Alterations of PNNs are often linked to neuropsychiatric diseases, changes in synaptic plasticity and in electrophysiology. Utilizing multielectrode array analysis (MEA), we observed a remarkable increase of the spontaneous network activity in neuronal networks after 21 days in vitro (DIV) when mother mice suffered a prenatal immune challenge. As PNNs are associated with GABAergic interneurons, our data indicate that this neuronal subtype might be stronger affected by a prenatal MIA. Degradation or damage of this subtype might cause the hyperexcitability observed in the whole network. In addition, embryonic neurons of the Poly I:C condition developed significantly shorter axons after five days in culture, while dendritic parameters and apoptosis rate remained unchanged. Structural analysis of synapse numbers revealed an increase of postsynaptic density 95 (PSD-95) puncta after 14 DIV and an increase of presynaptic vesicular glutamate transporter (vGlut) puncta after 21 DIV, while inhibitory synaptic proteins were not altered.
Collapse
Affiliation(s)
- David Wegrzyn
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Marie-Pierre Manitz
- Division of Experimental and Molecular Psychiatry, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL University Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Michael Kostka
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Nadja Freund
- Division of Experimental and Molecular Psychiatry, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL University Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Georg Juckel
- Department of Psychiatry, LWL University Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
41
|
Ajayi AM, Chidebe EO, Ben-Azu B, Umukoro S. Chrysophyllum albidum (African star apple) fruit-supplemented diet enhances cognitive functions and attenuates lipopolysaccharide-induced memory impairment, oxidative stress, and release of proinflammatory cytokines. NUTRIRE : REVISTA DE SOCIEDADE BRASILEIRA DE ALIMENTACAO E NUTRICAO = JOURNAL OF THE BRAZILIAN SOCIETY OF FOOD AND NUTRITION 2020; 45:20. [PMID: 38624427 PMCID: PMC7448960 DOI: 10.1186/s41110-020-00123-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 08/16/2020] [Indexed: 12/19/2022]
Abstract
Purpose Fruit-based supplement has an important role in protecting the brain against oxido-inflammatory stress. Chrysophyllum albidum fruit contained several phytonutrients that possess antioxidants and anti-inflammatory properties. Hence, this study investigated the effect of C. albidum fruit supplemented diet (CAFD) on cognitive functions and lipopolysaccharide (LPS)-induced memory impairment and oxido-inflammatory response in mice. Methods Mice were randomized into two experiments. Experiment 1 with naïve mice contained four groups (n = 6) while experiment 2 with LPS contains five groups (n = 6). Mice in experiments 1 and 2 were fed on CAFD (5%, 10%, and 20%) in naïve (6 weeks) and LPS (250 μg/kg, i.p.) in the 7th week, respectively. Cognitive performance was tested using Y-maze test (YMT) and novel object recognition test (NORT) in the naïve and LPS mice. Brain samples were obtained for determination of oxido-inflammatory parameters and acetylcholinesterase activity. Results The CAFD significantly enhanced cognitive performance in the YMT and NORT in naïve and LPS mice, as evidenced by increased % alternation and discrimination index, respectively. CAFD supplementation significantly reduced acetylcholinesterase enzyme activity while it attenuated depletion of reduced glutathione and catalase activities in brains of naive and LPS-treated animals. The CAFD significantly reduced LPS-induced increased malondialdehyde levels in mice brains. CAFD supplementation significantly attenuated LPS-induced pro-inflammatory cytokines (IL-6, TNF-α) in mice brains. Conclusion Chrysophyllum albidum fruit supplementation in diet enhances memory function and prevents cognitive deficits induced by LPS via mechanisms associated with inhibition of oxidative stress-related processes, acetylcholinesterase activity, and pro-inflammatory mediators.
Collapse
Affiliation(s)
- Abayomi Mayowa Ajayi
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, University of Ibadan, Ibadan, Oyo State Nigeria
| | - Emmanuel Oyinyechukwu Chidebe
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, University of Ibadan, Ibadan, Oyo State Nigeria
| | - Benneth Ben-Azu
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, University of Ibadan, Ibadan, Oyo State Nigeria
- Department of Pharmacology, Faculty of Basic Medical Sciences, PAMO University of Medical Sciences, Port Harcourt, Rivers State Nigeria
| | - Solomon Umukoro
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, University of Ibadan, Ibadan, Oyo State Nigeria
| |
Collapse
|
42
|
Nasirpour S, Kheirandish F, Fallahi S. Depression and Toxoplasma gondii infection: assess the possible relationship through a seromolecular case-control study. Arch Microbiol 2020; 202:2689-2695. [PMID: 32725599 DOI: 10.1007/s00203-020-01993-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 07/12/2020] [Accepted: 07/21/2020] [Indexed: 11/26/2022]
Abstract
Depression disorder is one of the most common psychological recognitions that characterized by sadness, low self-confidence, and disinterest in every activity. Considering evidence showing the effects of toxoplasmosis on the psychological disease, this study conducted to investigate the serological and molecular aspects of Toxoplasma gondii infection among patients with depression. In this study, after selecting the patients with depression and control groups under the supervision of a psychologist, the blood samples were collected and the serum samples and buffy coat were separated. The specific anti-Toxoplasma IgG antibodies in serum samples were evaluated using the commercial ELISA kit. Then the desired region of the Toxoplasma B1 gene was amplified using the specific primers. To confirm the specificity of primers to amplify the B1 gene of Toxoplasma, the extracted PCR product was sequenced. The overall prevalence of toxoplasmosis in patients with depression was 59.8 and 60.19% by ELISA and PCR, respectively. In the control group, the prevalence of Toxoplasma was 56.3 and 40.2% by serology and PCR. There was a significant correlation between the prevalence of toxoplasmosis and depression. Moreover, a significant difference was found between the variables of age, sex, kind of nutrition, level of education and toxoplasmosis among the two cases and control groups. The higher prevalence of Toxoplasma infection among patients with depression compared with the control group indicates the probable impact of this parasite on depression and exacerbates its symptoms, which requires special attention of specialist physicians and patient's relatives.
Collapse
Affiliation(s)
- Saber Nasirpour
- Department of Microbiology, Islamic Azad University, Farahan Branch, Farahan, Iran
| | - Farnaz Kheirandish
- Department of Parasitology and Mycology, Faculty of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran.
| | - Shirzad Fallahi
- Department of Parasitology and Mycology, Faculty of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran.
- Hepatitis Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran.
| |
Collapse
|
43
|
Preterm birth and sustained inflammation: consequences for the neonate. Semin Immunopathol 2020; 42:451-468. [PMID: 32661735 PMCID: PMC7508934 DOI: 10.1007/s00281-020-00803-2] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 06/24/2020] [Indexed: 12/15/2022]
Abstract
Almost half of all preterm births are caused or triggered by an inflammatory process at the feto-maternal interface resulting in preterm labor or rupture of membranes with or without chorioamnionitis (“first inflammatory hit”). Preterm babies have highly vulnerable body surfaces and immature organ systems. They are postnatally confronted with a drastically altered antigen exposure including hospital-specific microbes, artificial devices, drugs, nutritional antigens, and hypoxia or hyperoxia (“second inflammatory hit”). This is of particular importance to extremely preterm infants born before 28 weeks, as they have not experienced important “third-trimester” adaptation processes to tolerate maternal and self-antigens. Instead of a balanced adaptation to extrauterine life, the delicate co-regulation between immune defense mechanisms and immunosuppression (tolerance) to allow microbiome establishment is therefore often disturbed. Hence, preterm infants are predisposed to sepsis but also to several injurious conditions that can contribute to the onset or perpetuation of sustained inflammation (SI). This is a continuing challenge to clinicians involved in the care of preterm infants, as SI is regarded as a crucial mediator for mortality and the development of morbidities in preterm infants. This review will outline the (i) role of inflammation for short-term consequences of preterm birth and (ii) the effect of SI on organ development and long-term outcome.
Collapse
|
44
|
Lathe R, St Clair D. From conifers to cognition: Microbes, brain and behavior. GENES BRAIN AND BEHAVIOR 2020; 19:e12680. [PMID: 32515128 DOI: 10.1111/gbb.12680] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 05/12/2020] [Accepted: 05/29/2020] [Indexed: 12/25/2022]
Abstract
A diversity of bacteria, protozoans and viruses ("endozoites") were recently uncovered within healthy tissues including the human brain. By contrast, it was already recognized a century ago that healthy plants tissues contain abundant endogenous microbes ("endophytes"). Taking endophytes as an informative precedent, we overview the nature, prevalence, and role of endozoites in mammalian tissues, centrally focusing on the brain, concluding that endozoites are ubiquitous in diverse tissues. These passengers often remain subclinical, but they are not silent. We address their routes of entry, mechanisms of persistence, tissue specificity, and potential to cause long-term behavioral changes and/or immunosuppression in mammals, where rabies virus is the exemplar. We extend the discussion to Herpesviridae, Coronaviridae, and Toxoplasma, as well as to diverse bacteria and yeasts, and debate the advantages and disadvantages that endozoite infection might afford to the host and to the ecosystem. We provide a clinical perspective in which endozoites are implicated in neurodegenerative disease, anxiety/depression, and schizophrenia. We conclude that endozoites are instrumental in the delicate balance between health and disease, including age-related brain disease, and that endozoites have played an important role in the evolution of brain function and human behavior.
Collapse
Affiliation(s)
- Richard Lathe
- Division of Infection Medicine, University of Edinburgh Medical School, Edinburgh, UK
| | - David St Clair
- Institute of Medical Sciences, School of Medicine, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
45
|
Hoffman KW, Lee JJ, Corcoran CM, Kimhy D, Kranz TM, Malaspina D. Considering the Microbiome in Stress-Related and Neurodevelopmental Trajectories to Schizophrenia. Front Psychiatry 2020; 11:629. [PMID: 32719625 PMCID: PMC7350783 DOI: 10.3389/fpsyt.2020.00629] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 06/16/2020] [Indexed: 12/12/2022] Open
Abstract
Early life adversity and prenatal stress are consistently associated with an increased risk for schizophrenia, although the exact pathogenic mechanisms linking the exposures with the disease remain elusive. Our previous view of the HPA stress axis as an elegant but simple negative feedback loop, orchestrating adaptation to stressors among the hypothalamus, pituitary, and adrenal glands, needs to be updated. Research in the last two decades shows that important bidirectional signaling between the HPA axis and intestinal mucosa modulates brain function and neurochemistry, including effects on glucocorticoid hormones and brain-derived neurotrophic factor (BDNF). The intestinal microbiome in earliest life, which is seeded by the vaginal microbiome during delivery, programs the development of the HPA axis in a critical developmental window, determining stress sensitivity and HPA function as well as immune system development. The crosstalk between the HPA and the Microbiome Gut Brain Axis (MGBA) is particularly high in the hippocampus, the most consistently disrupted neural region in persons with schizophrenia. Animal models suggest that the MGBA remains influential on behavior and physiology across developmental stages, including the perinatal window, early childhood, adolescence, and young adulthood. Understanding the role of the microbiome on critical risk related stressors may enhance or transform of understanding of the origins of schizophrenia and offer new approaches to increase resilience against stress effects for preventing and treating schizophrenia.
Collapse
Affiliation(s)
- Kevin W. Hoffman
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jakleen J. Lee
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Cheryl M. Corcoran
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- James J. Peters VA Medical Center, Mental Illness Research, Education and Clinical Centers (MIRECC), New York, NY, United States
| | - David Kimhy
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- James J. Peters VA Medical Center, Mental Illness Research, Education and Clinical Centers (MIRECC), New York, NY, United States
| | - Thorsten M. Kranz
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt, Germany
| | - Dolores Malaspina
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
46
|
Dunn AL, Michie PT, Hodgson DM, Harms L. Adolescent cannabinoid exposure interacts with other risk factors in schizophrenia: A review of the evidence from animal models. Neurosci Biobehav Rev 2020; 116:202-220. [PMID: 32610181 DOI: 10.1016/j.neubiorev.2020.06.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 06/23/2020] [Accepted: 06/24/2020] [Indexed: 12/18/2022]
Abstract
Many factors and their interaction are linked to the aetiology of schizophrenia, leading to the development of animal models of multiple risk factors and adverse exposures. Differentiating between separate and combined effects for each factor could better elucidate schizophrenia pathology, and drive development of preventative strategies for high-load risk factors. An epidemiologically valid risk factor commonly associated with schizophrenia is adolescent cannabis use. The aim of this review is to evaluate how early-life adversity from various origins, in combination with adolescent cannabinoid exposure interact, and whether these interactions confer main, synergistic or protective effects in animal models of schizophrenia-like behavioural, cognitive and morphological alterations. Patterns emerge regarding which models show consistent synergistic or protective effects, particularly those models incorporating early-life exposure to maternal deprivation and maternal immune activation, and sex-specific effects are observed. It is evident that more research needs to be conducted to better understand the risks and alterations of interacting factors, with particular interest in sex differences, to better understand the translatability of these preclinical models to humans.
Collapse
Affiliation(s)
- Ariel L Dunn
- School of Psychology, Faculty of Science, University of Newcastle, Callaghan, NSW 2308, Australia; Priority Centre for Brain and Mental Health Research, University of Newcastle, Callaghan, NSW 2308, Australia.
| | - Patricia T Michie
- School of Psychology, Faculty of Science, University of Newcastle, Callaghan, NSW 2308, Australia; Priority Centre for Brain and Mental Health Research, University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia.
| | - Deborah M Hodgson
- School of Psychology, Faculty of Science, University of Newcastle, Callaghan, NSW 2308, Australia; Priority Centre for Brain and Mental Health Research, University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia.
| | - Lauren Harms
- Priority Centre for Brain and Mental Health Research, University of Newcastle, Callaghan, NSW 2308, Australia; School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia.
| |
Collapse
|
47
|
Haddad FL, Patel SV, Schmid S. Maternal Immune Activation by Poly I:C as a preclinical Model for Neurodevelopmental Disorders: A focus on Autism and Schizophrenia. Neurosci Biobehav Rev 2020; 113:546-567. [PMID: 32320814 DOI: 10.1016/j.neubiorev.2020.04.012] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 01/28/2020] [Accepted: 04/09/2020] [Indexed: 12/18/2022]
Abstract
Maternal immune activation (MIA) in response to a viral infection during early and mid-gestation has been linked through various epidemiological studies to a higher risk for the child to develop autism or schizophrenia-related symptoms.. This has led to the establishment of the pathogen-free poly I:C-induced MIA animal model for neurodevelopmental disorders, which shows relatively high construct and face validity. Depending on the experimental variables, particularly the timing of poly I:C administration, different behavioural and molecular phenotypes have been described that relate to specific symptoms of neurodevelopmental disorders such as autism spectrum disorder and/or schizophrenia. We here review and summarize epidemiological evidence for the effects of maternal infection and immune activation, as well as major findings in different poly I:C MIA models with a focus on poly I:C exposure timing, behavioural and molecular changes in the offspring, and characteristics of the model that relate it to autism spectrum disorder and schizophrenia.
Collapse
Affiliation(s)
- Faraj L Haddad
- Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada.
| | - Salonee V Patel
- Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada.
| | - Susanne Schmid
- Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada.
| |
Collapse
|
48
|
Bauman MD, Van de Water J. Translational opportunities in the prenatal immune environment: Promises and limitations of the maternal immune activation model. Neurobiol Dis 2020; 141:104864. [PMID: 32278881 DOI: 10.1016/j.nbd.2020.104864] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 03/03/2020] [Accepted: 04/02/2020] [Indexed: 12/15/2022] Open
Abstract
The prenatal environment, and in particular, the maternal-fetal immune environment, has emerged as a targeted area of research for central nervous system (CNS) diseases with neurodevelopmental origins. Converging evidence from both clinical and preclinical research indicates that changes in the maternal gestational immune environment can alter fetal brain development and increase the risk for certain neurodevelopmental disorders. Here we focus on the translational potential of one prenatal animal model - the maternal immune activation (MIA) model. This model stems from the observation that a subset of pregnant women who are exposed to infection during pregnancy have an increased risk of giving birth to a child who will later be diagnosed with a neurodevelopmental disorder, such as autism spectrum disorder (ASD) or schizophrenia (SZ). The preclinical MIA model provides a system in which to explore causal relationships, identify underlying neurobiological mechanisms, and, ultimately, develop novel therapeutic interventions and preventative strategies. In this review, we will highlight converging evidence from clinical and preclinical research that links changes in the maternal-fetal immune environment with lasting changes in offspring brain and behavioral development. We will then explore the promises and limitations of the MIA model as a translational tool to develop novel therapeutic interventions. As the translational potential of the MIA model has been the focus of several excellent review articles, here we will focus on what is perhaps the least well developed area of MIA model research - novel preventative strategies and therapeutic interventions.
Collapse
Affiliation(s)
- Melissa D Bauman
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, United States of America; California National Primate Research Center, University of California, Davis, United States of America; The MIND Institute, University of California, Davis, United States of America.
| | - Judy Van de Water
- The MIND Institute, University of California, Davis, United States of America; Rheumatology/Allergy and Clinical Immunology, University of California, Davis, United States of America
| |
Collapse
|
49
|
Tyebji S, Hannan AJ, Tonkin CJ. Pathogenic Infection in Male Mice Changes Sperm Small RNA Profiles and Transgenerationally Alters Offspring Behavior. Cell Rep 2020; 31:107573. [DOI: 10.1016/j.celrep.2020.107573] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 03/01/2020] [Accepted: 04/02/2020] [Indexed: 12/12/2022] Open
|
50
|
Mok SWF, Wong VKW, Lo HH, de Seabra Rodrigues Dias IR, Leung ELH, Law BYK, Liu L. Natural products-based polypharmacological modulation of the peripheral immune system for the treatment of neuropsychiatric disorders. Pharmacol Ther 2020; 208:107480. [DOI: 10.1016/j.pharmthera.2020.107480] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 12/31/2019] [Indexed: 02/06/2023]
|