51
|
Lech MA, Leśkiewicz M, Kamińska K, Rogóż Z, Lorenc-Koci E. Glutathione Deficiency during Early Postnatal Development Causes Schizophrenia-Like Symptoms and a Reduction in BDNF Levels in the Cortex and Hippocampus of Adult Sprague-Dawley Rats. Int J Mol Sci 2021; 22:ijms22126171. [PMID: 34201038 PMCID: PMC8229148 DOI: 10.3390/ijms22126171] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 12/11/2022] Open
Abstract
Growing body of evidence points to dysregulation of redox status in the brain as an important factor in the pathogenesis of schizophrenia. The aim of our study was to evaluate the effects of l-buthionine-(S,R)-sulfoximine (BSO), a glutathione (GSH) synthesis inhibitor, and 1-[2-Bis(4-fluorophenyl)methoxy]ethyl]-4-(3-phenylpropyl)piperazine dihydrochloride (GBR 12909), a dopamine reuptake inhibitor, given alone or in combination, to Sprague–Dawley pups during early postnatal development (p5–p16), on the time course of the onset of schizophrenia-like behaviors, and on the expression of brain-derived neurotrophic factor (BDNF) mRNA and its protein in the prefrontal cortex (PFC) and hippocampus (HIP) during adulthood. BSO administered alone decreased the levels of BDNF mRNA and its protein both in the PFC and HIP. Treatment with the combination of BSO + GBR 12909 also decreased BDNF mRNA and its protein in the PFC, but in the HIP, only the level of BDNF protein was decreased. Schizophrenia-like behaviors in rats were assessed at three time points of adolescence (p30, p42–p44, p60–p62) and in early adulthood (p90–p92) using the social interaction test, novel object recognition test, and open field test. Social and cognitive deficits first appeared in the middle adolescence stage and continued to occur into adulthood, both in rats treated with BSO alone or with the BSO + GBR 12909 combination. Behavior corresponding to positive symptoms in humans occurred in the middle adolescence period, only in rats treated with BSO + GBR 12909. Only in the latter group, amphetamine exacerbated the existing positive symptoms in adulthood. Our data show that rats receiving the BSO + GBR 12909 combination in the early postnatal life reproduced virtually all symptoms observed in patients with schizophrenia and, therefore, can be considered a valuable neurodevelopmental model of this disease.
Collapse
Affiliation(s)
- Marta Anna Lech
- Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, 31-343 Kraków, Poland; (M.A.L.); (K.K.); (Z.R.)
| | - Monika Leśkiewicz
- Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, 31-343 Kraków, Poland;
| | - Kinga Kamińska
- Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, 31-343 Kraków, Poland; (M.A.L.); (K.K.); (Z.R.)
| | - Zofia Rogóż
- Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, 31-343 Kraków, Poland; (M.A.L.); (K.K.); (Z.R.)
| | - Elżbieta Lorenc-Koci
- Department of Neuro-Psychopharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, 31-343 Kraków, Poland
- Correspondence: ; Tel.: +48-126-623-272
| |
Collapse
|
52
|
Testai L, Martelli A, Flori L, Cicero AFG, Colletti A. Coenzyme Q 10: Clinical Applications beyond Cardiovascular Diseases. Nutrients 2021; 13:1697. [PMID: 34067632 PMCID: PMC8156424 DOI: 10.3390/nu13051697] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/29/2021] [Accepted: 05/14/2021] [Indexed: 02/07/2023] Open
Abstract
Coenzyme Q10 (CoQ10) is an essential cofactor in oxidative phosphorylation (OXPHOS), present in mitochondria and cell membranes in reduced and oxidized forms. Acting as an energy transfer molecule, it occurs in particularly high levels in the liver, heart, and kidneys. CoQ10 is also an anti-inflammatory and antioxidant agent able to prevent the damage induced by free radicals and the activation of inflammatory signaling pathways. In this context, several studies have shown the possible inverse correlation between the blood levels of CoQ10 and some disease conditions. Interestingly, beyond cardiovascular diseases, CoQ10 is involved also in neuronal and muscular degenerative diseases, in migraine and in cancer; therefore, the supplementation with CoQ10 could represent a viable option to prevent these and in some cases might be used as an adjuvant to conventional treatments. This review is aimed to summarize the clinical applications regarding the use of CoQ10 in migraine, neurodegenerative diseases (including Parkinson and Alzheimer diseases), cancer, or degenerative muscle disorders (such as multiple sclerosis and chronic fatigue syndrome), analyzing its effect on patients' health and quality of life.
Collapse
Affiliation(s)
- Lara Testai
- Department of Pharmacy, University of Pisa, 56120 Pisa, Italy; (A.M.); (L.F.)
- Interdepartmental Research Centre ‘‘Nutraceuticals and Food for Health (NUTRAFOOD)’’, University of Pisa, 56120 Pisa, Italy
- Interdepartmental Research Centre of Ageing, Biology and Pathology, University of Pisa, 56120 Pisa, Italy
| | - Alma Martelli
- Department of Pharmacy, University of Pisa, 56120 Pisa, Italy; (A.M.); (L.F.)
- Interdepartmental Research Centre ‘‘Nutraceuticals and Food for Health (NUTRAFOOD)’’, University of Pisa, 56120 Pisa, Italy
- Interdepartmental Research Centre of Ageing, Biology and Pathology, University of Pisa, 56120 Pisa, Italy
| | - Lorenzo Flori
- Department of Pharmacy, University of Pisa, 56120 Pisa, Italy; (A.M.); (L.F.)
| | - Arrigo F. G. Cicero
- Medical and Surgical Sciences Department, University of Bologna, 40138 Bologna, Italy;
- Italian Nutraceutical Society (SINut), 40138 Bologna, Italy;
| | - Alessandro Colletti
- Italian Nutraceutical Society (SINut), 40138 Bologna, Italy;
- Department of Science and Drug Technology, University of Turin, 10125 Turin, Italy
| |
Collapse
|
53
|
Parellada E, Gassó P. Glutamate and microglia activation as a driver of dendritic apoptosis: a core pathophysiological mechanism to understand schizophrenia. Transl Psychiatry 2021; 11:271. [PMID: 33958577 PMCID: PMC8102516 DOI: 10.1038/s41398-021-01385-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 04/09/2021] [Accepted: 04/20/2021] [Indexed: 02/03/2023] Open
Abstract
Schizophrenia disorder remains an unsolved puzzle. However, the integration of recent findings from genetics, molecular biology, neuroimaging, animal models and translational clinical research offers evidence that the synaptic overpruning hypothesis of schizophrenia needs to be reassessed. During a critical period of neurodevelopment and owing to an imbalance of excitatory glutamatergic pyramidal neurons and inhibitory GABAergic interneurons, a regionally-located glutamate storm might occur, triggering excessive dendritic pruning with the activation of local dendritic apoptosis machinery. The apoptotic loss of dendritic spines would be aggravated by microglia activation through a recently described signaling system from complement abnormalities and proteins of the MHC, thus implicating the immune system in schizophrenia. Overpruning of dendritic spines coupled with aberrant synaptic plasticity, an essential function for learning and memory, would lead to brain misconnections and synaptic inefficiency underlying the primary negative symptoms and cognitive deficits of schizophrenia. This driving hypothesis has relevant therapeutic implications, including the importance of pharmacological interventions during the prodromal phase or the transition to psychosis, targeting apoptosis, microglia cells or the glutamate storm. Future research on apoptosis and brain integrity should combine brain imaging, CSF biomarkers, animal models and cell biology.
Collapse
Affiliation(s)
- Eduard Parellada
- Barcelona Clínic Schizophrenia Unit (BCSU). Institute of Neuroscience, Hospital Clínic of Barcelona, University of Barcelona, Barcelona, Catalonia, Spain.
- The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Catalonia, Spain.
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.
| | - Patricia Gassó
- Barcelona Clínic Schizophrenia Unit (BCSU). Institute of Neuroscience, Hospital Clínic of Barcelona, University of Barcelona, Barcelona, Catalonia, Spain
- The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Catalonia, Spain
- Department of Basic Clinical Practice, Unit of Pharmacology, University of Barcelona, Barcelona, Catalonia, Spain
| |
Collapse
|
54
|
Pan Y, Dempster K, Jeon P, Théberge J, Khan AR, Palaniyappan L. Acute conceptual disorganization in untreated first-episode psychosis: a combined magnetic resonance spectroscopy and diffusion imaging study of the cingulum. J Psychiatry Neurosci 2021; 46:E337-E346. [PMID: 33904669 PMCID: PMC8327974 DOI: 10.1503/jpn.200167] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Disorganized thinking is a core feature of acute psychotic episodes that is linked to social and vocational functioning. Several lines of evidence implicate disrupted cognitive control, excitatory overdrive and oxidative stress relating to the anterior cingulate cortex as mechanisms of conceptual disorganization (CD). We examined 3 candidate mechanistic markers related to CD in firstepisode psychosis: glutamate excess, cortical antioxidant (glutathione) status and the integrity of the cingulum bundle that connects regions implicated in cognitive control. METHODS We used fractional anisotropy maps from 7 T diffusion-weighted imaging to investigate the bilateral cingulum based on a probabilistic white matter atlas. We compared high CD, low CD and healthy control groups and performed probabilistic fibre tracking from the identified clusters (regions of interest within the cingulum) to the rest of the brain. We quantified glutamate and glutathione using magnetic resonance spectroscopy (MRS) in the dorsal anterior cingulate cortex. RESULTS We found a significant fractional anisotropy reduction in a cluster in the left cingulum in the high CD group compared to the low CD group (Cohen's d = 1.39; p < 0.001) and controls (Cohen's d = 0.86; p = 0.009). Glutamate levels did not vary among groups, but glutathione levels were higher in the high CD group than in the low CD group. We also found higher glutathione related to lower fractional anisotropy in the cingulum cluster in the high CD group. LIMITATIONS The MRS measures of glutamine were highly uncertain, and MRS was acquired from a single voxel only. CONCLUSION Acute CD relates to indicators of oxidative stress, as well as reduced white matter integrity of the cingulum, but not to MRI-based glutamatergic excess. We propose that both oxidative imbalance and structural dysconnectivity underlie acute disorganization.
Collapse
Affiliation(s)
- Yunzhi Pan
- From the Department of Psychiatry, Second Xiangya Hospital of Central South University, Changsha, Hunan, China (Pan); the Robarts Research Institute, University of Western Ontario, London, Ont., Canada (Pan, Khan, Palaniyappan); the Lawson Health Research Institute, London, Ont., Canada (Théberge, Palaniyappan); the Department of Medical Biophysics, University of Western Ontario, London, Ont., Canada (Jeon, Théberge, Khan, Palaniyappan); the Department of Psychiatry, University of Western Ontario, London, Ont., Canada (Palaniyappan, Théberge); the Department of Psychiatry, Dalhousie University, Halifax, NS, Canada (Dempster); the China National Clinical Research Center on Mental Disorders (Xiangya), Changsha, Hunan, China (Pan); the China National Technology Institute on Mental Disorders, Changsha, Hunan, China (Pan); the Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan, China (Pan); and the Institute of Mental Health of Second Xiangya Hospital, Central South University, Changsha, Hunan, China (Pan)
| | - Kara Dempster
- From the Department of Psychiatry, Second Xiangya Hospital of Central South University, Changsha, Hunan, China (Pan); the Robarts Research Institute, University of Western Ontario, London, Ont., Canada (Pan, Khan, Palaniyappan); the Lawson Health Research Institute, London, Ont., Canada (Théberge, Palaniyappan); the Department of Medical Biophysics, University of Western Ontario, London, Ont., Canada (Jeon, Théberge, Khan, Palaniyappan); the Department of Psychiatry, University of Western Ontario, London, Ont., Canada (Palaniyappan, Théberge); the Department of Psychiatry, Dalhousie University, Halifax, NS, Canada (Dempster); the China National Clinical Research Center on Mental Disorders (Xiangya), Changsha, Hunan, China (Pan); the China National Technology Institute on Mental Disorders, Changsha, Hunan, China (Pan); the Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan, China (Pan); and the Institute of Mental Health of Second Xiangya Hospital, Central South University, Changsha, Hunan, China (Pan)
| | - Peter Jeon
- From the Department of Psychiatry, Second Xiangya Hospital of Central South University, Changsha, Hunan, China (Pan); the Robarts Research Institute, University of Western Ontario, London, Ont., Canada (Pan, Khan, Palaniyappan); the Lawson Health Research Institute, London, Ont., Canada (Théberge, Palaniyappan); the Department of Medical Biophysics, University of Western Ontario, London, Ont., Canada (Jeon, Théberge, Khan, Palaniyappan); the Department of Psychiatry, University of Western Ontario, London, Ont., Canada (Palaniyappan, Théberge); the Department of Psychiatry, Dalhousie University, Halifax, NS, Canada (Dempster); the China National Clinical Research Center on Mental Disorders (Xiangya), Changsha, Hunan, China (Pan); the China National Technology Institute on Mental Disorders, Changsha, Hunan, China (Pan); the Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan, China (Pan); and the Institute of Mental Health of Second Xiangya Hospital, Central South University, Changsha, Hunan, China (Pan)
| | - Jean Théberge
- From the Department of Psychiatry, Second Xiangya Hospital of Central South University, Changsha, Hunan, China (Pan); the Robarts Research Institute, University of Western Ontario, London, Ont., Canada (Pan, Khan, Palaniyappan); the Lawson Health Research Institute, London, Ont., Canada (Théberge, Palaniyappan); the Department of Medical Biophysics, University of Western Ontario, London, Ont., Canada (Jeon, Théberge, Khan, Palaniyappan); the Department of Psychiatry, University of Western Ontario, London, Ont., Canada (Palaniyappan, Théberge); the Department of Psychiatry, Dalhousie University, Halifax, NS, Canada (Dempster); the China National Clinical Research Center on Mental Disorders (Xiangya), Changsha, Hunan, China (Pan); the China National Technology Institute on Mental Disorders, Changsha, Hunan, China (Pan); the Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan, China (Pan); and the Institute of Mental Health of Second Xiangya Hospital, Central South University, Changsha, Hunan, China (Pan)
| | - Ali R Khan
- From the Department of Psychiatry, Second Xiangya Hospital of Central South University, Changsha, Hunan, China (Pan); the Robarts Research Institute, University of Western Ontario, London, Ont., Canada (Pan, Khan, Palaniyappan); the Lawson Health Research Institute, London, Ont., Canada (Théberge, Palaniyappan); the Department of Medical Biophysics, University of Western Ontario, London, Ont., Canada (Jeon, Théberge, Khan, Palaniyappan); the Department of Psychiatry, University of Western Ontario, London, Ont., Canada (Palaniyappan, Théberge); the Department of Psychiatry, Dalhousie University, Halifax, NS, Canada (Dempster); the China National Clinical Research Center on Mental Disorders (Xiangya), Changsha, Hunan, China (Pan); the China National Technology Institute on Mental Disorders, Changsha, Hunan, China (Pan); the Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan, China (Pan); and the Institute of Mental Health of Second Xiangya Hospital, Central South University, Changsha, Hunan, China (Pan)
| | - Lena Palaniyappan
- From the Department of Psychiatry, Second Xiangya Hospital of Central South University, Changsha, Hunan, China (Pan); the Robarts Research Institute, University of Western Ontario, London, Ont., Canada (Pan, Khan, Palaniyappan); the Lawson Health Research Institute, London, Ont., Canada (Théberge, Palaniyappan); the Department of Medical Biophysics, University of Western Ontario, London, Ont., Canada (Jeon, Théberge, Khan, Palaniyappan); the Department of Psychiatry, University of Western Ontario, London, Ont., Canada (Palaniyappan, Théberge); the Department of Psychiatry, Dalhousie University, Halifax, NS, Canada (Dempster); the China National Clinical Research Center on Mental Disorders (Xiangya), Changsha, Hunan, China (Pan); the China National Technology Institute on Mental Disorders, Changsha, Hunan, China (Pan); the Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan, China (Pan); and the Institute of Mental Health of Second Xiangya Hospital, Central South University, Changsha, Hunan, China (Pan)
| |
Collapse
|
55
|
Cruz BF, de Campos-Carli SM, de Oliveira AM, de Brito CB, Garcia ZM, do Nascimento Arifa RD, de Souza DDG, Teixeira AL, Salgado JV. Investigating potential associations between neurocognition/social cognition and oxidative stress in schizophrenia. Psychiatry Res 2021; 298:113832. [PMID: 33652247 DOI: 10.1016/j.psychres.2021.113832] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 02/21/2021] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Deficits in neurocognition and social cognition play a critical role in the functional impairment of patients with schizophrenia. Increased oxidative stress has been evidenced in schizophrenia. Increased oxidative stress can affect neuronal function and lead to impairments in neurocognitive functions (especially working memory) and social cognition. OBJECTIVE To investigate deficits in neurocognition and social cognition and their potential association with oxidative stress biomarkers in schizophrenia. MATERIAL AND METHODS Eight-five clinically stable patients with schizophrenia and 75 controls were enrolled in this study. Neurocognition was evaluated through the Brief Assessment of Cognition in Schizophrenia (BACS). Social cognition was assessed through the Hinting Task - a test of theory of mind - and an emotion processing test, Facial Emotion Recognition Test (FERT-100). Oxidative stress was assessed by measuring serum levels of glutathione (GSH) and thiobarbituric acid reactive substances (TBARS). RESULTS Patients had decreased serum levels of GSH (Z=3.56; p<0.001) and increased TBARS (Z=5.51; P<0.001) when compared with controls. TBARS levels are higher in patients using first generation antipsychotics. Higher serum levels of TBARS in patients were associated with poor performance in working memory test (r=-0.39; p=0.002), even when controlling for age and negative symptoms (Standard Beta: -0.36; CI= -2.52 a -13.71). DISCUSSION The association between greater lipid peroxidation, as assessed by TBARS, and worse performance in working memory corroborates theoretical models of greater vulnerability of schizophrenia to oxidative stress.
Collapse
Affiliation(s)
- Breno Fiuza Cruz
- Mental Health Department, Federal University of Minas Gerais, Belo Horizonte, Brazil.
| | | | - Amanda Margarida de Oliveira
- Neuroscience Program, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Zélia Menezes Garcia
- Microbiology Department, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | | | - Antonio Lucio Teixeira
- Institute of Education and Research, Santa Casa BH, Belo Horizonte, Brazil; Neuropsychiatry Program, UTHealth Houston, TX, United States
| | - João Vinícius Salgado
- Neuroscience Program, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil; Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
56
|
Ansarey SH. Inflammation and JNK's Role in Niacin-GPR109A Diminished Flushed Effect in Microglial and Neuronal Cells With Relevance to Schizophrenia. Front Psychiatry 2021; 12:771144. [PMID: 34916973 PMCID: PMC8668869 DOI: 10.3389/fpsyt.2021.771144] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 11/02/2021] [Indexed: 12/28/2022] Open
Abstract
Schizophrenia is a neuropsychiatric illness with no single definitive aetiology, making its treatment difficult. Antipsychotics are not fully effective because they treat psychosis rather than the cognitive or negative symptoms. Antipsychotics fail to alleviate symptoms when patients enter the chronic stage of illness. Topical application of niacin showed diminished skin flush in the majority of patients with schizophrenia compared to the general population who showed flushing. The niacin skin flush test is useful for identifying patients with schizophrenia at their ultra-high-risk stage, and understanding this pathology may introduce an effective treatment. This review aims to understand the pathology behind the diminished skin flush response, while linking it back to neurons and microglia. First, it suggests that there are altered proteins in the GPR109A-COX-prostaglandin pathway, inflammatory imbalance, and kinase signalling pathway, c-Jun N-terminal kinase (JNK), which are associated with diminished flush. Second, genes from the GPR109A-COX-prostaglandin pathway were matched against the 128-loci genome wide association study (GWAS) for schizophrenia using GeneCards, suggesting that G-coupled receptor-109A (GPR109A) may have a genetic mutation, resulting in diminished flush. This review also suggests that there may be increased pro-inflammatory mediators in the GPR109A-COX-prostaglandin pathway, which contributes to the diminished flush pathology. Increased levels of pro-inflammatory markers may induce microglial-activated neuronal death. Lastly, this review explores the role of JNK on pro-inflammatory mediators, proteins in the GPR109A-COX-prostaglandin pathway, microglial activation, and neuronal death. Inhibiting JNK may reverse the changes observed in the diminished flush response, which might make it a good therapeutic target.
Collapse
Affiliation(s)
- Sabrina H Ansarey
- Department of Neuroscience and Psychology, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
57
|
Bessières B, Cruz E, Alberini CM. Metabolomic profiling reveals a differential role for hippocampal glutathione reductase in infantile memory formation. eLife 2021; 10:68590. [PMID: 34825649 PMCID: PMC8626085 DOI: 10.7554/elife.68590] [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: 03/19/2021] [Accepted: 11/09/2021] [Indexed: 01/12/2023] Open
Abstract
The metabolic mechanisms underlying the formation of early-life episodic memories remain poorly characterized. Here, we assessed the metabolomic profile of the rat hippocampus at different developmental ages both at baseline and following episodic learning. We report that the hippocampal metabolome significantly changes over developmental ages and that learning regulates differential arrays of metabolites according to age. The infant hippocampus had the largest number of significant changes following learning, with downregulation of 54 metabolites. Of those, a large proportion was associated with the glutathione-mediated cellular defenses against oxidative stress. Further biochemical, molecular, and behavioral assessments revealed that infantile learning evokes a rapid and persistent increase in the activity of neuronal glutathione reductase, the enzyme that regenerates reduced glutathione from its oxidized form. Inhibition of glutathione reductase selectively impaired long-term memory formation in infant but not in juvenile and adult rats, confirming its age-specific role. Thus, metabolomic profiling revealed that the hippocampal glutathione-mediated antioxidant pathway is differentially required for the formation of infantile memory.
Collapse
Affiliation(s)
| | - Emmanuel Cruz
- Center for Neural Science, New York UniversityNew YorkUnited States
| | | |
Collapse
|
58
|
Identification of a functional SNP rs7304782 at schizophrenia risk locus 12q24.31 and validation of its association with schiz ophrenia in Chinese populations. Psychiatry Res 2020; 294:113491. [PMID: 33070109 DOI: 10.1016/j.psychres.2020.113491] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/28/2020] [Indexed: 12/19/2022]
Abstract
Recent genome-wide association studies (GWAS) have identified multiple schizophrenia-associated risk loci. However, the potential functional (or causal) variant remains largely unknown for each of the identified risk locus. In this study, we utilized different functional annotation approaches (i.e., CADD, Eigen, GWAVA, RegulomeDB and LINSIGHT) to prioritize the most possible functional variant at schizophrenia risk locus 12q24.31, a risk locus that showed genome-wide significant association with schizophrenia. We found that four functional annotation methods prioritized rs7304782 as a potential functional variant at 12q24.31, suggesting the potential functional consequence of rs7304782. Consistent with the functional annotation, reporter gene assays showed that different allele of rs7304782 affected the luciferase activity significantly, further supporting that rs7304782 is a functional variant. We further performed genetic association study and validated that rs7304782 is also associated with schizophrenia in Chinese population (N=4,291 cases and 7,847 controls), with the same risk allele as in European population. Expression quantitative trait loci (eQTL) analysis indicated that rs7304782 was significantly associated with the expression of OGFOD2 in human brain tissues. Of note, differential expression analysis indicated that OGFOD2 was significantly down-regulated in schizophrenia cases compared with controls. Our study identified a potential functional variant (i.e., rs7304782) at schizophrenia risk locus 12q24.31 and suggested that this functional variant may confer schizophrenia risk through regulating OGFOD2 expression.
Collapse
|
59
|
Sydnor VJ, Roalf DR. A meta-analysis of ultra-high field glutamate, glutamine, GABA and glutathione 1HMRS in psychosis: Implications for studies of psychosis risk. Schizophr Res 2020; 226:61-69. [PMID: 32723493 PMCID: PMC7750272 DOI: 10.1016/j.schres.2020.06.028] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 06/25/2020] [Accepted: 06/26/2020] [Indexed: 12/20/2022]
Abstract
Ultra-high field proton magnetic resonance spectroscopy (1HMRS) offers a unique opportunity to measure the concentration of neurometabolites implicated in psychosis (PSY). The extant 7 T 1HMRS literature measuring glutamate-associated neurometabolites in the brain in PSY in vivo is small, but a comprehensive, quantitative summary of these data can offer insight and guidance to this emerging field. This meta-analysis examines proton spectroscopy (1HMRS) measures of glutamate (Glu), glutamine (Gln), glutamate+glutamine (Glx), gamma aminobutyric acid (GABA), and glutathione (GSH) across 255 individuals with PSY (121 first episode) and 293 healthy comparison participants (HC). While all five neurometabolites were lower in PSY as compared to HC, only Glu (Cohen's d = -0.18) and GSH (Cohen's d = -0.21) concentrations were significantly lower in PSY, whereas concentrations of Gln, Glx, and GABA did not significantly differ between groups. Notably, 1HMRS methodological choices and sample demographic characteristics did not impact study-specific effect sizes for PSY-related Glu or GSH differences. This review thus provides further evidence of neurometabolite dysfunction in first episode and chronic PSY, and thereby suggests that Glu and GSH abnormalities may additionally play a role in more incipient stages of the disorder: in clinical high risk stages. Additional 7 T neurochemical imaging studies in larger, longitudinal, and unmedicated samples and in youth at risk for developing psychosis are needed. Such studies will be critical for elucidating the neurodevelopmental and clinical time course of PSY-related neurometabolite alterations, and for assessing the potential for implicated metabolites to serve as druggable targets for decreasing PSY risk.
Collapse
Affiliation(s)
- Valerie J Sydnor
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, United States of America
| | - David R Roalf
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, United States of America; Lifespan Brain Institute at the Children's Hospital of Philadelphia & the University of Pennsylvania, Philadelphia, PA 19104, United States of America.
| |
Collapse
|
60
|
Naderi M, Kwong RWM. A comprehensive review of the neurobehavioral effects of bisphenol S and the mechanisms of action: New insights from in vitro and in vivo models. ENVIRONMENT INTERNATIONAL 2020; 145:106078. [PMID: 32911243 DOI: 10.1016/j.envint.2020.106078] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 08/12/2020] [Accepted: 08/17/2020] [Indexed: 06/11/2023]
Abstract
The normal brain development and function are delicately driven by an ever-changing milieu of steroid hormones arising from fetal, placental, and maternal origins. This reliance on the neuroendocrine system sets the stage for the exquisite sensitivity of the central nervous system to the adverse effects of endocrine-disrupting chemicals (EDCs). Bisphenol A (BPA) is one of the most common EDCs which has been a particular focus of environmental concern for decades due to its widespread nature and formidable threat to human and animal health. The heightened regulatory actions and the scientific and public concern over the adverse health effects of BPA have led to its replacement with a suite of structurally similar but less known alternative chemicals. Bisphenol S (BPS) is the main substitute for BPA that is increasingly being used in a wide array of consumer and industrial products. Although it was considered to be a safe BPA alternative, mounting evidence points to the deleterious effects of BPS on a wide range of neuroendocrine functions in animals. In addition to its reproductive toxicity, recent experimental efforts indicate that BPS has a considerable potential to induce neurotoxicity and behavioral dysfunction. This review analyzes the current state of knowledge regarding the neurobehavioral effects of BPS and discusses its potential mode of actions on several aspects of the neuroendocrine system. We summarize the role of certain hormones and their signaling pathways in the regulation of brain and behavior and discuss how BPS induces neurotoxicity through interactions with these pathways. Finally, we review potential links between BPS exposure and aberrant neurobehavioral functions in animals and identify key knowledge gaps and hypotheses for future research.
Collapse
Affiliation(s)
- Mohammad Naderi
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada.
| | - Raymond W M Kwong
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada.
| |
Collapse
|
61
|
Gomes FV, Zhu X, Grace AA. The pathophysiological impact of stress on the dopamine system is dependent on the state of the critical period of vulnerability. Mol Psychiatry 2020; 25:3278-3291. [PMID: 31488866 PMCID: PMC7056584 DOI: 10.1038/s41380-019-0514-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 04/02/2019] [Accepted: 07/18/2019] [Indexed: 12/18/2022]
Abstract
Unregulated stress during critical periods of development is proposed to drive deficits consistent with schizophrenia in adults. If accurate, reopening the critical period could make the adult susceptible to pathology. We evaluated the impact of early adolescent and adult stress exposure (combination of daily footshock for 10 days and 3 restraint sessions) on (1) midbrain dopamine (DA) neuron activity, (2) ventral hippocampal (vHipp) pyramidal neuron activity, and (3) the number of parvalbumin (PV) interneurons in the vHipp and their associated perineuronal nets (PNNs). Ventral tegmental area (VTA) DA neuron population activity and vHipp activity was increased 1-2 and 5-6 weeks post-adolescent stress, along with a decrease in the number of PV+, PNN+, PV + /PNN + cells in the vHipp, which are consistent with the MAM model of schizophrenia. In contrast, adult stress decreased VTA DA neuron population activity only at 1-2 weeks post stress, which is consistent with what has been observed in animal models of depression, without impacting vHipp activity and PV/PNN expression. Administration of valproate (VPA), which can re-instate the critical period of plasticity via histone deacetylase (HDAC) inhibition, caused adult stress to produce changes similar to those induced by adolescent stress, presumably by increasing stress vulnerability to early adolescent levels. Our findings indicate that timing of stress is a critical determinant of the pathology produced in the adult: adolescent stress led to circuit deficits that recapitulates schizophrenia, whereas adult stress induced a depression-like hypodopaminergic state. Reopening the critical period in the adult restores vulnerability to stress-induced pathology resembling schizophrenia.
Collapse
Affiliation(s)
- Felipe V. Gomes
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, PA, USA
| | - Xiyu Zhu
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, PA, USA
| | - Anthony A. Grace
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, PA, USA
| |
Collapse
|
62
|
Janickova L, Schwaller B. Parvalbumin-Deficiency Accelerates the Age-Dependent ROS Production in Pvalb Neurons in vivo: Link to Neurodevelopmental Disorders. Front Cell Neurosci 2020; 14:571216. [PMID: 33132847 PMCID: PMC7549402 DOI: 10.3389/fncel.2020.571216] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/12/2020] [Indexed: 12/26/2022] Open
Abstract
In neurodevelopmental disorders (NDDs) including autism spectrum disorder (ASD) and schizophrenia, impairment/malfunctioning of a subpopulation of interneurons expressing the calcium-binding protein parvalbumin (PV) -here termed Pvalb neurons- has gradually emerged as a possible cause. These neurons may represent a hub or point-of-convergence in the etiology of NDD. Increased oxidative stress associated with mitochondria impairment in Pvalb neurons is discussed as an essential step in schizophrenia etiology. Since PV downregulation is a common finding in ASD and schizophrenia individuals and PV-deficient (PV-/-) mice show a strong ASD-like behavior phenotype, we investigated the putative link between PV expression, alterations in mitochondria and oxidative stress. In a longitudinal study with 1, 3, and 6-months old PV-/- and wild type mice, oxidative stress was investigated in 9 Pvalb neuron subpopulations in the hippocampus, striatum, somatosensory cortex, medial prefrontal cortex, thalamic reticular nucleus (TRN) and cerebellum. In Pvalb neuron somata in the striatum and TRN, we additionally determined mitochondria volume and distribution at these three time points. In all Pvalb neuron subpopulations, we observed an age-dependent increase in oxidative stress and the increase strongly correlated with PV expression levels, but not with mitochondria density in these Pvalb neurons. Moreover, oxidative stress was elevated in Pvalb neurons of PV-/- mice and the magnitude of the effect was again correlated with PV expression levels in the corresponding wild type Pvalb neuron subpopulations. The PV-dependent effect was insignificant at 1 month and relative differences between WT and PV-/- Pvalb neurons were largest at 3 months. Besides the increase in mitochondria volume in PV's absence in TRN and striatal PV-/- Pvalb neurons fully present already at 1 month, we observed a redistribution of mitochondria from the perinuclear region toward the plasma membrane at all time points. We suggest that in absence of PV, slow Ca2+ buffering normally exerted by PV is compensated by a (mal)adaptive, mostly sub-plasmalemmal increase in mitochondria resulting in increased oxidative stress observed in 3- and 6-months old mice. Since PV-/- mice display core ASD-like symptoms already at 1 month, oxidative stress in Pvalb neurons is not a likely cause for their ASD-related behavior observed at this age.
Collapse
Affiliation(s)
| | - Beat Schwaller
- Department of Neurosciences amd Movement Science, Section of Medicine, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
63
|
Perkins DO, Jeffries CD, Do KQ. Potential Roles of Redox Dysregulation in the Development of Schizophrenia. Biol Psychiatry 2020; 88:326-336. [PMID: 32560962 PMCID: PMC7395886 DOI: 10.1016/j.biopsych.2020.03.016] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 03/03/2020] [Accepted: 03/22/2020] [Indexed: 12/20/2022]
Abstract
Converging evidence implicates redox dysregulation as a pathological mechanism driving the emergence of psychosis. Increased oxidative damage and decreased capacity of intracellular redox modulatory systems are consistent findings in persons with schizophrenia as well as in persons at clinical high risk who subsequently developed frank psychosis. Levels of glutathione, a key regulator of cellular redox status, are reduced in the medial prefrontal cortex, striatum, and thalamus in schizophrenia. In humans with schizophrenia and in rodent models recapitulating various features of schizophrenia, redox dysregulation is linked to reductions of parvalbumin containing gamma-aminobutyric acid (GABA) interneurons and volumes of their perineuronal nets, white matter abnormalities, and microglia activation. Importantly, the activity of transcription factors, kinases, and phosphatases regulating diverse aspects of neurodevelopment and synaptic plasticity varies according to cellular redox state. Molecules regulating interneuron function under redox control include NMDA receptor subunits GluN1 and GluN2A as well as KEAP1 (regulator of transcription factor NRF2). In a rodent schizophrenia model characterized by impaired glutathione synthesis, the Gclm knockout mouse, oxidative stress activated MMP9 (matrix metalloprotease 9) via its redox-responsive regulatory sites, causing a cascade of molecular events leading to microglia activation, perineural net degradation, and impaired NMDA receptor function. Molecular pathways under redox control are implicated in the etiopathology of schizophrenia and are attractive drug targets for individualized drug therapy trials in the contexts of prevention and treatment of psychosis.
Collapse
Affiliation(s)
- Diana O. Perkins
- corresponding author: CB 7160, Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, Office: 919-962-1401, Cell: 919-360-1602,
| | - Clark D. Jeffries
- Renaissance Computing Institute, University of North Carolina, Chapel Hill NC
| | - Kim Q. Do
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital-CHUV, Prilly-Lausanne, Switzerland
| |
Collapse
|
64
|
Phensy A, Lindquist KL, Lindquist KA, Bairuty D, Gauba E, Guo L, Tian J, Du H, Kroener S. Deletion of the Mitochondrial Matrix Protein CyclophilinD Prevents Parvalbumin Interneuron Dysfunctionand Cognitive Deficits in a Mouse Model of NMDA Hypofunction. J Neurosci 2020; 40:6121-6132. [PMID: 32605939 PMCID: PMC7406283 DOI: 10.1523/jneurosci.0880-20.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/28/2020] [Accepted: 06/22/2020] [Indexed: 12/23/2022] Open
Abstract
Redox dysregulation and oxidative stress are final common pathways in the pathophysiology of a variety of psychiatric disorders, including schizophrenia. Oxidative stress causes dysfunction of GABAergic parvalbumin (PV)-positive interneurons (PVI), which are crucial for the coordination of neuronal synchrony during sensory and cognitive processing. Mitochondria are the main source of reactive oxygen species (ROS) in neurons and they control synaptic activity through their roles in energy production and intracellular calcium homeostasis. We have previously shown that in male mice transient blockade of NMDA receptors (NMDARs) during development [subcutaneous injections of 30 mg/kg ketamine (KET) on postnatal days 7, 9, and 11] results in long-lasting alterations in synaptic transmission and reduced PV expression in the adult prefrontal cortex (PFC), contributing to a behavioral phenotype that mimics multiple symptoms associated with schizophrenia. These changes correlate with oxidative stress and impaired mitochondrial function in both PVI and pyramidal cells. Here, we show that genetic deletion (Ppif-/-) of the mitochondrial matrix protein cyclophilin D (CypD) prevents perinatal KET-induced increases in ROS and the resulting deficits in PVI function, and changes in excitatory and inhibitory synaptic transmission in the PFC. Deletion of CypD also prevented KET-induced behavioral deficits in cognitive flexibility, social interaction, and novel object recognition (NOR). Taken together, these data highlight how mitochondrial activity may play an integral role in modulating PVI-mediated cognitive processes.SIGNIFICANCE STATEMENT Mitochondria are important modulators of oxidative stress and cell function, yet how mitochondrial dysfunction affects cell activity and synaptic transmission in psychiatric illnesses is not well understood. NMDA receptor (NMDAR) blockade with ketamine (KET) during development causes oxidative stress, dysfunction of parvalbumin (PV)-positive interneurons (PVI), and long-lasting physiological and behavioral changes. Here we show that mice deficient for the mitochondrial matrix protein cyclophilin D (CypD) show robust protection from PVI dysfunction following perinatal NMDAR blockade. Mitochondria serve as an essential node for a number of stress-induced signaling pathways and our experiments suggest that failure of mitochondrial redox regulation can contribute to PVI dysfunction.
Collapse
Affiliation(s)
- Aarron Phensy
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, Texas 75080
| | - Kathy L Lindquist
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, Texas 75080
| | - Karen A Lindquist
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, Texas 75080
| | - Dania Bairuty
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, Texas 75080
| | - Esha Gauba
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, Texas 75080
| | - Lan Guo
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, Texas 75080
| | - Jing Tian
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, Texas 75080
| | - Heng Du
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, Texas 75080
| | - Sven Kroener
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, Texas 75080
| |
Collapse
|
65
|
Javitt DC, Siegel SJ, Spencer KM, Mathalon DH, Hong LE, Martinez A, Ehlers CL, Abbas AI, Teichert T, Lakatos P, Womelsdorf T. A roadmap for development of neuro-oscillations as translational biomarkers for treatment development in neuropsychopharmacology. Neuropsychopharmacology 2020; 45:1411-1422. [PMID: 32375159 PMCID: PMC7360555 DOI: 10.1038/s41386-020-0697-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 03/16/2020] [Accepted: 04/27/2020] [Indexed: 02/08/2023]
Abstract
New treatment development for psychiatric disorders depends critically upon the development of physiological measures that can accurately translate between preclinical animal models and clinical human studies. Such measures can be used both as stratification biomarkers to define pathophysiologically homogeneous patient populations and as target engagement biomarkers to verify similarity of effects across preclinical and clinical intervention. Traditional "time-domain" event-related potentials (ERP) have been used translationally to date but are limited by the significant differences in timing and distribution across rodent, monkey and human studies. By contrast, neuro-oscillatory responses, analyzed within the "time-frequency" domain, are relatively preserved across species permitting more precise translational comparisons. Moreover, neuro-oscillatory responses are increasingly being mapped to local circuit mechanisms and may be useful for investigating effects of both pharmacological and neuromodulatory interventions on excitatory/inhibitory balance. The present paper provides a roadmap for development of neuro-oscillatory responses as translational biomarkers in neuropsychiatric treatment development.
Collapse
Affiliation(s)
- Daniel C Javitt
- Department of Psychiatry, Columbia University Medical Center, New York, NY, 10032, USA.
- Schizophrenia Research Division, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, 10954, USA.
| | - Steven J Siegel
- Department of Psychiatry and Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Kevin M Spencer
- Research Service, VA Boston Healthcare System, and Dept. of Psychiatry, Harvard Medical School, Boston, MA, 02130, USA
| | - Daniel H Mathalon
- VA San Francisco Healthcare System, University of California, San Francisco, San Francisco, CA, 94121, USA
| | - L Elliot Hong
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Antigona Martinez
- Department of Psychiatry, Columbia University Medical Center, New York, NY, 10032, USA
- Schizophrenia Research Division, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, 10954, USA
| | - Cindy L Ehlers
- Department of Neuroscience, The Scripps Research Institute, 10550 N Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Atheir I Abbas
- VA Portland Health Care System, Portland, OR, 97239, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Tobias Teichert
- Departments of Psychiatry and Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Peter Lakatos
- Schizophrenia Research Division, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, 10954, USA
| | - Thilo Womelsdorf
- Department of Psychology, Vanderbilt University, Nashville, TN, 37203, USA
| |
Collapse
|
66
|
Alterations in the Antioxidant Enzyme Activities in the Neurodevelopmental Rat Model of Schizophrenia Induced by Glutathione Deficiency during Early Postnatal Life. Antioxidants (Basel) 2020; 9:antiox9060538. [PMID: 32575563 PMCID: PMC7346228 DOI: 10.3390/antiox9060538] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/10/2020] [Accepted: 06/17/2020] [Indexed: 01/24/2023] Open
Abstract
The aim of the present study was to assess the effects of l-buthionine-(S,R)-sulfoximine (BSO), a glutathione (GSH) synthesis inhibitor, and GBR 12909, a dopamine reuptake inhibitor, administered alone or in combination to Sprague-Dawley rats during early postnatal development (p5-p16), on the levels of reactive oxygen species (ROS), lipid peroxidation (LP) and the activities of antioxidant enzymes: superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx) and glutathione disulfide reductase (GR) in peripheral tissues (liver, kidney) and selected brain structures (prefrontal cortex, PFC; hippocampus, HIP; and striatum, STR) of 16-day-old rats. The studied parameters were analyzed with reference to the content of GSH and sulfur amino acids, methionine (Met) and cysteine (Cys) described in our previous study. This analysis showed that treatment with a BSO + GBR 12909 combination caused significant decreases in the lipid peroxidation levels in the PFC and HIP, in spite of there being no changes in ROS. The reduction of lipid peroxidation indicates a weakening of the oxidative power of the cells, and a shift in balance in favor of reducing processes. Such changes in cellular redox signaling in the PFC and HIP during early postnatal development may result in functional changes in adulthood.
Collapse
|
67
|
MK-801 Exposure during Adolescence Elicits Enduring Disruption of Prefrontal E-I Balance and Its Control of Fear Extinction Behavior. J Neurosci 2020; 40:4881-4887. [PMID: 32430298 DOI: 10.1523/jneurosci.0581-20.2020] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/05/2020] [Accepted: 05/11/2020] [Indexed: 01/04/2023] Open
Abstract
Understanding how disruption of prefrontal cortex (PFC) maturation during adolescence is crucial to reveal which neural processes could contribute to the onset of psychiatric disorders that display frontal cortical deficits. Of particular interest is the gain of GABAergic function in the PFC during adolescence and its susceptibility to the impact of transient blockade of NMDA receptor function. Here we assessed whether exposure to MK-801 during adolescence in male rats triggers a state of excitatory-inhibitory imbalance in the PFC that limits its functional capacity to regulate behavior in adulthood. Recordings from PFC brain slices revealed that MK-801 exposure during adolescence preferentially reduces the presynaptic functionality of GABAergic activity over that of excitatory synapses. As a result, an imbalance of excitatory-inhibitory synaptic activity emerges in the PFC that correlates linearly with the GABAergic deficit. Notably, the data also suggest that the diminished prefrontal GABAergic function could arise from a deficit in the recruitment of fast-spiking interneurons by excitatory inputs during adolescence. At the behavioral level, MK-801 exposure during adolescence did not disrupt the acquisition of trace fear conditioning, but markedly increased the level of freezing response during extinction testing. Infusion of the GABAA receptor-positive allosteric modulator Indiplon into the PFC before extinction testing reduced the level of freezing response in MK-801-treated rats to control levels. Collectively, the results indicate NMDA receptor signaling during adolescence enables the gain of prefrontal GABAergic function, which is required for maintaining proper excitatory-inhibitory balance in the PFC and its control of behavioral responses.SIGNIFICANCE STATEMENT A developmental disruption of prefrontal cortex maturation has been implicated in the pathophysiology of cognitive deficits in psychiatric disorders. Of particular interest is the susceptibility of the local GABAergic circuit to the impact of transient disruption of NMDA receptors. Here we found that NMDA receptor signaling is critical to enable the gain of prefrontal GABAergic transmission during adolescence for maintaining proper levels of excitatory-inhibitory balance in the PFC and its control of behavior.
Collapse
|
68
|
Bitanihirwe BKY, Woo TUW. A conceptualized model linking matrix metalloproteinase-9 to schizophrenia pathogenesis. Schizophr Res 2020; 218:28-35. [PMID: 32001079 DOI: 10.1016/j.schres.2019.12.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 12/18/2019] [Accepted: 12/18/2019] [Indexed: 12/11/2022]
Abstract
Matrix metalloproteinase 9 (MMP-9) is an extracellularly operating zinc-dependent endopeptidase that is commonly expressed in the brain, other tissues. It is synthesized in a latent zymogen form known as pro-MMP-9 that is subsequently converted to the active MMP-9 enzyme following cleavage of the pro-domain. Within the central nervous system, MMP-9 is localized and released from neurons, astrocytes and microglia where its expression levels are modulated by cytokines and growth factors during both normal and pathological conditions as well as by reactive oxygen species generated during oxidative stress. MMP-9 is involved in a number of key neurodevelopmental processes that are thought to be affected in schizophrenia, including maturation of the inhibitory neurons that contain the calcium-binding protein parvalbumin, developmental formation of the specialized extracellular matrix structure perineuronal net, synaptic pruning, and myelination. In this context, the present article provides a narrative synthesis of the existing evidence linking MMP-9 dysregulation to schizophrenia pathogenesis. We start by providing an overview of MMP-9 involvement in brain development and physiology. We then discuss the potential mechanisms through which MMP-9 dysregulation may affect neural circuitry maturation as well as how these anomalies may contribute to the disease process of schizophrenia. We conclude by articulating a comprehensive, cogent, and experimentally testable hypothesis linking MMP-9 to the developmental pathophysiologic cascade that triggers the onset and sustains the chronicity of the illness.
Collapse
Affiliation(s)
| | - Tsung-Ung W Woo
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA; Program in Cellular Neuropathology, McLean Hospital, Belmont, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
69
|
Hong J, Bang M. Anti-inflammatory Strategies for Schizophrenia: A Review of Evidence for Therapeutic Applications and Drug Repurposing. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2020; 18:10-24. [PMID: 31958901 PMCID: PMC7006977 DOI: 10.9758/cpn.2020.18.1.10] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 09/12/2019] [Accepted: 11/04/2019] [Indexed: 02/06/2023]
Abstract
Schizophrenia is a debilitating psychiatric disorder with a substantial socioeconomic and humanistic burden. Currently available treatment strategies mostly rely on antipsychotic drugs, which block dopaminergic effects in the mesolimbic pathway of the brain. Although antipsychotic drugs help relieve psychotic symptoms, a definitive cure for schizophrenia has yet to be achieved. Recent advances in neuroinflammation research suggest that proinflammatory processes in the brain could cause alterations in neurobehavioral development and increase vulnerability to schizophrenia. With a growing need for novel strategies in the treatment of schizophrenia, it would be meaningful to review the current evidence supporting the therapeutic potential of anti-inflammatory strategies. This review details the key findings of clinical trials that investigate the efficacy of anti-inflammatory agents as adjuvants to antipsychotic treatment. We further discuss the possibilities of repurposing anti-inflammatory agents and developing novel strategies for the treatment of schizophrenia.
Collapse
Affiliation(s)
- Jonghee Hong
- CHA University School of Medicine, Seongnam, Korea
| | - Minji Bang
- Department of Psychiatry, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Korea
| |
Collapse
|
70
|
Dempster K, Jeon P, MacKinley M, Williamson P, Théberge J, Palaniyappan L. Early treatment response in first episode psychosis: a 7-T magnetic resonance spectroscopic study of glutathione and glutamate. Mol Psychiatry 2020; 25:1640-1650. [PMID: 32205866 PMCID: PMC7387300 DOI: 10.1038/s41380-020-0704-x] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 02/18/2020] [Accepted: 02/24/2020] [Indexed: 01/07/2023]
Abstract
Early response to antipsychotic medications is one of the most important determinants of later symptomatic and functional outcomes in psychosis. Glutathione and glutamate have emerged as promising therapeutic targets for patients demonstrating inadequate response to dopamine-blocking antipsychotics. Nevertheless, the role of these neurochemicals in the mechanism of early antipsychotic response remains poorly understood. Using a longitudinal design and ultrahigh field 7-T magnetic resonance spectroscopy (MRS) protocol in 53 subjects, we report the association between dorsal anterior cingulate cortex glutamate and glutathione, with time to treatment response in drug naive (34.6% of the sample) or minimally medicated first episode patients with schizophreniform disorder, schizophrenia, and schizoaffective disorder. Time to response was defined as the number of weeks required to reach a 50% reduction in the PANSS-8 scores. Higher glutathione was associated with shorter time to response (F = 4.86, P = 0.017), while higher glutamate was associated with more severe functional impairment (F = 5.33, P = 0.008). There were no significant differences between patients and controls on measures of glutamate or glutathione. For the first time, we have demonstrated an association between higher glutathione and favorable prognosis in FEP. We propose that interventions that increase brain glutathione levels may improve outcomes of early intervention in psychosis.
Collapse
Affiliation(s)
- Kara Dempster
- 0000 0004 1936 8200grid.55602.34Department of Psychiatry, Dalhousie University, Halifax, NS Canada
| | - Peter Jeon
- 0000 0004 1936 8884grid.39381.30Department of Medical Biophysics, University of Western Ontario, London, ON Canada
| | - Michael MacKinley
- 0000 0004 1936 8884grid.39381.30Robarts Research Institute, London, ON Canada
| | - Peter Williamson
- 0000 0004 1936 8884grid.39381.30Robarts Research Institute, London, ON Canada ,0000 0004 1936 8884grid.39381.30Department of Psychiatry, University of Western Ontario, London, ON Canada ,0000 0001 0556 2414grid.415847.bLawson Health Research Institute, London, ON Canada
| | - Jean Théberge
- 0000 0004 1936 8884grid.39381.30Department of Medical Biophysics, University of Western Ontario, London, ON Canada ,0000 0004 1936 8884grid.39381.30Robarts Research Institute, London, ON Canada ,0000 0001 0556 2414grid.415847.bLawson Health Research Institute, London, ON Canada ,0000 0000 9674 4717grid.416448.bDepartment of Diagnostic Imaging, St. Joseph’s Health Care London, London, ON Canada
| | - Lena Palaniyappan
- Department of Medical Biophysics, University of Western Ontario, London, ON, Canada. .,Robarts Research Institute, London, ON, Canada. .,Department of Psychiatry, University of Western Ontario, London, ON, Canada. .,Lawson Health Research Institute, London, ON, Canada.
| |
Collapse
|
71
|
Bittle J, Menezes EC, McCormick ML, Spitz DR, Dailey M, Stevens HE. The Role of Redox Dysregulation in the Effects of Prenatal Stress on Embryonic Interneuron Migration. Cereb Cortex 2019; 29:5116-5130. [PMID: 30877797 PMCID: PMC7199998 DOI: 10.1093/cercor/bhz052] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 02/18/2019] [Accepted: 02/22/2019] [Indexed: 01/09/2023] Open
Abstract
Maternal stress during pregnancy is associated with increased risk of psychiatric disorders in offspring, but embryonic brain mechanisms disrupted by prenatal stress are not fully understood. Our lab has shown that prenatal stress delays inhibitory neural progenitor migration. Here, we investigated redox dysregulation as a mechanism for embryonic cortical interneuron migration delay, utilizing direct manipulation of pro- and antioxidants and a mouse model of maternal repetitive restraint stress starting on embryonic day 12. Time-lapse, live-imaging of migrating GAD67GFP+ interneurons showed that normal tangential migration of inhibitory progenitor cells was disrupted by the pro-oxidant, hydrogen peroxide. Interneuron migration was also delayed by in utero intracerebroventricular rotenone. Prenatal stress altered glutathione levels and induced changes in activity of antioxidant enzymes and expression of redox-related genes in the embryonic forebrain. Assessment of dihydroethidium (DHE) fluorescence after prenatal stress in ganglionic eminence (GE), the source of migrating interneurons, showed increased levels of DHE oxidation. Maternal antioxidants (N-acetylcysteine and astaxanthin) normalized DHE oxidation levels in GE and ameliorated the migration delay caused by prenatal stress. Through convergent redox manipula-tions, delayed interneuron migration after prenatal stress was found to critically involve redox dysregulation. Redox biology during prenatal periods may be a target for protecting brain development.
Collapse
Affiliation(s)
- Jada Bittle
- Department of Psychiatry, University of Iowa Carver College of Medicine, 1310 PBDB, 169 Newton Rd, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Neuroscience, University of Iowa, 356 Medical Research Center, Iowa City, IA, USA
| | - Edenia C Menezes
- Department of Psychiatry, University of Iowa Carver College of Medicine, 1310 PBDB, 169 Newton Rd, Iowa City, IA, USA
| | - Michael L McCormick
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Carver College of Medicine, B180 Medical Laboratories, Iowa City, IA, USA
| | - Douglas R Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Carver College of Medicine, B180 Medical Laboratories, Iowa City, IA, USA
| | - Michael Dailey
- Interdisciplinary Graduate Program in Neuroscience, University of Iowa, 356 Medical Research Center, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, 2312 PBDB, 169 Newton Rd, Iowa City, IA, USA
| | - Hanna E Stevens
- Department of Psychiatry, University of Iowa Carver College of Medicine, 1310 PBDB, 169 Newton Rd, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Neuroscience, University of Iowa, 356 Medical Research Center, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, 2312 PBDB, 169 Newton Rd, Iowa City, IA, USA
| |
Collapse
|
72
|
Reiterer M, Schmidt-Kastner R, Milton SL. Methionine sulfoxide reductase (Msr) dysfunction in human brain disease. Free Radic Res 2019; 53:1144-1154. [PMID: 31775527 DOI: 10.1080/10715762.2019.1662899] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Extensive research has shown that oxidative stress is strongly associated with aging, senescence and several diseases, including neurodegenerative and psychiatric disorders. Oxidative stress is caused by the overproduction of reactive oxygen species (ROS) that can be counteracted by both enzymatic and nonenzymatic antioxidants. One of these antioxidant mechanisms is the widely studied methionine sulfoxide reductase system (Msr). Methionine is one of the most easily oxidized amino acids and Msr can reverse this oxidation and restore protein function, with MsrA and MsrB reducing different stereoisomers. This article focuses on experimental and genetic research performed on Msr and its link to brain diseases. Studies on several model systems as well as genome-wide association studies are compiled to highlight the role of MSRA in schizophrenia, Alzheimer's disease, and Parkinson's disease. Genetic variation of MSRA may also contribute to the risk of psychosis, personality traits, and metabolic factors.
Collapse
Affiliation(s)
- Melissa Reiterer
- Charles E. Schmidt College of Science, Florida Atlantic University, Boca Raton, FL, USA
| | | | - Sarah L Milton
- Charles E. Schmidt College of Science, Florida Atlantic University, Boca Raton, FL, USA
| |
Collapse
|
73
|
Górny M, Wnuk A, Kamińska A, Kamińska K, Chwatko G, Bilska-Wilkosz A, Iciek M, Kajta M, Rogóż Z, Lorenc-Koci E. Glutathione Deficiency and Alterations in the Sulfur Amino Acid Homeostasis during Early Postnatal Development as Potential Triggering Factors for Schizophrenia-Like Behavior in Adult Rats. Molecules 2019; 24:molecules24234253. [PMID: 31766654 PMCID: PMC6930621 DOI: 10.3390/molecules24234253] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 11/15/2019] [Accepted: 11/18/2019] [Indexed: 01/09/2023] Open
Abstract
Impaired glutathione (GSH) synthesis and dopaminergic transmission are important factors in the pathophysiology of schizophrenia. Our research aimed to assess the effects of l-buthionine-(S,R)-sulfoximine (BSO), a GSH synthesis inhibitor, and GBR 12909, a dopamine reuptake inhibitor, administered alone or in combination, to Sprague–Dawley rats during early postnatal development (p5–p16), on the levels of GSH, sulfur amino acids, global DNA methylation, and schizophrenia-like behavior. GSH, methionine (Met), homocysteine (Hcy), and cysteine (Cys) contents were determined in the liver, kidney, and in the prefrontal cortex (PFC) and hippocampus (HIP) of 16-day-old rats. DNA methylation in the PFC and HIP and schizophrenia-like behavior were assessed in adulthood (p90–p93). BSO caused the tissue-dependent decreases in GSH content and alterations in Met, Hcy, and Cys levels in the peripheral tissues and in the PFC and HIP. The changes in these parameters were accompanied by alterations in the global DNA methylation in the studied brain structures. Parallel to changes in the global DNA methylation, deficits in the social behaviors and cognitive functions were observed in adulthood. Only BSO + GBR 12909-treated rats exhibited behavioral alterations resembling positive symptoms in schizophrenia patients. Our results suggest the usefulness of this neurodevelopmental model for research on the pathomechanism of schizophrenia.
Collapse
Affiliation(s)
- Magdalena Górny
- The Chair of Medical Biochemistry, Jagiellonian University Medical College, 7 Kopernika Street, 31–034 Kraków, Poland; (M.G.); (A.B.-W.); (M.I.)
| | - Agnieszka Wnuk
- Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, 31–343 Kraków, Poland; (A.W.); (K.K.); (M.K.); (Z.R.)
| | - Adrianna Kamińska
- Department of Environmental Chemistry, University of Łódź, 163 Pomorska Street, 90-236 Łódź, Poland; (A.K.); (G.C.)
| | - Kinga Kamińska
- Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, 31–343 Kraków, Poland; (A.W.); (K.K.); (M.K.); (Z.R.)
| | - Grażyna Chwatko
- Department of Environmental Chemistry, University of Łódź, 163 Pomorska Street, 90-236 Łódź, Poland; (A.K.); (G.C.)
| | - Anna Bilska-Wilkosz
- The Chair of Medical Biochemistry, Jagiellonian University Medical College, 7 Kopernika Street, 31–034 Kraków, Poland; (M.G.); (A.B.-W.); (M.I.)
| | - Małgorzata Iciek
- The Chair of Medical Biochemistry, Jagiellonian University Medical College, 7 Kopernika Street, 31–034 Kraków, Poland; (M.G.); (A.B.-W.); (M.I.)
| | - Małgorzata Kajta
- Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, 31–343 Kraków, Poland; (A.W.); (K.K.); (M.K.); (Z.R.)
| | - Zofia Rogóż
- Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, 31–343 Kraków, Poland; (A.W.); (K.K.); (M.K.); (Z.R.)
| | - Elżbieta Lorenc-Koci
- Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, 31–343 Kraków, Poland; (A.W.); (K.K.); (M.K.); (Z.R.)
- Correspondence: ; Tel.: +48-126-623-272
| |
Collapse
|
74
|
Cabungcal JH, Steullet P, Kraftsik R, Cuenod M, Do KQ. A developmental redox dysregulation leads to spatio-temporal deficit of parvalbumin neuron circuitry in a schizophrenia mouse model. Schizophr Res 2019; 213:96-106. [PMID: 30857872 DOI: 10.1016/j.schres.2019.02.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 02/19/2019] [Accepted: 02/22/2019] [Indexed: 11/26/2022]
Abstract
The fast-spiking parvalbumin (PV) interneurons play a critical role in neural circuit activity and dysfunction of these cells has been implicated in the cognitive deficits typically observed in schizophrenia patients. Due to the high metabolic demands of PV neurons, they are particularly susceptible to oxidative stress. Given the extant literature exploring the pathological effects of oxidative stress on PV cells in cortical regions linked to schizophrenia, we decided to investigate whether PV neurons in other select brain regions, including sub-cortical structures, may be differentially affected by redox dysregulation induced oxidative stress during neurodevelopment in mice with a genetically compromised glutathione synthesis (Gclm KO mice). Our analyses revealed a spatio-temporal sequence of PV cell deficit in Gclm KO mice, beginning with the thalamic reticular nucleus at postnatal day (P) 20 followed by a PV neuronal deficit in the amygdala at P40, then in the lateral globus pallidus and the ventral hippocampus Cornu Ammonis 3 region at P90 and finally the anterior cingulate cortex at P180. We suggest that PV neurons in different brain regions are developmentally susceptible to oxidative stress and that anomalies in the neurodevelopmental calendar of metabolic regulation can interfere with neural circuit maturation and functional connectivity contributing to the emergence of developmental psychopathology.
Collapse
Affiliation(s)
- Jan-Harry Cabungcal
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital-CHUV, Prilly-Lausanne, Switzerland
| | - Pascal Steullet
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital-CHUV, Prilly-Lausanne, Switzerland
| | - Rudolf Kraftsik
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Michel Cuenod
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital-CHUV, Prilly-Lausanne, Switzerland
| | - Kim Q Do
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital-CHUV, Prilly-Lausanne, Switzerland.
| |
Collapse
|
75
|
Gomes FV, Zhu X, Grace AA. Stress during critical periods of development and risk for schizophrenia. Schizophr Res 2019; 213:107-113. [PMID: 30711313 PMCID: PMC6667322 DOI: 10.1016/j.schres.2019.01.030] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 01/15/2019] [Accepted: 01/18/2019] [Indexed: 12/20/2022]
Abstract
Schizophrenia is a neurodevelopmental disorder with genetic predisposition, and stress has long been linked to its etiology. While stress affects all stages of the illness, increasing evidence suggests that stress during critical periods of development may be particularly detrimental, increasing individual's vulnerability to psychosis. To thoroughly understand the potential causative role of stress, our group has been focusing on the prenatal methylazoxymethanol acetate (MAM) rodent model, and discovered that MAM offspring display abnormal stress reactivity and heightened anxiety prepubertally, prior to the manifestation of a hyperdopaminergic state. Furthermore, pharmacologically treating anxiety during prepuberty prevented the emergence of the dopamine dysfunction in adulthood. Interestingly, sufficiently strong stressors applied to normal rats selectively during early development can recapitulate multiple schizophrenia-related phenotypes of MAM rats, whereas the same stress paradigm during adulthood only produced short-term depression-related deficits. Altogether, the evidence is thus converging: developmental disruption (genetic or environmental) might render animals more susceptible to the deleterious effects of stress during critical time windows, during which unregulated stress can lead to the emergence of psychosis later in life. As an important region regulating the midbrain dopamine system, the ventral hippocampus is particularly vulnerable to stress, and the distinct maturational profile of its fast-spiking parvalbumin interneurons may largely underlie such vulnerability. In this review, by discussing emerging evidence spanning clinical and basic science studies, we propose developmental stress vulnerability as a novel link between early predispositions and environmental triggering events in the pathophysiology of schizophrenia. This promising line of research can potentially provide not only insights into the etiology, but also a "roadmap" for disease prevention.
Collapse
Affiliation(s)
| | | | - Anthony A. Grace
- Corresponding author: Dr. Anthony A. Grace - Department of Neuroscience, A210 Langley Hall, University of Pittsburgh, Pittsburgh, PA, 15260, USA. Phone: +1 412 624 4609.
| |
Collapse
|
76
|
Nakazawa K, Sapkota K. The origin of NMDA receptor hypofunction in schizophrenia. Pharmacol Ther 2019; 205:107426. [PMID: 31629007 DOI: 10.1016/j.pharmthera.2019.107426] [Citation(s) in RCA: 157] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 10/10/2019] [Indexed: 12/12/2022]
Abstract
N-methyl-d-aspartate (NMDA) receptor (NMDAR) hypofunction plays a key role in pathophysiology of schizophrenia. Since NMDAR hypofunction has also been reported in autism, Alzheimer's disease and cognitive dementia, it is crucial to identify the location, timing, and mechanism of NMDAR hypofunction for schizophrenia for better understanding of disease etiology and for novel therapeutic intervention. In this review, we first discuss the shared underlying mechanisms of NMDAR hypofunction in NMDAR antagonist models and the anti-NMDAR autoantibody model of schizophrenia and suggest that NMDAR hypofunction could occur in GABAergic neurons in both models. Preclinical models using transgenic mice have shown that NMDAR hypofunction in cortical GABAergic neurons, in particular parvalbumin-positive fast-spiking interneurons, in the early postnatal period confers schizophrenia-related phenotypes. Recent studies suggest that NMDAR hypofunction can also occur in PV-positive GABAergic neurons with alterations of NMDAR-associated proteins, such as neuregulin/ErbB4, α7nAChR, and serine racemase. Furthermore, several environmental factors, such as oxidative stress, kynurenic acid and hypoxia, may also potentially elicit NMDAR hypofunction in GABAergic neurons in early postnatal period. Altogether, the studies discussed here support a central role for GABAergic abnormalities in the context of NMDAR hypofunction. We conclude by suggesting potential therapeutic strategies to improve the function of fast-spiking neurons.
Collapse
|
77
|
Tsugawa S, Noda Y, Tarumi R, Mimura Y, Yoshida K, Iwata Y, Elsalhy M, Kuromiya M, Kurose S, Masuda F, Morita S, Ogyu K, Plitman E, Wada M, Miyazaki T, Graff-Guerrero A, Mimura M, Nakajima S. Glutathione levels and activities of glutathione metabolism enzymes in patients with schizophrenia: A systematic review and meta-analysis. J Psychopharmacol 2019; 33:1199-1214. [PMID: 31039654 DOI: 10.1177/0269881119845820] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Glutathione is among the important antioxidants to prevent oxidative stress. However, the relationships between abnormality in the glutathione system and pathophysiology of schizophrenia remain uncertain due to inconsistent findings on glutathione levels and/or glutathione-related enzyme activities in patients with schizophrenia. METHODS A systematic literature search was conducted using Embase, Medline, PsycINFO, and PubMed. Original studies, in which three metabolite levels (glutathione, glutathione disulfide, and total glutathione (glutathione+glutathione disulfide)) and five enzyme activities (glutathione peroxidase, glutathione reductase, glutamate-cysteine ligase, glutathione synthetase, and glutathione S-transferase) were measured with any techniques in both patients with schizophrenia and healthy controls, were included. Standardized mean differences were calculated to determine the group differences in the glutathione levels with a random-effects model. RESULTS We identified 41, 9, 15, 38, and seven studies which examined glutathione, glutathione disulfide, total glutathione, glutathione peroxidase, and glutathione reductase, respectively. Patients with schizophrenia had lower levels of both glutathione and total glutathione and decreased activity of glutathione peroxidase compared to controls. Glutathione levels were lower in unmedicated patients with schizophrenia than those in controls while glutathione levels did not differ between patients with first-episode psychosis and controls. CONCLUSIONS Our findings suggested that there may be glutathione deficits and abnormalities in the glutathione redox cycle in patients with schizophrenia. However, given the small number of studies examined the entire glutathione system, further studies are needed to elucidate a better understanding of disrupted glutathione function in schizophrenia, which may pave the way for the development of novel therapeutic strategies in this disorder.
Collapse
Affiliation(s)
- Sakiko Tsugawa
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Yoshihiro Noda
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Ryosuke Tarumi
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Yu Mimura
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Kazunari Yoshida
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan.,Pharmacogenetic Research Clinic, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Yusuke Iwata
- Multimodal Imaging Group, University of Toronto, Toronto, ON, Canada
| | - Muhammad Elsalhy
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Minori Kuromiya
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Shin Kurose
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Fumi Masuda
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Shinji Morita
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Kamiyu Ogyu
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Eric Plitman
- Multimodal Imaging Group, University of Toronto, Toronto, ON, Canada
| | - Masataka Wada
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Takahiro Miyazaki
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | | | - Masaru Mimura
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Shinichiro Nakajima
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan.,Multimodal Imaging Group, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
78
|
Kim E, Keskey Z, Kang M, Kitchen C, Bentley WE, Chen S, Kelly DL, Payne GF. Validation of oxidative stress assay for schizophrenia. Schizophr Res 2019; 212:126-133. [PMID: 31399268 DOI: 10.1016/j.schres.2019.07.057] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 05/22/2019] [Accepted: 07/29/2019] [Indexed: 01/27/2023]
Abstract
Accumulating evidence implicates oxidative stress in a range of diseases, yet no objective measurement has emerged that characterizes the global nature of oxidative stress. Previously, we reported a measurement that employs the moderately strong oxidant iridium (Ir) to probe the oxidative damage in a serum sample and reported that in a small study (N = 15) the Ir-reducing capacity assay could distinguish schizophrenia from healthy control groups based on their levels of oxidative stress. Here, we used a larger sample size to evaluate the Ir-reducing capacity assay to assess its ability to discriminate the schizophrenia (N = 73) and healthy control groups (N = 45). Each serum sample was measured (in triplicate) at three different times that were separated by several weeks. The Intraclass Correlation Coefficient (ICC = 0.69) for these repeated measurements indicates the assay detects stable components in the sample (i.e., it is not detecting transient reactive species or air-oxidizable serum components). Correlations between the Ir-reducing capacity assay and independently-measured total serum protein levels (r = +0.74, p < 2.2 × 10-16) suggest the assay is detecting information in the protein pool. For cross-validation of the discrimination ability, we used machine learning and receiver operating characteristic (ROC) analysis. After adjusting for potential confounders (age and smoking status), an area under the curve (AUC) of ROC curve was calculated to be 0.89 (p = 9.3 × 10-5). In conclusion, this validation indicates the Ir-reducing capacity assay provides a simple global measure of oxidative stress, and further supports the hypothesis that oxidative stress is linked with schizophrenia.
Collapse
Affiliation(s)
- Eunkyoung Kim
- Institute for Bioscience & Biotechnology Research, University of Maryland, College Park, MD 20742, USA
| | - Zoe Keskey
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD 21228, USA
| | - Mijeong Kang
- Institute for Bioscience & Biotechnology Research, University of Maryland, College Park, MD 20742, USA
| | - Christopher Kitchen
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD 21228, USA
| | - William E Bentley
- Institute for Bioscience & Biotechnology Research, University of Maryland, College Park, MD 20742, USA
| | - Shuo Chen
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD 21228, USA
| | - Deanna L Kelly
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD 21228, USA
| | - Gregory F Payne
- Institute for Bioscience & Biotechnology Research, University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
79
|
Moslem M, Olive J, Falk A. Stem cell models of schizophrenia, what have we learned and what is the potential? Schizophr Res 2019; 210:3-12. [PMID: 30587427 DOI: 10.1016/j.schres.2018.12.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 12/14/2018] [Accepted: 12/16/2018] [Indexed: 12/13/2022]
Abstract
Schizophrenia is a complex disorder with clinical manifestations in early adulthood. However, it may start with disruption of brain development caused by genetic or environmental factors, or both. Early deteriorating effects of genetic/environmental factors on neural development might be key to described disease causing mechanisms. Establishing cellular models with cells from affected individual using the induced pluripotent stem cells (iPSC) technology could be used to mimic early neurodevelopment alterations caused by risk genes or environmental stressors. Indeed, cellular models have allowed identification and further study of risk factors and the biological pathways in which they are involved. New advancements in differentiation methods such as defined and robust monolayer protocols and cerebral 3D organoids have made it possible to faithfully mimic neural development and neuronal functionality while CRISPR-editing tools assist to engineer isogenic cell lines to precisely explore genetic variation in polygenic diseases such as schizophrenia. Here we review the current field of iPSC models of schizophrenia and how risk factors can be modelled as well as discussing the common biological pathways involved.
Collapse
Affiliation(s)
- Mohsen Moslem
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Jessica Olive
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden; Department of Life Sciences, Imperial College London, United Kingdom.
| | - Anna Falk
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
80
|
A five-year follow-up study of antioxidants, oxidative stress and polyunsaturated fatty acids in schizophrenia. Acta Neuropsychiatr 2019; 31:202-212. [PMID: 31178002 DOI: 10.1017/neu.2019.14] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Oxidative stress and dysregulated antioxidant defence may be involved in the pathophysiology of schizophrenia. In the present study, we investigated changes in antioxidants and oxidative stress from an acute to a later stable phase. We hypothesised that the levels of oxidative markers are increased in schizophrenia compared with healthy controls; change from the acute to the stable phase; and are associated with the levels of membrane polyunsaturated fatty acids (PUFAs) and symptom severity. METHODS Fifty-five patients with schizophrenia spectrum disorders, assessed during an acute phase and 5 years later during a stable phase, and 51 healthy controls were included. We measured antioxidants (α-tocopherol, uric acid, albumin and bilirubin), markers of oxidative stress (F2-isoprostane and reactive oxygen metabolites) and membrane fatty acids. Antioxidants and oxidative stress markers were compared in schizophrenia versus healthy controls, adjusting for differences in sex, age and smoking, and changes over time. Associations between symptoms and PUFA were also investigated. RESULTS In the acute phase, α-tocopherol was significantly higher (p < 0.001), while albumin was lower (p < 0.001) compared with the stable phase. Changes in α-tocopherol were associated with PUFA levels in the acute phase. In the stable phase, schizophrenia patients had higher uric acid (p = 0.009) and lower bilirubin (p = 0.046) than healthy controls. CRP was higher in patients in the stable phase (p < 0.001), and there was no significant change from the acute phase. CONCLUSION The present findings of change in antioxidant levels in the acute versus stable phase of schizophrenia the present findings suggest that redox regulation is dynamic and changes during different phases of the disorder.
Collapse
|
81
|
Valdovinos-Flores C, Limón-Pacheco JH, León-Rodríguez R, Petrosyan P, Garza-Lombó C, Gonsebatt ME. Systemic L-Buthionine -S-R-Sulfoximine Treatment Increases Plasma NGF and Upregulates L-cys/L-cys2 Transporter and γ-Glutamylcysteine Ligase mRNAs Through the NGF/TrkA/Akt/Nrf2 Pathway in the Striatum. Front Cell Neurosci 2019; 13:325. [PMID: 31396052 PMCID: PMC6664075 DOI: 10.3389/fncel.2019.00325] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 07/03/2019] [Indexed: 01/31/2023] Open
Abstract
Glutathione (GSH) is the most abundant intracellular antioxidant. GSH depletion leads to oxidative stress and neuronal damage in the central nervous system (CNS). In mice, the acute systemic inhibition of GSH synthesis by L-buthionine-S-R-sulfoximine (BSO) triggers a protective response and a subsequent increase in the CNS GSH content. This response might be modulated by a peripheral increment of circulating nerve growth factor (NGF). NGF is an important activator of antioxidant pathways mediated by tropomyosin-related kinase receptor A (TrkA). Here, we report that peripheral administration of BSO increased plasma NGF levels. Additionally, BSO increased NGF levels and activated the NGF/TrkA/Akt pathway in striatal neurons. Moreover, the response in the striatum included an increased transcription of nrf2, gclm, lat1, eaac1, and xct, all of which are involved in antioxidant responses, and L-cys/L-cys2 and glutamate transporters. Using antibody against NGF confirmed that peripheral NGF activated the NGF/TrkA/Akt/Nrf2 pathway in the striatum and subsequently increased the transcription of gclm, nrf2, lat1, eaac1, and xct. These results provide evidence that the reduction of peripheral GSH pools increases peripheral NGF circulation that orchestrates a neuroprotective response in the CNS, at least in the striatum, through the NGF/TrkA/Akt/Nrf2 pathway.
Collapse
Affiliation(s)
- Cesar Valdovinos-Flores
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Jorge H Limón-Pacheco
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Renato León-Rodríguez
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Pavel Petrosyan
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Carla Garza-Lombó
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Maria E Gonsebatt
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
82
|
Dowell J, Elser BA, Schroeder RE, Stevens HE. Cellular stress mechanisms of prenatal maternal stress: Heat shock factors and oxidative stress. Neurosci Lett 2019; 709:134368. [PMID: 31299286 DOI: 10.1016/j.neulet.2019.134368] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/19/2019] [Accepted: 07/03/2019] [Indexed: 12/24/2022]
Abstract
Development of the brain prenatally is affected by maternal experience and exposure. Prenatal maternal psychological stress changes brain development and results in increased risk for neuropsychiatric disorders. In this review, multiple levels of prenatal stress mechanisms (offspring brain, placenta, and maternal physiology) are discussed and their intersection with cellular stress mechanisms explicated. Heat shock factors and oxidative stress are closely related to each other and converge with the inflammation, hormones, and cellular development that have been more deeply explored as the basis of prenatal stress risk. Increasing evidence implicates cellular stress mechanisms in neuropsychiatric disorders associated with prenatal stress including affective disorders, schizophrenia, and child-onset psychiatric disorders. Heat shock factors and oxidative stress also have links with the mechanisms involved in other kinds of prenatal stress including external exposures such as environmental toxicants and internal disruptions such as preeclampsia. Integrative understanding of developmental neurobiology with these cellular and physiological mechanisms is necessary to reduce risks and promote healthy brain development.
Collapse
Affiliation(s)
- Jonathan Dowell
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA.
| | - Benjamin A Elser
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Interdisciplinary Graduate Program in Human Toxicology, University of Iowa, Iowa City, IA, USA.
| | - Rachel E Schroeder
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA, USA.
| | - Hanna E Stevens
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Interdisciplinary Graduate Program in Human Toxicology, University of Iowa, Iowa City, IA, USA; Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA, USA; Iowa Neuroscience Institute, Iowa City, IA, USA.
| |
Collapse
|
83
|
Mullier E, Roine T, Griffa A, Xin L, Baumann PS, Klauser P, Cleusix M, Jenni R, Alemàn-Gómez Y, Gruetter R, Conus P, Do KQ, Hagmann P. N-Acetyl-Cysteine Supplementation Improves Functional Connectivity Within the Cingulate Cortex in Early Psychosis: A Pilot Study. Int J Neuropsychopharmacol 2019; 22:478-487. [PMID: 31283822 PMCID: PMC6672595 DOI: 10.1093/ijnp/pyz022] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 04/10/2019] [Accepted: 06/26/2019] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND There is increasing evidence that redox dysregulation, which can lead to oxidative stress and eventually to impairment of oligodendrocytes and parvalbumin interneurons, may underlie brain connectivity alterations in schizophrenia. Accordingly, we previously reported that levels of brain antioxidant glutathione in the medial prefrontal cortex were positively correlated with increased functional connectivity along the cingulum bundle in healthy controls but not in early psychosis patients. In a recent randomized controlled trial, we observed that 6-month supplementation with a glutathione precursor, N-acetyl-cysteine, increased brain glutathione levels and improved symptomatic expression and processing speed. METHODS We investigated the effect of N-acetyl-cysteine supplementation on the functional connectivity between regions of the cingulate cortex, which have been linked to positive symptoms and processing speed decline. In this pilot study, we compared structural connectivity and resting-state functional connectivity between early psychosis patients treated with 6-month N-acetyl-cysteine (n = 9) or placebo (n = 11) supplementation with sex- and age-matched healthy control subjects (n = 74). RESULTS We observed that 6-month N-acetyl-cysteine supplementation increases functional connectivity along the cingulum and more precisely between the caudal anterior part and the isthmus of the cingulate cortex. These functional changes can be partially explained by an increase of centrality of these regions in the functional brain network. CONCLUSIONS N-acetyl-cysteine supplementation has a positive effect on functional connectivity within the cingulate cortex in early psychosis patients. To our knowledge, this is the first study suggesting that increased brain glutathione levels via N-acetyl-cysteine supplementation may improve brain functional connectivity.
Collapse
Affiliation(s)
- Emeline Mullier
- Department of Radiology, Lausanne University Hospital (CHUV), Lausanne, Switzerland,Correspondence: Emeline Mullier, Centre de recherche en Radiologie RC7, CHUV, Rue du Bugnon 46, 1011 Lausanne, Suisse ()
| | - Timo Roine
- Department of Radiology, Lausanne University Hospital (CHUV), Lausanne, Switzerland,Turku Brain and Mind Center, University of Turku, Turku, Finland,Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Alessandra Griffa
- Department of Radiology, Lausanne University Hospital (CHUV), Lausanne, Switzerland,Dutch Connectome Lab, Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research (CNCR), VU Amsterdam, Amsterdam, The Netherlands
| | - Lijing Xin
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Philipp S Baumann
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Paul Klauser
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Martine Cleusix
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Raoul Jenni
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Yasser Alemàn-Gómez
- Department of Radiology, Lausanne University Hospital (CHUV), Lausanne, Switzerland,Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland,Medical Image Analysis Laboratory (MIAL), Centre d’Imagerie BioMédicale (CIBM), Lausanne, Switzerland
| | - Rolf Gruetter
- Laboratory of Functional and Metabolic Imaging, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Philippe Conus
- Treatment and Early Intervention in Psychosis Program (TIPP), Service of General Psychiatry, Department of Psychiatry, Lausanne, Switzerland
| | - Kim Q Do
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland,Treatment and Early Intervention in Psychosis Program (TIPP), Service of General Psychiatry, Department of Psychiatry, Lausanne, Switzerland
| | - Patric Hagmann
- Department of Radiology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| |
Collapse
|
84
|
Kim E, Li J, Kang M, Kelly DL, Chen S, Napolitano A, Panzella L, Shi X, Yan K, Wu S, Shen J, Bentley WE, Payne GF. Redox Is a Global Biodevice Information Processing Modality. PROCEEDINGS OF THE IEEE. INSTITUTE OF ELECTRICAL AND ELECTRONICS ENGINEERS 2019; 107:1402-1424. [PMID: 32095023 PMCID: PMC7036710 DOI: 10.1109/jproc.2019.2908582] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Biology is well-known for its ability to communicate through (i) molecularly-specific signaling modalities and (ii) a globally-acting electrical modality associated with ion flow across biological membranes. Emerging research suggests that biology uses a third type of communication modality associated with a flow of electrons through reduction/oxidation (redox) reactions. This redox signaling modality appears to act globally and has features of both molecular and electrical modalities: since free electrons do not exist in aqueous solution, the electrons must flow through molecular intermediates that can be switched between two states - with electrons (reduced) or without electrons (oxidized). Importantly, this global redox modality is easily accessible through its electrical features using convenient electrochemical instrumentation. In this review, we explain this redox modality, describe our electrochemical measurements, and provide four examples demonstrating that redox enables communication between biology and electronics. The first two examples illustrate how redox probing can acquire biologically relevant information. The last two examples illustrate how redox inputs can transduce biologically-relevant transitions for patterning and the induction of a synbio transceiver for two-hop molecular communication. In summary, we believe redox provides a unique ability to bridge bio-device communication because simple electrochemical methods enable global access to biologically meaningful information. Further, we envision that redox may facilitate the application of information theory to the biological sciences.
Collapse
Affiliation(s)
- Eunkyoung Kim
- Institute for Bioscience & Biotechnology Research, University of Maryland, College Park, MD 20742, USA
| | - Jinyang Li
- Institute for Bioscience & Biotechnology Research, Fischell Department of Bioengineering University of Maryland, College Park, MD 20742, USA
| | - Mijeong Kang
- Institute for Bioscience & Biotechnology Research, University of Maryland, College Park, MD 20742, USA
| | - Deanna L Kelly
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD 21228, USA
| | - Shuo Chen
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD 21228, USA
| | - Alessandra Napolitano
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 4, I-80126 Naples, Italy
| | - Lucia Panzella
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 4, I-80126 Naples, Italy
| | - Xiaowen Shi
- School of Resource and Environmental Science, Hubei Biomass-Resource Chemistry, Environmental Biotechnology Key Laboratory, Wuhan University, Wuhan 430079, China
| | - Kun Yan
- School of Resource and Environmental Science, Hubei Biomass-Resource Chemistry, Environmental Biotechnology Key Laboratory, Wuhan University, Wuhan 430079, China
| | - Si Wu
- School of Resource and Environmental Science, Hubei Biomass-Resource Chemistry, Environmental Biotechnology Key Laboratory, Wuhan University, Wuhan 430079, China
| | - Jana Shen
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| | - William E Bentley
- Institute for Bioscience & Biotechnology Research, Fischell Department of Bioengineering University of Maryland, College Park, MD 20742, USA
| | - Gregory F Payne
- Institute for Bioscience & Biotechnology Research, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
85
|
Abstract
PURPOSE OF REVIEW The objective of this article is to highlight the potential role of the galantamine-memantine combination as a novel antioxidant treatment for schizophrenia. RECENT FINDINGS In addition to the well-known mechanisms of action of galantamine and memantine, these medications also have antioxidant activity. Furthermore, an interplay exists between oxidative stress, inflammation (redox-inflammatory hypothesis), and kynurenine pathway metabolites. Also, there is an interaction between brain-derived neurotrophic factor and oxidative stress in schizophrenia. Oxidative stress may be associated with positive, cognitive, and negative symptoms and impairments in white matter integrity in schizophrenia. The antipsychotic-galantamine-memantine combination may provide a novel strategy in schizophrenia to treat positive, cognitive, and negative symptoms. SUMMARY A "single antioxidant" may be inadequate to counteract the complex cascade of oxidative stress. The galantamine-memantine combination as "double antioxidants" is promising. Hence, randomized controlled trials are warranted with the antipsychotic-galantamine-memantine combination with oxidative stress and antioxidant biomarkers in schizophrenia.
Collapse
|
86
|
Das TK, Javadzadeh A, Dey A, Sabesan P, Théberge J, Radua J, Palaniyappan L. Antioxidant defense in schizophrenia and bipolar disorder: A meta-analysis of MRS studies of anterior cingulate glutathione. Prog Neuropsychopharmacol Biol Psychiatry 2019; 91:94-102. [PMID: 30125624 DOI: 10.1016/j.pnpbp.2018.08.006] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 07/21/2018] [Accepted: 08/13/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND Glutathione [GSH] is a major intracellular antioxidant that disposes peroxides and protects neurons and glial cells from oxidative stress. In both schizophrenia and bipolar disorder, atypical levels of GSH have been demonstrated, particularly in the anterior cingulate cortex (ACC), though no consistent results have emerged due to limitations in sample size. Our objective was to evaluate if GSH levels in the ACC are abnormal in these 2 disorder, when compared to healthy controls. METHODS We reviewed all 1H-MRS studies reporting GSH values for patients satisfying DSM or ICD based criteria for (1) the psychotic disorders - schizophrenia or schizoaffective disorder or (2) bipolar disorder in comparison to a healthy controls (HC) group in the Anterior Cingulate Cortex (ACC) published until June 2018. A random-effects model was used to calculate the pooled effect size. A meta-regression analysis of moderator variables was also undertaken. RESULTS The literature search identified 18 studies with a total sample size of 581 controls, 578 patients with schizophrenia or bipolar disorder. There is a small but significant reduction in ACC GSH in patients with schizophrenia compared to HC (N = 13; RFX SMD =0.26; 95% CI [0.07 to 0.44]; p = 0.008; heterogeneity p = 0.11). There is a significant increase in the ACC GSH concentration in bipolar disorder compared to HC (N = 6; RFX SMD = -0.28, 95% CI [-0.09 to -0.47]; p = 0.003; heterogeneity p = 0.95). CONCLUSIONS We report a small, but significant reduction in GSH concentration in the ACC in schizophrenia, and a similar sized increase in bipolar disorder. A notable limitation is the lack of sufficient data to examine the moderating effect of the symptom profile. Schizophrenia and bipolar disorder have notably different patterns of redox abnormalities in the ACC. Reduced ACC GSH may confer a schizophrenia-like clinical phenotype, while an excess favouring a bipolar disorder-like profile.
Collapse
Affiliation(s)
- Tushar Kanti Das
- Department of Psychiatry, University of Western Ontario, London, ON, Canada; Robarts Research Institute, London, ON. Canada; Lawson Health Research Institute, London, ON. Canada
| | - Alborz Javadzadeh
- Department of Psychiatry, University of Western Ontario, London, ON, Canada
| | - Avyarthana Dey
- Department of Psychiatry, University of Western Ontario, London, ON, Canada; Robarts Research Institute, London, ON. Canada
| | | | - Jean Théberge
- Lawson Health Research Institute, London, ON. Canada; Department of Medical Biophysics, University of Western Ontario, London, ON, Canada; Department of Diagnostic Imaging, St. Joseph's Health Care London, ON, Canada
| | - Joaquim Radua
- FIDMAG Germanes Hospitalàries, CIBERSAM, Sant Boi de Llobregat, Spain; Institute of Psychiatry, King's College London, De Crespigny Park, London,UK; Centre for Psychiatric Research and Education, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Lena Palaniyappan
- Department of Psychiatry, University of Western Ontario, London, ON, Canada; Robarts Research Institute, London, ON. Canada; Lawson Health Research Institute, London, ON. Canada; Department of Medical Biophysics, University of Western Ontario, London, ON, Canada.
| |
Collapse
|
87
|
Lin CH, Lane HY. Early Identification and Intervention of Schizophrenia: Insight From Hypotheses of Glutamate Dysfunction and Oxidative Stress. Front Psychiatry 2019; 10:93. [PMID: 30873052 PMCID: PMC6400883 DOI: 10.3389/fpsyt.2019.00093] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 02/08/2019] [Indexed: 12/22/2022] Open
Abstract
Schizophrenia is a severe mental disorder which leads to functional deterioration. Early detection and intervention are vital for better prognosis. However, the diagnosis of schizophrenia still depends on clinical observation to date. Without reliable biomarkers, schizophrenia is difficult to detect in its early phase. Further, there is no approved medication for prodromal schizophrenia because current antipsychotics fail to show satisfactory efficacy and safety. Therefore, to develop an effective early diagnostic and therapeutic approach for schizophrenia, especially in its prodromal phase, is crucial. Glutamate signaling dysfunction and dysregulation of oxidative stress have been considered to play important roles in schizophrenic prodrome. The N-methyl-D-aspartate receptor (NMDAR) is one of three types of ionotropic glutamate receptors. In this article, we reviewed literature regarding NMDAR hypofunction, oxidative stress, and the linkage between both in prodromal schizophrenia. The efficacy of NMDAR enhancers such as D-amino acid oxidase inhibitor was addressed. Finally, we highlighted potential biomarkers related to NMDAR and oxidative stress regulation, and therefore suggested the strategies of early detection and intervention of prodromal schizophrenia. Future larger-scale studies combining biomarkers and novel drug development for early psychosis are warranted.
Collapse
Affiliation(s)
- Chieh-Hsin Lin
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Hsien-Yuan Lane
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Department of Psychiatry and Brain Disease Research Center, China Medical University Hospital, Taichung, Taiwan
- Department of Psychology, College of Medical and Health Sciences, Asia University, Taichung, Taiwan
| |
Collapse
|
88
|
Ueno H, Suemitsu S, Murakami S, Kitamura N, Wani K, Matsumoto Y, Okamoto M, Ishihara T. Region-specific reduction of parvalbumin neurons and behavioral changes in adult mice following single exposure to cranial irradiation. Int J Radiat Biol 2019; 95:611-625. [PMID: 30601685 DOI: 10.1080/09553002.2019.1564081] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
PURPOSE Ionizing irradiation has several long-term effects including progressive cognitive impairment. Cognitive deterioration generally appears to be caused by abnormalities in the hippocampal dentate gyrus, with abnormal function of parvalbumin-expressing interneurons (PV neurons) in the cerebral cortex. PV neurons are vulnerable to oxidative stress, which can be caused by ionizing irradiation. We speculated that selective impairment of specific brain regions due to ionizing irradiation may alter the degree of cognitive impairment. METHODS We irradiated mature mouse brains with 20 Gy-ionizing irradiation. Subsequently, we analyzed behavioral abnormalities and changes in the number of PV neurons. RESULTS PV neuron density was significantly lower in some cortical regions of irradiated mice than in control mice. Within 1 week of irradiation, both body weight and temperature of irradiated mice decreased. In the forced swim test, irradiated mice spent significantly less time immobile than did control mice. However, irradiated mice did not display any abnormalities in the elevated plus maze test, Y-maze test, tail suspension test, and social interaction test between 3 to 6 days after irradiation. CONCLUSIONS These results suggest that high-dose irradiation is less likely to cause brain dysfunction in the subacute phase. Moreover, the vulnerability of PV neurons appears to be brain-region specific.
Collapse
Affiliation(s)
- Hiroshi Ueno
- a Department of Medical Technology , Kawasaki University of Medical Welfare , Okayama , Japan.,b Department of Medical Technology, Graduate School of Health Sciences , Okayama University , Okayama , Japan
| | - Shunsuke Suemitsu
- c Department of Psychiatry , Kawasaki Medical School , Kurashiki , Japan
| | - Shinji Murakami
- c Department of Psychiatry , Kawasaki Medical School , Kurashiki , Japan
| | - Naoya Kitamura
- c Department of Psychiatry , Kawasaki Medical School , Kurashiki , Japan
| | - Kenta Wani
- c Department of Psychiatry , Kawasaki Medical School , Kurashiki , Japan
| | - Yosuke Matsumoto
- d Department of Neuropsychiatry, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences , Okayama University , Okayama , Japan
| | - Motoi Okamoto
- b Department of Medical Technology, Graduate School of Health Sciences , Okayama University , Okayama , Japan
| | - Takeshi Ishihara
- c Department of Psychiatry , Kawasaki Medical School , Kurashiki , Japan
| |
Collapse
|
89
|
Abstract
Glutamate is the most abundant excitatory neurotransmitter, present at the bulk of cortical synapses, and participating in many physiologic and pathologic processes ranging from learning and memory to stroke. The tripeptide, glutathione, is one-third glutamate and present at up to low millimolar intracellular concentrations in brain, mediating antioxidant defenses and drug detoxification. Because of the substantial amounts of brain glutathione and its rapid turnover under homeostatic control, we hypothesized that glutathione is a relevant reservoir of glutamate and could influence synaptic excitability. We find that drugs that inhibit generation of glutamate by the glutathione cycle elicit decreases in cytosolic glutamate and decreased miniature excitatory postsynaptic potential (mEPSC) frequency. In contrast, pharmacologically decreasing the biosynthesis of glutathione leads to increases in cytosolic glutamate and enhanced mEPSC frequency. The glutathione cycle can compensate for decreased excitatory neurotransmission when the glutamate-glutamine shuttle is inhibited. Glutathione may be a physiologic reservoir of glutamate neurotransmitter.
Collapse
|
90
|
Chaumette B, Kebir O, Pouch J, Ducos B, Selimi F, Gaillard R, Krebs MO. Longitudinal Analyses of Blood Transcriptome During Conversion to Psychosis. Schizophr Bull 2019; 45:247-255. [PMID: 29471546 PMCID: PMC6293211 DOI: 10.1093/schbul/sby009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The biological processes associated with the onset of schizophrenia remain largely unknown. Current hypotheses favor gene × environment interactions as supported by our recent report about DNA methylation changes during the onset of psychosis. Here, we conducted the first longitudinal transcriptomic analysis of blood samples from 31 at-risk individuals who later converted to psychosis and 63 at-risk individuals who did not. Individuals were followed for a maximum of 1 year. Blood samples were collected at baseline and at the end of follow-up and individuals served as their own controls. Differentially expressed genes between the 2 groups were identified using the RNA sequencing of an initial discovery subgroup (n = 15 individuals). The most promising results were replicated using high-throughput real-time qPCR in the whole cohort (n = 94 individuals). We identified longitudinal changes in 4 brain-expressed genes based on RNAseq analysis. One of these genes (CPT1A) was replicated in the whole cohort. The previously observed hypermethylation in NRP1 and GSTM5 during the onset of psychosis correlated with a decrease in corresponding gene expression. RNA sequencing also identified 2 co-expression networks that were impaired after conversion compared with baseline-the Wnt pathway including AKT1, CPT1A and semaphorins, and the Toll-like receptor pathway, related to innate immunity. This longitudinal study of transcriptomic changes in individuals with at-risk mental state revealed alterations during conversion to psychosis in pathways and genes relevant to schizophrenia. These results may be a first step toward better understanding psychosis onset. They may also help to identify new biomarkers and targets for disease-modifying therapeutic strategies.
Collapse
Affiliation(s)
- Boris Chaumette
- Universite Paris Descartes, Université Sorbonne Paris Cite, Paris, France,INSERM, Laboratoire de Physiopathologie des Maladies Psychiatriques, Centre de Psychiatrie et Neurosciences, Paris, France,GDR3557-Institut de Psychiatrie, Paris, France,Centre Hospitalier Sainte-Anne, Service Hospitalo-Universitaire, Paris, France
| | - Oussama Kebir
- Universite Paris Descartes, Université Sorbonne Paris Cite, Paris, France,INSERM, Laboratoire de Physiopathologie des Maladies Psychiatriques, Centre de Psychiatrie et Neurosciences, Paris, France,GDR3557-Institut de Psychiatrie, Paris, France,Centre Hospitalier Sainte-Anne, Service Hospitalo-Universitaire, Paris, France
| | - Juliette Pouch
- Plateforme qPCR-HD-GPC, Ecole Normale Supérieure, Fondation Pierre-Gilles de Gennes, PSL Research University, Paris, France
| | - Bertrand Ducos
- Plateforme qPCR-HD-GPC, Ecole Normale Supérieure, Fondation Pierre-Gilles de Gennes, PSL Research University, Paris, France,Laboratoire de Physique Statistique, Ecole normale Supérieure, PSL Research University, Université Paris-Diderot Sorbonne Paris-Cité, Sorbonne Universités Univ Paris, CNRS UMR, Paris, France
| | - Fekrije Selimi
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR and INSERM U1050, Paris, France
| | - ICAAR study group
- Centre Hospitalier Sainte-Anne, Service Hospitalo-Universitaire, Paris, France
| | - Raphael Gaillard
- Universite Paris Descartes, Université Sorbonne Paris Cite, Paris, France,INSERM, Laboratoire de Physiopathologie des Maladies Psychiatriques, Centre de Psychiatrie et Neurosciences, Paris, France,GDR3557-Institut de Psychiatrie, Paris, France,Centre Hospitalier Sainte-Anne, Service Hospitalo-Universitaire, Paris, France
| | - Marie-Odile Krebs
- Universite Paris Descartes, Université Sorbonne Paris Cite, Paris, France,INSERM, Laboratoire de Physiopathologie des Maladies Psychiatriques, Centre de Psychiatrie et Neurosciences, Paris, France,GDR3557-Institut de Psychiatrie, Paris, France,Centre Hospitalier Sainte-Anne, Service Hospitalo-Universitaire, Paris, France,To whom correspondence should be addressed; Service Hospitalo-Universitaire, Centre Hospitalier Sainte Anne, 7 rue Cabanis, 75014 Paris, France; tel: +33 14 5658 646, fax: +33 14 5658 160, e-mail:
| |
Collapse
|
91
|
Bygrave AM, Kilonzo K, Kullmann DM, Bannerman DM, Kätzel D. Can N-Methyl-D-Aspartate Receptor Hypofunction in Schizophrenia Be Localized to an Individual Cell Type? Front Psychiatry 2019; 10:835. [PMID: 31824347 PMCID: PMC6881463 DOI: 10.3389/fpsyt.2019.00835] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 10/21/2019] [Indexed: 01/07/2023] Open
Abstract
Hypofunction of N-methyl-D-aspartate glutamate receptors (NMDARs), whether caused by endogenous factors like auto-antibodies or mutations, or by pharmacological or genetic manipulations, produces a wide variety of deficits which overlap with-but do not precisely match-the symptom spectrum of schizophrenia. In order to understand how NMDAR hypofunction leads to different components of the syndrome, it is necessary to take into account which neuronal subtypes are particularly affected by it in terms of detrimental functional alterations. We provide a comprehensive overview detailing findings in rodent models with cell type-specific knockout of NMDARs. Regarding inhibitory cortical cells, an emerging model suggests that NMDAR hypofunction in parvalbumin (PV) positive interneurons is a potential risk factor for this disease. PV interneurons display a selective vulnerability resulting from a combination of genetic, cellular, and environmental factors that produce pathological multi-level positive feedback loops. Central to this are two antioxidant mechanisms-NMDAR activity and perineuronal nets-which are themselves impaired by oxidative stress, amplifying disinhibition. However, NMDAR hypofunction in excitatory pyramidal cells also produces a range of schizophrenia-related deficits, in particular maladaptive learning and memory recall. Furthermore, NMDAR blockade in the thalamus disturbs thalamocortical communication, and NMDAR ablation in dopaminergic neurons may provoke over-generalization in associative learning, which could relate to the positive symptom domain. Therefore, NMDAR hypofunction can produce schizophrenia-related effects through an action on various different circuits and cell types.
Collapse
Affiliation(s)
- Alexei M Bygrave
- Department of Neuroscience, Johns Hopkins University, Baltimore, MD, United States
| | - Kasyoka Kilonzo
- Institute of Applied Physiology, Ulm University, Ulm, Germany
| | - Dimitri M Kullmann
- UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - David M Bannerman
- Department of Experimental Psychology, University of Oxford, Oxford, United Kingdom
| | - Dennis Kätzel
- Institute of Applied Physiology, Ulm University, Ulm, Germany
| |
Collapse
|
92
|
van de Looij Y, Larpin C, Cabungcal JH, Sanches EF, Toulotte A, Do KQ, Sizonenko SV. Nutritional Intervention for Developmental Brain Damage: Effects of Lactoferrin Supplementation in Hypocaloric Induced Intrauterine Growth Restriction Rat Pups. Front Endocrinol (Lausanne) 2019; 10:46. [PMID: 30800096 PMCID: PMC6375847 DOI: 10.3389/fendo.2019.00046] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 01/21/2019] [Indexed: 12/13/2022] Open
Abstract
Introduction: Intrauterine Growth Restriction (IUGR) refers to an impaired development of the fetus and hence results in adverse neurodevelopmental and psychiatric consequences later in life. Lactoferrin (Lf) is a glycoprotein present in milk that has already shown neuroprotective effects through its anti-inflammatory and antioxidant properties on impaired developing brains. The aim of this study was to characterize a rat model of IUGR and assess the neuroprotective effect of a nutritional supplementation with bovine Lf during pregnancy and lactation on this model. Methods: A model of 50% gestational caloric restriction (CR) was used. Three groups were designed, and pregnant rats had either ad libitum access to food (control group, CTL) or 50% of the controls' intake (restricted group, IUGR). The diet was isocaloric and supplemented with bovine Lf for the caloric restricted dams (restricted-Lf, IUGR_Lf). At postnatal day 7 and 21, advanced ex-vivo diffusion MRI techniques at 9.4T were used to investigate brain cortical and white matter microstructure. Further, genes and proteins involved in structure (synaptophysin, MBP), microglia (Iba-1), metabolism (MCT2, βCaMKII) and apoptosis (Bcl-2) were analyzed in the cortex and striatum. In the cortex, the number of parvalbumin immunoreactive interneurons and their perineuronal nets were quantified. Behavioral tests were performed at P31. Results: Effects of the CR were significant in the cortex and striatum with reduction of synaptophysin (marker of synaptogenesis) at P7 and MBP (marker of myelin) at P21 in the cortex. Indeed, MCT2 (energy metabolism), Bcl-2 (anti-apoptotic protein) and βCaMKII (synapse activity) expressions were reduced in IUGR groups at P7. In the striatum NG2 (marker of oligodendrocyte precursor cells) and Bcl-2 at P7 as well as βCaMKII at P21 were decreased following IUGR and restored by Lf. Cortical microstructure was impaired following CR with partial effect of Lf. Lf prevented oxidative stress induced parvalbumin interneurons impairments whereas striatum and external capsule showed alterations in microstructure depicted by diffusion MRI, which were also partially reversed by Lf. Discussion and Conclusion: The model of 50% caloric restriction induced mild impairment partially reversed by nutritional intervention using Lf during pregnancy and lactation.
Collapse
Affiliation(s)
- Yohan van de Looij
- Division of Child Development and Growth, Department of Pediatrics, School of Medicine, University of Geneva, Geneva, Switzerland
- Laboratory for Functional and Metabolic Imaging, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Camille Larpin
- Division of Child Development and Growth, Department of Pediatrics, School of Medicine, University of Geneva, Geneva, Switzerland
| | - Jan-Harry Cabungcal
- Department of Psychiatry, Centre for Psychiatric Neuroscience, Lausanne University Hospital, Lausanne, Switzerland
| | - Eduardo F. Sanches
- Division of Child Development and Growth, Department of Pediatrics, School of Medicine, University of Geneva, Geneva, Switzerland
| | - Audrey Toulotte
- Division of Child Development and Growth, Department of Pediatrics, School of Medicine, University of Geneva, Geneva, Switzerland
| | - Kim Q. Do
- Department of Psychiatry, Centre for Psychiatric Neuroscience, Lausanne University Hospital, Lausanne, Switzerland
| | - Stéphane V. Sizonenko
- Division of Child Development and Growth, Department of Pediatrics, School of Medicine, University of Geneva, Geneva, Switzerland
- *Correspondence: Stéphane V. Sizonenko
| |
Collapse
|
93
|
Turkmen R, Akosman MS, Demirel HH. Protective effect of N-acetylcysteine on MK-801-induced testicular oxidative stress in mice. Biomed Pharmacother 2019; 109:1988-1993. [DOI: 10.1016/j.biopha.2018.09.139] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 09/13/2018] [Accepted: 09/26/2018] [Indexed: 02/07/2023] Open
|
94
|
Synaptic loss in schizophrenia: a meta-analysis and systematic review of synaptic protein and mRNA measures. Mol Psychiatry 2019; 24:549-561. [PMID: 29511299 PMCID: PMC6004314 DOI: 10.1038/s41380-018-0041-5] [Citation(s) in RCA: 158] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 01/05/2018] [Accepted: 01/31/2018] [Indexed: 02/06/2023]
Abstract
Although synaptic loss is thought to be core to the pathophysiology of schizophrenia, the nature, consistency and magnitude of synaptic protein and mRNA changes has not been systematically appraised. Our objective was thus to systematically review and meta-analyse findings. The entire PubMed database was searched for studies from inception date to the 1st of July 2017. We selected case-control postmortem studies in schizophrenia quantifying synaptic protein or mRNA levels in brain tissue. The difference in protein and mRNA levels between cases and controls was extracted and meta-analysis conducted. Among the results, we found a significant reduction in synaptophysin in schizophrenia in the hippocampus (effect size: -0.65, p < 0.01), frontal (effect size: -0.36, p = 0.04), and cingulate cortices (effect size: -0.54, p = 0.02), but no significant changes for synaptophysin in occipital and temporal cortices, and no changes for SNAP-25, PSD-95, VAMP, and syntaxin in frontal cortex. There were insufficient studies for meta-analysis of complexins, synapsins, rab3A and synaptotagmin and mRNA measures. Findings are summarised for these, which generally show reductions in SNAP-25, PSD-95, synapsin and rab3A protein levels in the hippocampus but inconsistency in other regions. Our findings of moderate-large reductions in synaptophysin in hippocampus and frontal cortical regions, and a tendency for reductions in other pre- and postsynaptic proteins in the hippocampus are consistent with models that implicate synaptic loss in schizophrenia. However, they also identify potential differences between regions and proteins, suggesting synaptic loss is not uniform in nature or extent.
Collapse
|
95
|
Maguire Á, Hargreaves A, Gill M. Coenzyme Q10 and neuropsychiatric and neurological disorders: relevance for schizophrenia. Nutr Neurosci 2018; 23:756-769. [PMID: 30537908 DOI: 10.1080/1028415x.2018.1556481] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Objective: Mitochondrial dysfunction has been implicated in the pathophysiology of schizophrenia and other neuropsychiatric disorders. Though the exact mechanisms and clinical implications for this dysfunction are not fully determined, there is a hypothesis that deficiency in coenzyme Q10 (CoQ10) may contribute to mitochondrial impairments and be reflected in cognitive, affective, and energy disturbances in the disorders. CoQ10 is a critical component of the mitochondrial respiratory chain and an essential free radical scavenger, necessary for mitochondrial function. Here, we review the results of CoQ10 supplementation interventions for adults with various neurological and neuropsychiatric disorders and consider the therapeutic potential of CoQ10 supplementation for schizophrenia in light of these studies. Methods: A literature review of randomised controlled trials and open-label studies investigating the effect of CoQ10 as a single intervention in adults with neurological and neuropsychiatric disorders was conducted. Results: CoQ10 supplementation has some positive effects on fatigue, cognitive impairment and affective difficulties in several neurological and neuropsychiatric conditions with associated mitochondrial dysfunction. Discussion: CoQ10 may be of therapeutic value to schizophrenia given evidence of mitochondrial dysfunction in the disorder.
Collapse
Affiliation(s)
- Áine Maguire
- Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - April Hargreaves
- Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin, Ireland.,Department of Psychology, National College of Ireland, Dublin, Ireland
| | - Michael Gill
- Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
96
|
Redox dysregulation as a link between childhood trauma and psychopathological and neurocognitive profile in patients with early psychosis. Proc Natl Acad Sci U S A 2018; 115:12495-12500. [PMID: 30455310 PMCID: PMC6298080 DOI: 10.1073/pnas.1812821115] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Early traumatic experiences interact with redox regulation and oxidative stress. Blood glutathione peroxidase (GPx) activity, involved in reducing peroxides, may reflect the oxidation status of the organism, thus allowing for the stratification of patients. Traumatized patients with psychosis who have a high blood oxidation status (high-GPx) have smaller hippocampal volumes (but not a smaller amygdala or intracranial volume), and this is associated with more severe clinical symptoms, while those with a lower oxidation status (low-GPx) showed better cognition and a correlated activation of the antioxidant thioredoxin/peroxiredoxin system. Thus, in patients with psychosis, traumatic experiences during childhood may interact with various redox systems, leading to long-term neuroanatomical and clinical defects. This redox profile may represent important biomarkers for patient stratification, defining treatment strategies at early stages of psychosis. Exposure to childhood trauma (CT) increases the risk for psychosis and affects the development of brain structures, possibly through oxidative stress. As oxidative stress is also linked to psychosis, it may interact with CT, leading to a more severe clinical phenotype. In 133 patients with early psychosis (EPP), we explored the relationships between CT and hippocampal, amygdala, and intracranial volume (ICV); blood antioxidant defenses [glutathione peroxidase (GPx) and thioredoxin/peroxiredoxin (Trx/Prx)]; psychopathological results; and neuropsychological results. Nonadjusted hippocampal volume correlated negatively with GPx activity in patients with CT, but not in patients without CT. In patients with CT with high GPx activity (high-GPx+CT), hippocampal volume was decreased compared with that in patients with low-GPx+CT and patients without CT, who had similar hippocampal volumes. Patients with high-GPx+CT had more severe positive and disorganized symptoms than other patients. Interestingly, Trx and oxidized Prx levels correlated negatively with GPx only in patients with low-GPx+CT. Moreover, patients with low-GPx+CT performed better than other patients on cognitive tasks. Discriminant analysis combining redox markers, hippocampal volume, clinical scores, and cognitive scores allowed for stratification of the patients into subgroups. In conclusion, traumatized EPP with high peripheral oxidation status (high-GPx activity) had smaller hippocampal volumes and more severe symptoms, while those with lower oxidation status (low-GPx activity) showed better cognition and regulation of GPx and Trx/Prx systems. These results suggest that maintained regulation of various antioxidant systems allowed for compensatory mechanisms preventing long-term neuroanatomical and clinical impacts. The redox marker profile may thus represent important biomarkers for defining treatment strategies in patients with psychosis.
Collapse
|
97
|
Rampon C, Volovitch M, Joliot A, Vriz S. Hydrogen Peroxide and Redox Regulation of Developments. Antioxidants (Basel) 2018; 7:E159. [PMID: 30404180 PMCID: PMC6262372 DOI: 10.3390/antiox7110159] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 10/10/2018] [Accepted: 10/10/2018] [Indexed: 01/16/2023] Open
Abstract
Reactive oxygen species (ROS), which were originally classified as exclusively deleterious compounds, have gained increasing interest in the recent years given their action as bona fide signalling molecules. The main target of ROS action is the reversible oxidation of cysteines, leading to the formation of disulfide bonds, which modulate protein conformation and activity. ROS, endowed with signalling properties, are mainly produced by NADPH oxidases (NOXs) at the plasma membrane, but their action also involves a complex machinery of multiple redox-sensitive protein families that differ in their subcellular localization and their activity. Given that the levels and distribution of ROS are highly dynamic, in part due to their limited stability, the development of various fluorescent ROS sensors, some of which are quantitative (ratiometric), represents a clear breakthrough in the field and have been adapted to both ex vivo and in vivo applications. The physiological implication of ROS signalling will be presented mainly in the frame of morphogenetic processes, embryogenesis, regeneration, and stem cell differentiation. Gain and loss of function, as well as pharmacological strategies, have demonstrated the wide but specific requirement of ROS signalling at multiple stages of these processes and its intricate relationship with other well-known signalling pathways.
Collapse
Affiliation(s)
- Christine Rampon
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, 75231 Paris, France.
- Sorbonne Paris Cité, Univ Paris Diderot, Biology Department, 75205 Paris CEDEX 13, France.
| | - Michel Volovitch
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, 75231 Paris, France.
- École Normale Supérieure, Department of Biology, PSL Research University, 75005 Paris, France.
| | - Alain Joliot
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, 75231 Paris, France.
| | - Sophie Vriz
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, 75231 Paris, France.
- Sorbonne Paris Cité, Univ Paris Diderot, Biology Department, 75205 Paris CEDEX 13, France.
| |
Collapse
|
98
|
Ximenes NC, Dos Santos Júnior MA, Vasconcelos GS, Dias KCF, Jucá MM, Silva AH, Leal LKAM, Viana GSB, de Sousa FCF, Vasconcelos SMM. Ethanolic extract of Erythrina velutina Willd ameliorate schizophrenia-like behavior induced by ketamine in mice. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2018; 16:/j/jcim.ahead-of-print/jcim-2018-0038/jcim-2018-0038.xml. [PMID: 30315736 DOI: 10.1515/jcim-2018-0038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 09/12/2018] [Indexed: 06/08/2023]
Abstract
Background Schizophrenia is a chronic mental disorder, characterized by positive, negative and cognitive symptoms. In general, several plants have shown activity in diseases related to the central nervous system (e.g., Erythrina velutina (EEEV), also known as "mulungu"). For this reason, we aimed to investigate the effects of standardized ethanol extract obtained from the stem bark of EEEV on the schizophrenia-like behaviors induced by ketamine (KET) administration. Methods Swiss mice were treated with KET (20 mg/kg, i.p.) or saline for 14 days. In addition, from 8th to 14th days, saline, EEEV (200 or 400 mg/kg, p.o.) or olanzapine (OLAN 2 mg/kg, p.o.) were associated to the protocol. On the 14th day of treatment, schizophrenia-like symptoms were evaluated by the prepulse inhibition of the startle reflex (PPI), locomotor activity evaluated by the open field test (OFT), spatial recognition memory evaluated by the Y-maze task and social interaction test (SIT). Results KET has caused deficits in PPI, and it has also has caused hyperlocomotion in OFT and deficits in SIT as compared to control. EEEV in both doses used, reversed behavioral changes induced by KET, likewise results obtained with the administration of OLAN. Conclusions Taken together, the results demonstrate that the standard extract of EEEV was able to revert schizophrenia-like symptoms, due to the administration in repeated doses of ketamine. Thus, our findings lead to a new perspective for the use of EEEV an interesting alternative for drug discovery in schizophrenia.
Collapse
Affiliation(s)
- Naiara Coelho Ximenes
- Department of Physiology and Pharmacology, Federal University of Ceara, Cel. Nunes de Melo 1127, CEP 60431-270, Fortaleza, Brazil
| | - Manuel Alves Dos Santos Júnior
- Department of Physiology and Pharmacology, Federal University of Ceara, Cel. Nunes de Melo 1127, CEP 60431-270, Fortaleza, Brazil
| | - Germana Silva Vasconcelos
- Department of Physiology and Pharmacology, Federal University of Ceara, Cel. Nunes de Melo 1127, CEP 60431-270, Fortaleza, Brazil
| | - Kátia Cilene Ferreira Dias
- Department of Physiology and Pharmacology, Federal University of Ceara, Cel. Nunes de Melo 1127, CEP 60431-270, Fortaleza, Brazil
| | - Mércia Marques Jucá
- Department of Physiology and Pharmacology, Federal University of Ceara, Cel. Nunes de Melo 1127, CEP 60431-270, Fortaleza, Brazil
| | - Aline Holanda Silva
- Department of Pharmacy, Federal University of Ceará, Capitão Francisco Pedro Street 1210, Fortaleza, CE CEP: 60430-170, Brazil
| | | | - Glauce Socorro Barros Viana
- Department of Physiology and Pharmacology, Federal University of Ceara, Cel. Nunes de Melo 1127, CEP 60431-270, Fortaleza, Brazil
| | - Francisca Cléa Florenço de Sousa
- Department of Physiology and Pharmacology, Federal University of Ceara, Cel. Nunes de Melo 1127, CEP 60431-270, Fortaleza, Brazil
| | | |
Collapse
|
99
|
Klauser P, Xin L, Fournier M, Griffa A, Cleusix M, Jenni R, Cuenod M, Gruetter R, Hagmann P, Conus P, Baumann PS, Do KQ. N-acetylcysteine add-on treatment leads to an improvement of fornix white matter integrity in early psychosis: a double-blind randomized placebo-controlled trial. Transl Psychiatry 2018; 8:220. [PMID: 30315150 PMCID: PMC6185923 DOI: 10.1038/s41398-018-0266-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 09/05/2018] [Accepted: 09/10/2018] [Indexed: 11/09/2022] Open
Abstract
Mechanism-based treatments for schizophrenia are needed, and increasing evidence suggests that oxidative stress may be a target. Previous research has shown that N-acetylcysteine (NAC), an antioxidant and glutathione (GSH) precursor almost devoid of side effects, improved negative symptoms, decreased the side effects of antipsychotics, and improved mismatch negativity and local neural synchronization in chronic schizophrenia. In a recent double-blind randomized placebo-controlled trial by Conus et al., early psychosis patients received NAC add-on therapy (2700 mg/day) for 6 months. Compared with placebo-treated controls, NAC patients showed significant improvements in neurocognition (processing speed) and a reduction of positive symptoms among patients with high peripheral oxidative status. NAC also led to a 23% increase in GSH levels in the medial prefrontal cortex (GSHmPFC) as measured by 1H magnetic resonance spectroscopy. A subgroup of the patients in this study were also scanned with multimodal MR imaging (spectroscopy, diffusion, and structural) at baseline (prior to NAC/placebo) and after 6 months of add-on treatment. Based on prior translational research, we hypothesized that NAC would protect white matter integrity in the fornix. A group × time interaction indicated a difference in the 6-month evolution of white matter integrity (as measured by generalized fractional anisotropy, gFA) in favor of the NAC group, which showed an 11% increase. The increase in gFA correlated with an increase in GSHmPFC over the same 6-month period. In this secondary study, we suggest that NAC add-on treatment may be a safe and effective way to protect white matter integrity in early psychosis patients.
Collapse
Affiliation(s)
- Paul Klauser
- 0000 0001 0423 4662grid.8515.9Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland ,0000 0001 0423 4662grid.8515.9Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland ,National Center of Competence in Research (NCCR) “SYNAPSY – The Synaptic Bases of Mental Diseases”, Lausanne, Switzerland
| | - Lijing Xin
- 0000000121839049grid.5333.6Animal Imaging and Technology Core (AIT), Center for Biomedical Imaging (CIBM), Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Margot Fournier
- 0000 0001 0423 4662grid.8515.9Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland ,National Center of Competence in Research (NCCR) “SYNAPSY – The Synaptic Bases of Mental Diseases”, Lausanne, Switzerland
| | - Alessandra Griffa
- 0000 0001 0423 4662grid.8515.9Department of Radiology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland ,Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Martine Cleusix
- 0000 0001 0423 4662grid.8515.9Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland ,National Center of Competence in Research (NCCR) “SYNAPSY – The Synaptic Bases of Mental Diseases”, Lausanne, Switzerland
| | - Raoul Jenni
- 0000 0001 0423 4662grid.8515.9Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland ,National Center of Competence in Research (NCCR) “SYNAPSY – The Synaptic Bases of Mental Diseases”, Lausanne, Switzerland
| | - Michel Cuenod
- 0000 0001 0423 4662grid.8515.9Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Rolf Gruetter
- 0000000121839049grid.5333.6Animal Imaging and Technology Core (AIT), Center for Biomedical Imaging (CIBM), Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland ,0000 0001 0423 4662grid.8515.9Department of Radiology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Patric Hagmann
- National Center of Competence in Research (NCCR) “SYNAPSY – The Synaptic Bases of Mental Diseases”, Lausanne, Switzerland ,0000 0001 0423 4662grid.8515.9Department of Radiology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Philippe Conus
- 0000 0001 0423 4662grid.8515.9Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland ,National Center of Competence in Research (NCCR) “SYNAPSY – The Synaptic Bases of Mental Diseases”, Lausanne, Switzerland
| | - Philipp S. Baumann
- 0000 0001 0423 4662grid.8515.9Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland ,0000 0001 0423 4662grid.8515.9Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland ,National Center of Competence in Research (NCCR) “SYNAPSY – The Synaptic Bases of Mental Diseases”, Lausanne, Switzerland
| | - Kim Q. Do
- 0000 0001 0423 4662grid.8515.9Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland ,National Center of Competence in Research (NCCR) “SYNAPSY – The Synaptic Bases of Mental Diseases”, Lausanne, Switzerland
| |
Collapse
|
100
|
de Almeida V, Martins-de-Souza D. Cannabinoids and glial cells: possible mechanism to understand schizophrenia. Eur Arch Psychiatry Clin Neurosci 2018; 268:727-737. [PMID: 29392440 DOI: 10.1007/s00406-018-0874-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 01/24/2018] [Indexed: 01/03/2023]
Abstract
Clinical and neurobiological findings have reported the involvement of endocannabinoid signaling in the pathophysiology of schizophrenia. This system modulates dopaminergic and glutamatergic neurotransmission that is associated with positive, negative, and cognitive symptoms of schizophrenia. Despite neurotransmitter impairments, increasing evidence points to a role of glial cells in schizophrenia pathobiology. Glial cells encompass three main groups: oligodendrocytes, microglia, and astrocytes. These cells promote several neurobiological functions, such as myelination of axons, metabolic and structural support, and immune response in the central nervous system. Impairments in glial cells lead to disruptions in communication and in the homeostasis of neurons that play role in pathobiology of disorders such as schizophrenia. Therefore, data suggest that glial cells may be a potential pharmacological tool to treat schizophrenia and other brain disorders. In this regard, glial cells express cannabinoid receptors and synthesize endocannabinoids, and cannabinoid drugs affect some functions of these cells that can be implicated in schizophrenia pathobiology. Thus, the aim of this review is to provide data about the glial changes observed in schizophrenia, and how cannabinoids could modulate these alterations.
Collapse
Affiliation(s)
- Valéria de Almeida
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Monteiro Lobato 255, Campinas, SP, 13083-862, Brazil.
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Monteiro Lobato 255, Campinas, SP, 13083-862, Brazil.,Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBION), Conselho Nacional de Desenvolvimento Científico e Tecnológico, São Paulo, Brazil
| |
Collapse
|