1
|
Ratsika A, Codagnone MG, Bastiaanssen TFS, Hoffmann Sarda FA, Lynch CMK, Ventura-Silva AP, Rosell-Cardona C, Caputi V, Stanton C, Fülling C, Clarke G, Cryan JF. Maternal high-fat diet-induced microbiota changes are associated with alterations in embryonic brain metabolites and adolescent behaviour. Brain Behav Immun 2024; 121:317-330. [PMID: 39032541 DOI: 10.1016/j.bbi.2024.07.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 07/09/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024] Open
Abstract
The developing central nervous system is highly sensitive to nutrient changes during the perinatal period, emphasising the potential impact of alterations of maternal diet on offspring brain development and behaviour. A growing body of research implicates the gut microbiota in neurodevelopment and behaviour. Maternal overweight and obesity during the perinatal period has been linked to changes in neurodevelopment, plasticity and affective disorders in the offspring, with implications for microbial signals from the maternal gut. Here we investigate the impact of maternal high-fat diet (mHFD)-induced changes in microbial signals on offspring brain development, and neuroimmune signals, and the enduring effects on behaviour into adolescence. We first demonstrate that maternal caecal microbiota composition at term pregnancy (embryonic day 18: E18) differs significantly in response to maternal diet. Moreover, mHFD resulted in the upregulation of microbial genes in the maternal intestinal tissue linked to alterations in quinolinic acid synthesis and elevated kynurenine levels in the maternal plasma, both neuronal plasticity mediators related to glutamate metabolism. Metabolomics of mHFD embryonic brains at E18 also detected molecules linked to glutamate-glutamine cycle, including glutamic acid, glutathione disulphide, and kynurenine. During adolescence, the mHFD offspring exhibited increased locomotor activity and anxiety-like behaviour in a sex-dependent manner, along with upregulation of glutamate-related genes compared to controls. Overall, our results demonstrate that maternal exposure to high-fat diet results in microbiota changes, behavioural imprinting, altered brain metabolism, and glutamate signalling during critical developmental windows during the perinatal period.
Collapse
Affiliation(s)
- Anna Ratsika
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland
| | - Martin G Codagnone
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland
| | - Thomaz F S Bastiaanssen
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland
| | - Fabiana A Hoffmann Sarda
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Caoimhe M K Lynch
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland
| | - Ana Paula Ventura-Silva
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland
| | - Cristina Rosell-Cardona
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland
| | - Valentina Caputi
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland
| | | | - Christine Fülling
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork T12YT20, Ireland
| | - John F Cryan
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland.
| |
Collapse
|
2
|
Pristner M, Wasinger D, Seki D, Klebermaß-Schrehof K, Berger A, Berry D, Wisgrill L, Warth B. Neuroactive metabolites and bile acids are altered in extremely premature infants with brain injury. Cell Rep Med 2024; 5:101480. [PMID: 38518769 PMCID: PMC11031385 DOI: 10.1016/j.xcrm.2024.101480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 10/02/2023] [Accepted: 02/27/2024] [Indexed: 03/24/2024]
Abstract
The gut microbiome is associated with pathological neurophysiological evolvement in extremely premature infants suffering from brain injury. The exact underlying mechanism and its associated metabolic signatures in infants are not fully understood. To decipher metabolite profiles linked to neonatal brain injury, we investigate the fecal and plasma metabolome of samples obtained from a cohort of 51 extremely premature infants at several time points, using liquid chromatography (LC)-high-resolution mass spectrometry (MS)-based untargeted metabolomics and LC-MS/MS-based targeted analysis for investigating bile acids and amidated bile acid conjugates. The data are integrated with 16S rRNA gene amplicon gut microbiome profiles as well as patient cytokine, growth factor, and T cell profiles. We find an early onset of differentiation in neuroactive metabolites between infants with and without brain injury. We detect several bacterially derived bile acid amino acid conjugates in plasma and feces. These results provide insights into the early-life metabolome of extremely premature infants.
Collapse
Affiliation(s)
- Manuel Pristner
- Department of Food Chemistry and Toxicology, University of Vienna, 1090 Vienna, Austria
| | - Daniel Wasinger
- Department of Food Chemistry and Toxicology, University of Vienna, 1090 Vienna, Austria
| | - David Seki
- Center for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, 1090 Vienna, Austria; Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, 1090 Vienna, Austria
| | - Katrin Klebermaß-Schrehof
- Department of Pediatrics and Adolescent Medicine, Division of Neonatology, Pediatric Intensive Care and Neuropediatrics, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria
| | - Angelika Berger
- Department of Pediatrics and Adolescent Medicine, Division of Neonatology, Pediatric Intensive Care and Neuropediatrics, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria
| | - David Berry
- Center for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, 1090 Vienna, Austria; Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, 1090 Vienna, Austria
| | - Lukas Wisgrill
- Department of Pediatrics and Adolescent Medicine, Division of Neonatology, Pediatric Intensive Care and Neuropediatrics, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria
| | - Benedikt Warth
- Department of Food Chemistry and Toxicology, University of Vienna, 1090 Vienna, Austria.
| |
Collapse
|
3
|
Kolar D, Krajcovic B, Kleteckova L, Kuncicka D, Vales K, Brozka H. Review: Genes Involved in Mitochondrial Physiology Within 22q11.2 Deleted Region and Their Relevance to Schizophrenia. Schizophr Bull 2023; 49:1637-1653. [PMID: 37379469 PMCID: PMC10686339 DOI: 10.1093/schbul/sbad066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/30/2023]
Abstract
BACKGROUND AND HYPOTHESIS Schizophrenia is associated with altered energy metabolism, but the cause and potential impact of these metabolic changes remain unknown. 22q11.2 deletion syndrome (22q11.2DS) represents a genetic risk factor for schizophrenia, which is associated with the loss of several genes involved in mitochondrial physiology. Here we examine how the haploinsufficiency of these genes could contribute to the emergence of schizophrenia in 22q11.2DS. STUDY DESIGN We characterize changes in neuronal mitochondrial function caused by haploinsufficiency of mitochondria-associated genes within the 22q11.2 region (PRODH, MRPL40, TANGO2, ZDHHC8, SLC25A1, TXNRD2, UFD1, and DGCR8). For that purpose, we combine data from 22q11.2DS carriers and schizophrenia patients, in vivo (animal models) and in vitro (induced pluripotent stem cells, IPSCs) studies. We also review the current knowledge about seven non-coding microRNA molecules located in the 22q11.2 region that may be indirectly involved in energy metabolism by acting as regulatory factors. STUDY RESULTS We found that the haploinsufficiency of genes of interest is mainly associated with increased oxidative stress, altered energy metabolism, and calcium homeostasis in animal models. Studies on IPSCs from 22q11.2DS carriers corroborate findings of deficits in the brain energy metabolism, implying a causal role between impaired mitochondrial function and the development of schizophrenia in 22q11.2DS. CONCLUSIONS The haploinsufficiency of genes within the 22q11.2 region leads to multifaceted mitochondrial dysfunction with consequences to neuronal function, viability, and wiring. Overlap between in vitro and in vivo studies implies a causal role between impaired mitochondrial function and the development of schizophrenia in 22q11.2DS.
Collapse
Affiliation(s)
- David Kolar
- National Institute of Mental Health, Klecany, Czech Republic
| | - Branislav Krajcovic
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
- Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | | | - Daniela Kuncicka
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Karel Vales
- National Institute of Mental Health, Klecany, Czech Republic
| | - Hana Brozka
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
4
|
Carletti B, Banaj N, Piras F, Bossù P. Schizophrenia and Glutathione: A Challenging Story. J Pers Med 2023; 13:1526. [PMID: 38003841 PMCID: PMC10672475 DOI: 10.3390/jpm13111526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/13/2023] [Accepted: 10/20/2023] [Indexed: 11/26/2023] Open
Abstract
Schizophrenia (SZ) is a devastating mental illness with a complex and heterogeneous clinical state. Several conditions like symptoms, stage and severity of the disease are only some of the variables that have to be considered to define the disorder and its phenotypes. SZ pathophysiology is still unclear, and the diagnosis is currently relegated to the analysis of clinical symptoms; therefore, the search for biomarkers with diagnostic relevance is a major challenge in the field, especially in the era of personalized medicine. Though the mechanisms implicated in SZ are not fully understood, some processes are beginning to be elucidated. Oxidative stress, and in particular glutathione (GSH) dysregulation, has been demonstrated to play a crucial role in SZ pathophysiology. In fact, glutathione is a leading actor of oxidative-stress-mediated damage in SZ and appears to reflect the heterogeneity of the disease. The literature reports differing results regarding the levels of glutathione in SZ patients. However, each GSH state may be a sign of specific symptoms or groups of symptoms, candidating glutathione as a biomarker useful for discriminating SZ phenotypes. Here, we summarize the literature about the levels of glutathione in SZ and analyze the role of this molecule and its potential use as a biomarker.
Collapse
Affiliation(s)
- Barbara Carletti
- Laboratory of Neuropsychiatry, Clinical Neuroscience and Neurorehabilitation Department, IRCCS Santa Lucia Foundation, Via Ardeatina 306, 00179 Rome, Italy; (N.B.); (F.P.)
| | - Nerisa Banaj
- Laboratory of Neuropsychiatry, Clinical Neuroscience and Neurorehabilitation Department, IRCCS Santa Lucia Foundation, Via Ardeatina 306, 00179 Rome, Italy; (N.B.); (F.P.)
| | - Fabrizio Piras
- Laboratory of Neuropsychiatry, Clinical Neuroscience and Neurorehabilitation Department, IRCCS Santa Lucia Foundation, Via Ardeatina 306, 00179 Rome, Italy; (N.B.); (F.P.)
| | - Paola Bossù
- Laboratory of Experimental Neuropsychobiology, Clinical Neuroscience and Neurorehabilitation Department, IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143 Rome, Italy;
| |
Collapse
|
5
|
Karanikas E. The Gordian knot of the immune-redox systems' interactions in psychosis. Int Clin Psychopharmacol 2023; 38:285-296. [PMID: 37351570 DOI: 10.1097/yic.0000000000000481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/24/2023]
Abstract
During the last decades the attempt to enlighten the pathobiological substrate of psychosis, from merely focusing on neurotransmitters, has expanded into new areas like the immune and redox systems. Indeed, the inflammatory hypothesis concerning psychosis etiopathology has exponentially grown with findings reflecting dysfunction/aberration of the immune/redox systems' effector components namely cytokines, chemokines, CRP, complement system, antibodies, pro-/anti-oxidants, oxidative stress byproducts just to name a few. Yet, we still lie far from comprehending the underlying cellular mechanisms, their causality directions, and the moderating/mediating parameters affecting these systems; let alone the inter-systemic (between immune and redox) interactions. Findings from preclinical studies on the stress field have provided evidence indicative of multifaceted interactions among the immune and redox components so tightly intertwined as a Gordian knot. Interestingly the literature concerning the interactions between these same systems in the context of psychosis appears minimal (if not absent) and ambiguous. This review attempts to draw a frame of the immune-redox systems' interactions starting from basic research on the stress field and expanding on clinical studies with cohorts with psychosis, hoping to instigate new avenues of research.
Collapse
Affiliation(s)
- Evangelos Karanikas
- Department of Psychiatry, 424 General Military Hospital, Ring Road, Nea Efkarpia, Thessaloniki, Greece
| |
Collapse
|
6
|
Fesharaki-Zadeh A. Oxidative Stress in Traumatic Brain Injury. Int J Mol Sci 2022; 23:ijms232113000. [PMID: 36361792 PMCID: PMC9657447 DOI: 10.3390/ijms232113000] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 10/21/2022] [Accepted: 10/25/2022] [Indexed: 11/17/2022] Open
Abstract
Traumatic Brain Injury (TBI) remains a major cause of disability worldwide. It involves a complex neurometabolic cascade, including oxidative stress. The products of this manuscript is examining the underlying pathophysiological mechanism, including reactive oxygen species (ROS) and reactive nitrogen species (RNS). This process in turn leads to secondary injury cascade, which includes lipid peroxidation products. These reactions ultimately play a key role in chronic inflammation and synaptic dysfunction in a synergistic fashion. Although there are no FDA approved antioxidant therapy for TBI, there is a number of antioxidant therapies that have been tested and include free radical scavengers, activators of antioxidant systems, inhibitors of free radical generating enzymes, and antioxidant enzymes. Antioxidant therapies have led to cognitive and functional recovery post TBI, and they offer a promising treatment option for patients recovering from TBI. Current major challenges in treatment of TBI symptoms include heterogenous nature of injury, as well as access to timely treatment post injury. The inherent benefits of antioxidant therapies include minimally reported side effects, and relative ease of use in the clinical setting. The current review also provides a highlight of the more studied anti-oxidant regimen with applicability for TBI treatment with potential use in the real clinical setting.
Collapse
Affiliation(s)
- Arman Fesharaki-Zadeh
- Yale School of Medicine, Department of Neurology, Yale University, New Haven, CT 06510, USA
| |
Collapse
|
7
|
Schnider M, Jenni R, Ramain J, Camporesi S, Golay P, Alameda L, Conus P, Do KQ, Steullet P. Time of exposure to social defeat stress during childhood and adolescence and redox dysregulation on long-lasting behavioral changes, a translational study. Transl Psychiatry 2022; 12:413. [PMID: 36163247 PMCID: PMC9512907 DOI: 10.1038/s41398-022-02183-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/11/2022] [Accepted: 09/13/2022] [Indexed: 11/09/2022] Open
Abstract
Traumatic events during childhood/early adolescence can cause long-lasting physiological and behavioral changes with increasing risk for psychiatric conditions including psychosis. Genetic factors and trauma (and their type, degree of repetition, time of occurrence) are believed to influence how traumatic experiences affect an individual. Here, we compared long-lasting behavioral effects of repeated social defeat stress (SD) applied during either peripuberty or late adolescence in adult male WT and Gclm-KO mice, a model of redox dysregulation relevant to schizophrenia. As SD disrupts redox homeostasis and causes oxidative stress, we hypothesized that KO mice would be particularly vulnerable to such stress. We first found that peripubertal and late adolescent SD led to different behavioral outcomes. Peripubertal SD induced anxiety-like behavior in anxiogenic environments, potentiated startle reflex, and increased sensitivity to the NMDA-receptor antagonist, MK-801. In contrast, late adolescent SD led to increased exploration in novel environments. Second, the long-lasting impact of peripubertal but not late adolescent SD differed in KO and WT mice. Peripubertal SD increased anxiety-like behavior in anxiogenic environments and MK-801-sensitivity mostly in KO mice, while it increased startle reflex in WT mice. These suggest that a redox dysregulation during peripuberty interacts with SD to remodel the trajectory of brain maturation, but does not play a significant role during later SD. As peripubertal SD induced persisting anxiety- and fear-related behaviors in male mice, we then investigated anxiety in a cohort of 89 early psychosis male patients for whom we had information about past abuse and clinical assessment during the first year of psychosis. We found that a first exposure to physical/sexual abuse (analogous to SD) before age 12, but not after, was associated with higher anxiety at 6-12 months after psychosis onset. This supports that childhood/peripuberty is a vulnerable period during which physical/sexual abuse in males has wide and long-lasting consequences.
Collapse
Affiliation(s)
- Mirko Schnider
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), 1008, Prilly-Lausanne, Switzerland
| | - Raoul Jenni
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), 1008, Prilly-Lausanne, Switzerland
| | - Julie Ramain
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital (CHUV), 1008, Prilly-Lausanne, Switzerland
| | - Sara Camporesi
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital (CHUV), 1008, Prilly-Lausanne, Switzerland
| | - Philippe Golay
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital (CHUV), 1008, Prilly-Lausanne, Switzerland
| | - Luis Alameda
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital (CHUV), 1008, Prilly-Lausanne, Switzerland
| | - Philippe Conus
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital (CHUV), 1008, Prilly-Lausanne, Switzerland
| | - Kim Q Do
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), 1008, Prilly-Lausanne, Switzerland
| | - Pascal Steullet
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), 1008, Prilly-Lausanne, Switzerland.
| |
Collapse
|
8
|
Giron LB, Peluso MJ, Ding J, Kenny G, Zilberstein NF, Koshy J, Hong KY, Rasmussen H, Miller GE, Bishehsari F, Balk RA, Moy JN, Hoh R, Lu S, Goldman AR, Tang HY, Yee BC, Chenna A, Winslow JW, Petropoulos CJ, Kelly JD, Wasse H, Martin JN, Liu Q, Keshavarzian A, Landay A, Deeks SG, Henrich TJ, Abdel-Mohsen M. Markers of fungal translocation are elevated during post-acute sequelae of SARS-CoV-2 and induce NF-κB signaling. JCI Insight 2022; 7:e160989. [PMID: 35727635 PMCID: PMC9462470 DOI: 10.1172/jci.insight.160989] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 06/17/2022] [Indexed: 11/24/2022] Open
Abstract
Long COVID, a type of post-acute sequelae of SARS-CoV-2 (PASC), has been associated with sustained elevated levels of immune activation and inflammation. However, the mechanisms that drive this inflammation remain unknown. Inflammation during acute coronavirus disease 2019 could be exacerbated by microbial translocation (from the gut and/or lung) to blood. Whether microbial translocation contributes to inflammation during PASC is unknown. We did not observe a significant elevation in plasma markers of bacterial translocation during PASC. However, we observed higher levels of fungal translocation - measured as β-glucan, a fungal cell wall polysaccharide - in the plasma of individuals experiencing PASC compared with those without PASC or SARS-CoV-2-negative controls. The higher β-glucan correlated with higher inflammation and elevated levels of host metabolites involved in activating N-methyl-d-aspartate receptors (such as metabolites within the tryptophan catabolism pathway) with established neurotoxic properties. Mechanistically, β-glucan can directly induce inflammation by binding to myeloid cells (via Dectin-1) and activating Syk/NF-κB signaling. Using a Dectin-1/NF-κB reporter model, we found that plasma from individuals experiencing PASC induced higher NF-κB signaling compared with plasma from negative controls. This higher NF-κB signaling was abrogated by piceatannol (Syk inhibitor). These data suggest a potential targetable mechanism linking fungal translocation and inflammation during PASC.
Collapse
Affiliation(s)
| | | | - Jianyi Ding
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Grace Kenny
- Centre for Experimental Pathogen Host Research, University College Dublin, Dublin, Ireland
| | | | - Jane Koshy
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Kai Ying Hong
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | | | | | - Faraz Bishehsari
- Department of Internal Medicine, Rush University, Chicago, Illinois, USA
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University, Chicago, Illinois, USA
| | - Robert A. Balk
- Department of Internal Medicine, Rush University, Chicago, Illinois, USA
| | - James N. Moy
- Department of Internal Medicine, Rush University, Chicago, Illinois, USA
| | | | - Scott Lu
- UCSF, San Francisco, California, USA
| | | | - Hsin-Yao Tang
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Brandon C. Yee
- Monogram Biosciences, Inc., Labcorp, South San Francisco, California, USA
| | - Ahmed Chenna
- Monogram Biosciences, Inc., Labcorp, South San Francisco, California, USA
| | - John W. Winslow
- Monogram Biosciences, Inc., Labcorp, South San Francisco, California, USA
| | | | | | - Haimanot Wasse
- Department of Internal Medicine, Rush University, Chicago, Illinois, USA
| | | | - Qin Liu
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Ali Keshavarzian
- Department of Internal Medicine, Rush University, Chicago, Illinois, USA
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University, Chicago, Illinois, USA
| | - Alan Landay
- Department of Internal Medicine, Rush University, Chicago, Illinois, USA
| | | | | | | |
Collapse
|
9
|
Cuenod M, Steullet P, Cabungcal JH, Dwir D, Khadimallah I, Klauser P, Conus P, Do KQ. Caught in vicious circles: a perspective on dynamic feed-forward loops driving oxidative stress in schizophrenia. Mol Psychiatry 2022; 27:1886-1897. [PMID: 34759358 PMCID: PMC9126811 DOI: 10.1038/s41380-021-01374-w] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/18/2021] [Accepted: 10/20/2021] [Indexed: 12/18/2022]
Abstract
A growing body of evidence has emerged demonstrating a pathological link between oxidative stress and schizophrenia. This evidence identifies oxidative stress as a convergence point or "central hub" for schizophrenia genetic and environmental risk factors. Here we review the existing experimental and translational research pinpointing the complex dynamics of oxidative stress mechanisms and their modulation in relation to schizophrenia pathophysiology. We focus on evidence supporting the crucial role of either redox dysregulation, N-methyl-D-aspartate receptor hypofunction, neuroinflammation or mitochondria bioenergetics dysfunction, initiating "vicious circles" centered on oxidative stress during neurodevelopment. These processes would amplify one another in positive feed-forward loops, leading to persistent impairments of the maturation and function of local parvalbumin-GABAergic neurons microcircuits and myelinated fibers of long-range macrocircuitry. This is at the basis of neural circuit synchronization impairments and cognitive, emotional, social and sensory deficits characteristic of schizophrenia. Potential therapeutic approaches that aim at breaking these different vicious circles represent promising strategies for timely and safe interventions. In order to improve early detection and increase the signal-to-noise ratio for adjunctive trials of antioxidant, anti-inflammatory and NMDAR modulator drugs, a reverse translation of validated circuitry approach is needed. The above presented processes allow to identify mechanism based biomarkers guiding stratification of homogenous patients groups and target engagement required for successful clinical trials, paving the way towards precision medicine in psychiatry.
Collapse
Affiliation(s)
- Michel Cuenod
- 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
| | - Jan-Harry Cabungcal
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Prilly, Lausanne, Switzerland
| | - Daniella Dwir
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Prilly, Lausanne, Switzerland
| | - Ines Khadimallah
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Prilly, Lausanne, Switzerland
| | - Paul Klauser
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Prilly, Lausanne, Switzerland
- Service of Child and Adolescent Psychiatry, Department of Psychiatry, Lausanne University Hospital, Prilly, Lausanne, Switzerland
| | - Philippe Conus
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital, Prilly, Lausanne, Switzerland
| | - Kim Q Do
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Prilly, Lausanne, Switzerland.
| |
Collapse
|
10
|
Veselinović T, Neuner I. Progress and Pitfalls in Developing Agents to Treat Neurocognitive Deficits Associated with Schizophrenia. CNS Drugs 2022; 36:819-858. [PMID: 35831706 PMCID: PMC9345797 DOI: 10.1007/s40263-022-00935-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/06/2022] [Indexed: 12/11/2022]
Abstract
Cognitive impairments associated with schizophrenia (CIAS) represent a central element of the symptomatology of this severe mental disorder. CIAS substantially determine the disease prognosis and hardly, if at all, respond to treatment with currently available antipsychotics. Remarkably, all drugs presently approved for the treatment of schizophrenia are, to varying degrees, dopamine D2/D3 receptor blockers. In turn, rapidly growing evidence suggests the immense significance of systems other than the dopaminergic system in the genesis of CIAS. Accordingly, current efforts addressing the unmet needs of patients with schizophrenia are primarily based on interventions in other non-dopaminergic systems. In this review article, we provide a brief overview of the available evidence on the importance of specific systems in the development of CIAS. In addition, we describe the promising targets for the development of new drugs that have been used so far. In doing so, we present the most important candidates that have been investigated in the field of the specific systems in recent years and present a summary of the results available at the time of drafting this review (May 2022), as well as the currently ongoing studies.
Collapse
Affiliation(s)
- Tanja Veselinović
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich, Jülich, Germany.
| | - Irene Neuner
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich, Jülich, Germany
- JARA-BRAIN, Aachen, Germany
| |
Collapse
|
11
|
Guler EM, Kurtulmus A, Gul AZ, Kocyigit A, Kirpinar I. Oxidative stress and schizophrenia: A comparative cross-sectional study of multiple oxidative markers in patients and their first-degree relatives. Int J Clin Pract 2021; 75:e14711. [PMID: 34370389 DOI: 10.1111/ijcp.14711] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 08/06/2021] [Indexed: 01/03/2023] Open
Abstract
OBJECTIVE Schizophrenia (SCZ) is a chronic, disruptive mental disorder with unknown pathogenic mechanisms. Several studies evidenced that oxidative stress (OS) may be one of the causal factors to play a role in the pathophysiology of the disease. Our study aims to contribute to the SCZ research by investigating a possible relationship between the severity of illness (scored with "The Positive and Negative Syndrome Scale [PANSS]") and OS biomarkers in patients. We additionally assess the "first-degree-relatives (FDRs)" oxidative status with multiple parameters to test the idea of oxidative imbalance leads to disease progression as a genetical susceptibility factor. METHODS This study included: 50 adult patients with SCZ, 50 unaffected FDRs, and 50 controls. OS biomarkers included myeloperoxidase (MPO), total oxidant status (TOS), total antioxidant status (TAS), total thiol (TT), native thiol (NT). Photometric methods were used to measure the parameters in the peripheral blood samples of participants. Disulphide (DS) and oxidative stress index (OSI) parameters were calculated. RESULTS TOS, DS, OSI levels were significantly higher, and TAS, TT, NT levels were significantly lower in both SCZ and FDRs than controls. In the SCZ group, MPO activity was significantly higher compared with other groups. Results in this study did not provide a strong correlation between the PANSS and selected biomarkers. There was a slightly negative correlation between TT and PANSS in the SCZ group (P = .041, r = -.297). CONCLUSION OS biomarkers increased significantly in the peripheral blood of SCZ patients compared with other groups indicates the presence of OS in the aetiology of the disease. Mid-levels of oxidative markers found in FDRs imply that unaffected first-degree relatives have an increased risk for turning up to the clinical presentation stage.
Collapse
Affiliation(s)
- Eray Metin Guler
- Department of Medical Biochemistry, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
- Department of Medical Biochemistry, Hamidiye School of Medicine, University of Health Sciences Turkey, Istanbul, Turkey
- Department of Medical Biochemistry, Haydarpasa Numune Health Application and Research Center, University of Health Sciences Turkey, Istanbul, Turkey
| | - Ayse Kurtulmus
- Department of Psychiatry, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
- Department of Psychiatry, Istanbul Medeniyet University Goztepe Education Research Hospital, Istanbul, Turkey
| | - Ayse Zehra Gul
- Department of Medical Biochemistry, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
| | - Abdurrahim Kocyigit
- Department of Medical Biochemistry, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
| | - Ismet Kirpinar
- Department of Psychiatry, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
| |
Collapse
|
12
|
Metzner C, Steuber V. The beta component of gamma-band auditory steady-state responses in patients with schizophrenia. Sci Rep 2021; 11:20387. [PMID: 34650135 PMCID: PMC8516862 DOI: 10.1038/s41598-021-99793-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 09/24/2021] [Indexed: 01/18/2023] Open
Abstract
The mechanisms underlying circuit dysfunctions in schizophrenia (SCZ) remain poorly understood. Auditory steady-state responses (ASSRs), especially in the gamma and beta band, have been suggested as a potential biomarker for SCZ. While the reduction of 40 Hz power for 40 Hz drive has been well established and replicated in SCZ patients, studies are inconclusive when it comes to an increase in 20 Hz power during 40 Hz drive. There might be several factors explaining the inconsistencies, including differences in the sensitivity of the recording modality (EEG vs MEG), differences in stimuli (click-trains vs amplitude-modulated tones) and large differences in the amplitude of the stimuli. Here, we used a computational model of ASSR deficits in SCZ and explored the effect of three SCZ-associated microcircuit alterations: reduced GABA activity, increased GABA decay times and NMDA receptor hypofunction. We investigated the effect of input strength on gamma (40 Hz) and beta (20 Hz) band power during gamma ASSR stimulation and saw that the pronounced increase in beta power during gamma stimulation seen experimentally could only be reproduced in the model when GABA decay times were increased and only for a specific range of input strengths. More specifically, when the input was in this specific range, the rhythmic drive at 40 Hz produced a strong 40 Hz rhythm in the control network; however, in the 'SCZ-like' network, the prolonged inhibition led to a so-called 'beat-skipping', where the network would only strongly respond to every other input. This mechanism was responsible for the emergence of the pronounced 20 Hz beta peak in the power spectrum. The other two microcircuit alterations were not able to produce a substantial 20 Hz component but they further narrowed the input strength range for which the network produced a beta component when combined with increased GABAergic decay times. Our finding that the beta component only existed for a specific range of input strengths might explain the seemingly inconsistent reporting in experimental studies and suggests that future ASSR studies should systematically explore different amplitudes of their stimuli. Furthermore, we provide a mechanistic link between a microcircuit alteration and an electrophysiological marker in schizophrenia and argue that more complex ASSR stimuli are needed to disentangle the nonlinear interactions of microcircuit alterations. The computational modelling approach put forward here is ideally suited to facilitate the development of such stimuli in a theory-based fashion.
Collapse
Affiliation(s)
- Christoph Metzner
- Neural Information Processing Group, Institute of Software Engineering and Theoretical Computer Science, Technische Universität Berlin, Berlin, Germany.
- School of Physics, Engineering and Computer Science, University of Hertfordshire, Hatfield, UK.
| | - Volker Steuber
- School of Physics, Engineering and Computer Science, University of Hertfordshire, Hatfield, UK
| |
Collapse
|
13
|
Chiang TI, Yu YH, Lin CH, Lane HY. Novel Biomarkers of Alzheimer's Disease: Based Upon N-methyl-D-aspartate Receptor Hypoactivation and Oxidative Stress. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2021; 19:423-433. [PMID: 34294612 PMCID: PMC8316669 DOI: 10.9758/cpn.2021.19.3.423] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/07/2020] [Accepted: 12/14/2020] [Indexed: 12/29/2022]
Abstract
Early detection and prevention of Alzheimer’s disease (AD) is important. The current treatment for early AD is acetylcholine esterase inhibitors (AChEIs); however, the efficacy is poor. Besides, AChEI did not show efficacy in mild cognitive impairment (MCI). Beta-amyloid (Aβ) deposits have been regarded to be highly related to the pathogenesis of AD. However, many clinical trials aiming at the clearance of Aβ deposits failed to improve the cognitive decline of AD, even at its early phase. There should be other important mechanisms unproven in the course of AD and MCI. Feasible biomarkers for the diagnosis and treatment response of AD are lacking to date. The N-methyl-D-aspartate receptor (NMDAR) activation plays an important role in learning and memory. On the other hand, oxidative stress has been regarded to contribute to aging with the assumption that free radicals damage cell constituents and connective tissues. Our recent study found that an NMDAR enhancer, sodium benzoate (the pivotal inhibitor of D-amino acid oxidase [DAAO]), improved the cognitive and global function of patients with early-phase AD. Further, we found that peripheral DAAO levels were higher in patients with MCI and AD than healthy controls. We also found that sodium benzoate was able to change the activity of antioxidant. These pieces of evidence suggest that the NMDAR function is associated with anti-oxidation, and have potential to be biomarkers for the diagnosis and treatment response of AD.
Collapse
Affiliation(s)
- Ting-I Chiang
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yi-Hsiang Yu
- Department of Dermatology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - 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 Science, Asia University, Taichung, Taiwan
| |
Collapse
|
14
|
Morgan AM, Hassanen EI, Ogaly HA, Al Dulmani SA, Al-Zahrani FAM, Galal MK, Kamel S, Rashad MM, Ibrahim MA, Hussien AM. The ameliorative effect of N-acetylcysteine against penconazole induced neurodegenerative and neuroinflammatory disorders in rats. J Biochem Mol Toxicol 2021; 35:e22884. [PMID: 34392569 DOI: 10.1002/jbt.22884] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/24/2021] [Accepted: 08/06/2021] [Indexed: 11/07/2022]
Abstract
Penconazole (PEN) is a widely used systemic fungicide to treat various fungal diseases in plants but it leaves residues in crops and food products causing serious environmental and health problems. N-acetylcysteine (NAC) is a precursor of the antioxidant glutathione in the body and exerts prominent antioxidant and anti-inflammatory effects. The present study aimed to explore the mechanistic way of NAC to ameliorate the PEN neurotoxicity in male rats. Twenty-eight male rats were randomly divided into four groups (n = 7) and given the treated material via oral gavage for 10 days as the following: Group I (distilled water), Group II (50 mg/kg body weight [bwt] PEN), Group III (200 mg/kg bwt NAC), and Group IV (NAC + PEN). After 10 days all rats were subjected to behavioral assessment and then euthanized to collect brain tissues to perform oxidative stress, molecular studies, and pathological examination. Our results revealed that PEN exhibits neurobehavioral toxicity manifested by alteration in the forced swim test, elevated plus maze test, and Y-maze test. There were marked elevations in malondialdehyde levels with reduction in total antioxidant capacity levels, upregulation of messenger RNA levels of bax, caspase 3, and caspase 9 genes with downregulation of bcl2 genes. In addition, brain sections showed marked histopathological alteration in the cerebrum and cerebellum with strong bax and inducible nitric oxide synthetase protein expression. On the contrary, cotreatment of rats with NAC had the ability to improve all the abovementioned neurotoxic parameters. The present study can conclude that NAC has a neuroprotective effect against PEN-induced neurotoxicity via its antioxidant, anti-inflammatory, and antiapoptotic effect. We recommend using NAC as a preventive and therapeutic agent for a wide variety of neurodegenerative and neuroinflammatory disorders.
Collapse
Affiliation(s)
- Ashraf M Morgan
- Toxicology and Forensic Medicine Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Eman I Hassanen
- Pathology Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Hanan A Ogaly
- Chemistry Department, Faculty of Science, King Khalid University, Abha, Saudi Arabia
| | - Sharah A Al Dulmani
- Chemistry Department, Faculty of Science, King Khalid University, Abha, Saudi Arabia
| | | | - Mona K Galal
- Biochemistry and Molecular Biology Department, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| | - Shaimaa Kamel
- Biochemistry and Molecular Biology Department, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| | - Maha M Rashad
- Biochemistry and Molecular Biology Department, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| | - Marwa A Ibrahim
- Biochemistry and Molecular Biology Department, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| | - Ahmed M Hussien
- Toxicology and Forensic Medicine Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
15
|
Kwon N, Lim CS, Ko G, Ha J, Lee D, Yin J, Kim HM, Yoon J. Fluorescence Probe for Imaging N-Methyl-d-aspartate Receptors and Monitoring GSH Selectively Using Two-Photon Microscopy. Anal Chem 2021; 93:11612-11616. [PMID: 34382767 DOI: 10.1021/acs.analchem.1c02350] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
N-Methyl-d-aspartate (NMDA) is an excitotoxic amino acid used to identify a specific subset of glutamate receptors. The activity of NMDA receptors is closely related to the redox level of the biological system. Glutathione (GSH) as an antioxidant plays a key role with regard to modulation of the redox environment. In this work we designed and developed a GSH-specific fluorescent probe with the capability of targeting NMDA receptors, which was composed of a two-photon naphthalimide fluorophore, a GSH-reactive group sulfonamide, and an ifenprodil targeting group for the NMDA receptor. This probe exhibited high selectivity toward GSH in comparison to other similar amino acids. Two-photon fluorescence microscopy allowed this probe to successfully monitor GSH in neuronal cells and hippocampal tissues with an excitation at 750 nm. It could serve as a potential practical imaging tool to explore the function of GSH and related biological processes in the brain.
Collapse
Affiliation(s)
- Nahyun Kwon
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul 03760, Korea
| | - Chang Su Lim
- Department of Energy Systems Research, Ajou University, Suwon 443-749, Korea
| | - Gyeongju Ko
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul 03760, Korea
| | - Jeongsun Ha
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul 03760, Korea
| | - Dayoung Lee
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul 03760, Korea
| | - Jun Yin
- Key Laboratory of Pesticide and Chemical Biology, Ministry of Education; Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis; International Joint Research Center for Intelligent Biosensing Technology and Health; College of Chemistry, Central China Normal University, Wuhan 430079, People's Republic of China
| | - Hwan Myung Kim
- Department of Energy Systems Research, Ajou University, Suwon 443-749, Korea
| | - Juyoung Yoon
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
16
|
Pearson SA, Cowan JA. Glutathione-coordinated metal complexes as substrates for cellular transporters. Metallomics 2021; 13:mfab015. [PMID: 33770183 PMCID: PMC8086996 DOI: 10.1093/mtomcs/mfab015] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 03/15/2021] [Indexed: 11/15/2022]
Abstract
Glutathione is the major thiol-containing species in both prokaryotes and eukaryotes and plays a wide variety of roles, including detoxification of metals by sequestration, reduction, and efflux. ABC transporters such as MRP1 and MRP2 detoxify the cell from certain metals by exporting the cations as a metal-glutathione complex. The ability of the bacterial Atm1 protein to efflux metal-glutathione complexes appears to have evolved over time to become the ABCB7 transporter in mammals, located in the inner mitochondrial membrane. No longer needed for the role of cellular detoxification, ABCB7 appears to be used to transport glutathione-coordinated iron-sulfur clusters from mitochondria to the cytosol.
Collapse
Affiliation(s)
- Stephen A Pearson
- The Ohio State University Biophysics Program, The Ohio State University, 484 West 12th Avenue, Columbus, OH 43210, USA
| | - J A Cowan
- The Ohio State University Biophysics Program, The Ohio State University, 484 West 12th Avenue, Columbus, OH 43210, USA
- Department of Chemistry and Biochemistry, The Ohio State University, 100 West 18th Avenue, Columbus, OH 43210, USA
| |
Collapse
|
17
|
Wu Q, Huang J, Wu R. Drugs Based on NMDAR Hypofunction Hypothesis in Schizophrenia. Front Neurosci 2021; 15:641047. [PMID: 33912003 PMCID: PMC8072017 DOI: 10.3389/fnins.2021.641047] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 03/12/2021] [Indexed: 12/30/2022] Open
Abstract
Treatments for negative symptoms and cognitive dysfunction in schizophrenia remain issues that psychiatrists around the world are trying to solve. Their mechanisms may be associated with N-methyl-D-aspartate receptors (NMDARs). The NMDAR hypofunction hypothesis for schizophrenia was brought to the fore mainly based on the clinical effects of NMDAR antagonists and anti-NMDAR encephalitis pathology. Drugs targeted at augmenting NMDAR function in the brain seem to be promising in improving negative symptoms and cognitive dysfunction in patients with schizophrenia. In this review, we list NMDAR-targeted drugs and report on related clinical studies. We then summarize their effects on negative symptoms and cognitive dysfunction and analyze the unsatisfactory outcomes of these clinical studies according to the improved glutamate hypothesis that has been revealed in animal models. We aimed to provide perspectives for scientists who sought therapeutic strategies for negative symptoms and cognitive dysfunction in schizophrenia based on the NMDAR hypofunction hypothesis.
Collapse
Affiliation(s)
- Qiongqiong Wu
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jing Huang
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Renrong Wu
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
18
|
Bjørklund G, Doşa MD, Maes M, Dadar M, Frye RE, Peana M, Chirumbolo S. The impact of glutathione metabolism in autism spectrum disorder. Pharmacol Res 2021; 166:105437. [PMID: 33493659 DOI: 10.1016/j.phrs.2021.105437] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 12/31/2020] [Accepted: 01/12/2021] [Indexed: 12/14/2022]
Abstract
This paper reviews the potential role of glutathione (GSH) in autism spectrum disorder (ASD). GSH plays a key role in the detoxification of xenobiotics and maintenance of balance in intracellular redox pathways. Recent data showed that imbalances in the GSH redox system are an important factor in the pathophysiology of ASD. Furthermore, ASD is accompanied by decreased concentrations of reduced GSH in part caused by oxidation of GSH into glutathione disulfide (GSSG). GSSG can react with protein sulfhydryl (SH) groups, thereby causing proteotoxic stress and other abnormalities in SH-containing enzymes in the brain and blood. Moreover, alterations in the GSH metabolism via its effects on redox-independent mechanisms are other processes associated with the pathophysiology of ASD. GSH-related regulation of glutamate receptors such as the N-methyl-D-aspartate receptor can contribute to glutamate excitotoxicity. Synergistic and antagonistic interactions between glutamate and GSH can result in neuronal dysfunction. These interactions can involve transcription factors of the immune pathway, such as activator protein 1 and nuclear factor (NF)-κB, thereby interacting with neuroinflammatory mechanisms, ultimately leading to neuronal damage. Neuronal apoptosis and mitochondrial dysfunction are recently outlined as significant factors linking GSH impairments with the pathophysiology of ASD. Moreover, GSH regulates the methylation of DNA and modulates epigenetics. Existing data support a protective role of the GSH system in ASD development. Future research should focus on the effects of GSH redox signaling in ASD and should explore new therapeutic approaches by targeting the GSH system.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine (CONEM), Toften 24, 8610, Mo i Rana, Norway.
| | - Monica Daniela Doşa
- Department of Pharmacology, Faculty of Medicine, Ovidius University of Constanta, Campus, 900470, Constanta, Romania.
| | - Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Impact Research Center, Deakin University, Geelong, Australia
| | - Maryam Dadar
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Richard E Frye
- Barrow Neurological Institute, Phoenix Children's Hospital, Phoenix, AZ, USA; Department of Child Health, University of Arizona College of Medicine, Phoenix, AZ, USA
| | | | - Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy; CONEM Scientific Secretary, Verona, Italy
| |
Collapse
|
19
|
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
|
20
|
Raffa M, Bel Hadj Youssef I, Ben Othman L, Fendri C, Mechri A. [Plasmatic glutathione levels and their relationships with clinical and therapeutic features in patients with schizophrenia]. Encephale 2020; 47:10-14. [PMID: 33358006 DOI: 10.1016/j.encep.2020.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 01/27/2020] [Accepted: 01/30/2020] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Altered glutathione systems (GSH) are suggested to participate in the pathophysiology of schizophrenia. The purpose of this study was to determine the plasmatic glutathione levels of patients with schizophrenia compared to healthy controls and to examine their relationships with clinical and therapeutic features. METHODS It was a case-control study carried out on 100 patients with schizophrenia according to DSM-IV-TR criteria and 95 healthy controls. All patients were assessed by Clinical Global Impressions-severity (CGI-severity) and Global Assessment of Functioning (EGF). Most of the patients (55%) were under first-generation antipsychotics. Plasmatic glutathione levels (total glutathione GSHt, reduced glutathione GSHr, oxidized glutathione GSSG) were determined by spectrophotometry. RESULTS The levels of GSHt and GSHr were significantly decreased in schizophrenic patients in comparison with the healthy controls. These reductions were noted to be more pronounced in the untreated patients. No correlation was observed between the GSH levels and the clinical subtypes of schizophrenia and EGF scores. Depending on the therapeutic status, there were no significant differences in the GSH levels. In addition, there was no correlation between the GSH levels and the daily dosage of the antipsychotic treatment. CONCLUSION Our results suggest that the observed changes are related to the physiopathology of schizophrenia rather than to the presence of neuroleptic treatment. These results provide support for further studies of the possible role of antioxidants as neuroprotective therapeutic strategies.
Collapse
Affiliation(s)
- M Raffa
- Laboratoire de recherche « Éléments trace, radicaux libres, antioxydants et pathologies humaines et environnement », département de biophysique, faculté de médecine de Monastir, rue Ibn Sina, 5000 Monastir, Tunisie.
| | - I Bel Hadj Youssef
- Laboratoire de recherche « Vulnérabilité aux psychoses », service de psychiatrie, hôpital universitaire de Monastir, rue 1(er) Juin, 5000 Monastir, Tunisie
| | - L Ben Othman
- Laboratoire de recherche « Éléments trace, radicaux libres, antioxydants et pathologies humaines et environnement », département de biophysique, faculté de médecine de Monastir, rue Ibn Sina, 5000 Monastir, Tunisie
| | - C Fendri
- Laboratoire de recherche « Vulnérabilité aux psychoses », service de psychiatrie, hôpital universitaire de Monastir, rue 1(er) Juin, 5000 Monastir, Tunisie
| | - A Mechri
- Laboratoire de recherche « Vulnérabilité aux psychoses », service de psychiatrie, hôpital universitaire de Monastir, rue 1(er) Juin, 5000 Monastir, Tunisie
| |
Collapse
|
21
|
Veschsanit N, Yang JL, Ngampramuan S, Viwatpinyo K, Pinyomahakul J, Lwin T, Chancharoen P, Rungruang S, Govitrapong P, Mukda S. Melatonin reverts methamphetamine-induced learning and memory impairments and hippocampal alterations in mice. Life Sci 2020; 265:118844. [PMID: 33278389 DOI: 10.1016/j.lfs.2020.118844] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 11/26/2020] [Accepted: 11/27/2020] [Indexed: 02/07/2023]
Abstract
AIMS Methamphetamine (METH) has become a major public health problem because of its abuse and profound neurotoxic effects, causing alterations in brain structure and function, and impairing cognitive functions, including attention, decision making, emotional memory, and working memory. This study aimed to determine whether melatonin (MEL), the circadian-control hormone, which has roles beyond circadian rhythm regulation, could restore METH-induced cognitive and neuronal impairment. MAIN METHODS Mice were treated with either METH (1 mg/kg) or saline for 7 days, followed by MEL (10 mg/kg) or saline for another 14 days. The Morris water maze (MWM) test was performed one day after the last saline or MEL injection. The hippocampal neuronal density, synaptic density, and receptors involved in learning and memory, along with downstream signaling molecules (NMDA receptor subunits GluN2A, GluN2B, and CaMKII) were investigated by immunoblotting. KEY FINDINGS METH administration significantly extended escape latency in learning phase and reduced the number of target crossings in memory test-phase as well as decreased the expression of BDNF, NMDA receptors, TrkB receptors, CaMKII, βIII tubulin, and synaptophysin. MEL treatment significantly ameliorated METH-induced increased escape latency, decreased the number of target crossings and decreased expression of BDNF, NMDA receptors, TrkB receptors, CaMKII, βIII tubulin and synaptophysin. SIGNIFICANCE METH administration impairs learning and memory in mice, and MEL administration restores METH-induced neuronal impairments which is probably through the changes in BDNF, NMDA receptors, TrkB receptors, CaMKII, βIII tubulin and synaptophysin. Therefore, MEL is potentially an innovative and promising treatment for learning and memory impairment of humans.
Collapse
Affiliation(s)
- Nisarath Veschsanit
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakornpathom 73170, Thailand
| | - Jenq-Lin Yang
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Sukonthar Ngampramuan
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakornpathom 73170, Thailand
| | - Kittikun Viwatpinyo
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakornpathom 73170, Thailand
| | - Jitrapa Pinyomahakul
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakornpathom 73170, Thailand
| | - Thit Lwin
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakornpathom 73170, Thailand; Department of Anatomy, Defence Services Medical Academy, Mingalardon, Yangon 11021, Myanmar
| | - Pongrung Chancharoen
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakornpathom 73170, Thailand; Faculty of Allied Health Sciences, Burapha University, Seansuk, Chonburi 20131, Thailand
| | - Saowalak Rungruang
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Piyarat Govitrapong
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakornpathom 73170, Thailand; Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Bangkok 10210, Thailand
| | - Sujira Mukda
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakornpathom 73170, Thailand.
| |
Collapse
|
22
|
Bjørklund G, Tinkov AA, Hosnedlová B, Kizek R, Ajsuvakova OP, Chirumbolo S, Skalnaya MG, Peana M, Dadar M, El-Ansary A, Qasem H, Adams JB, Aaseth J, Skalny AV. The role of glutathione redox imbalance in autism spectrum disorder: A review. Free Radic Biol Med 2020; 160:149-162. [PMID: 32745763 DOI: 10.1016/j.freeradbiomed.2020.07.017] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 07/02/2020] [Accepted: 07/13/2020] [Indexed: 12/22/2022]
Abstract
The role of glutathione in autism spectrum disorder (ASD) is emerging as a major topic, due to its role in the maintenance of the intracellular redox balance. Several studies have implicated glutathione redox imbalance as a leading factor in ASD, and both ASD and many other neurodevelopmental disorders involve low levels of reduced glutathione (GSH), high levels of oxidized glutathione (GSSG), and abnormalities in the expressions of glutathione-related enzymes in the blood or brain. Glutathione metabolism, through its impact on redox environment or redox-independent mechanisms, interferes with multiple mechanisms involved in ASD pathogenesis. Glutathione-mediated regulation of glutamate receptors [e.g., N-methyl-d-aspartate (NMDA) receptor], as well as the role of glutamate as a substrate for glutathione synthesis, may be involved in the regulation of glutamate excitotoxicity. However, the interaction between glutathione and glutamate in the pathogenesis of brain diseases may vary from synergism to antagonism. Modulation of glutathione is also associated with regulation of redox-sensitive transcription factors nuclear factor kappa B (NF-κB) and activator protein 1 (AP-1) and downstream signaling (proinflammatory cytokines and inducible enzymes), thus providing a significant impact on neuroinflammation. Mitochondrial dysfunction, as well as neuronal apoptosis, may also provide a significant link between glutathione metabolism and ASD. Furthermore, it has been recently highlighted that glutathione can affect and modulate DNA methylation and epigenetics. Review analysis including research studies meeting the required criteria for analysis showed statistically significant differences between the plasma GSH and GSSG levels as well as GSH:GSSG ratio in autistic patients compared with healthy individuals (P = 0.0145, P = 0.0150 and P = 0.0202, respectively). Therefore, the existing data provide a strong background on the role of the glutathione system in ASD pathogenesis. Future research is necessary to investigate the role of glutathione redox signaling in ASD, which could potentially also lead to promising therapeutics.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine (CONEM), Mo I Rana, Norway.
| | - Alexey A Tinkov
- IM Sechenov First Moscow State Medical University, Moscow, Russia; Yaroslavl State University, Yaroslavl, Russia; Federal Research Centre of Biological Systems, Agro-technologies of the Russian Academy of Sciences, Orenburg, Russia
| | - Božena Hosnedlová
- Department of Human Pharmacology and Toxicology, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences Brno, Brno, Czech Republic; CONEM Metallomics Nanomedicine Research Group (CMNRG), Brno, Czech Republic
| | - Rene Kizek
- Department of Human Pharmacology and Toxicology, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences Brno, Brno, Czech Republic; CONEM Metallomics Nanomedicine Research Group (CMNRG), Brno, Czech Republic; Faculty of Pharmacy with Division of Laboratory Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Olga P Ajsuvakova
- IM Sechenov First Moscow State Medical University, Moscow, Russia; Yaroslavl State University, Yaroslavl, Russia; Federal Research Centre of Biological Systems, Agro-technologies of the Russian Academy of Sciences, Orenburg, Russia
| | - Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy; CONEM Scientific Secretary, Verona, Italy
| | - Margarita G Skalnaya
- IM Sechenov First Moscow State Medical University, Moscow, Russia; Federal Research Centre of Biological Systems, Agro-technologies of the Russian Academy of Sciences, Orenburg, Russia
| | | | - Maryam Dadar
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Afaf El-Ansary
- Medicinal Chemistry Department, King Saud University, Riyadh, Saudi Arabia; Autism Research and Treatment Center, Riyadh, Saudi Arabia; CONEM Saudi Autism Research Group, King Saud University, Riyadh, Saudi Arabia
| | - Hanan Qasem
- Autism Research and Treatment Center, Riyadh, Saudi Arabia; CONEM Saudi Autism Research Group, King Saud University, Riyadh, Saudi Arabia
| | - James B Adams
- School for Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ, USA
| | - Jan Aaseth
- Research Department, Innlandet Hospital Trust, Brumunddal, Norway
| | - Anatoly V Skalny
- IM Sechenov First Moscow State Medical University, Moscow, Russia; Federal Research Centre of Biological Systems, Agro-technologies of the Russian Academy of Sciences, Orenburg, Russia
| |
Collapse
|
23
|
Bjørklund G, Peana M, Maes M, Dadar M, Severin B. The glutathione system in Parkinson's disease and its progression. Neurosci Biobehav Rev 2020; 120:470-478. [PMID: 33068556 DOI: 10.1016/j.neubiorev.2020.10.004] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 09/25/2020] [Accepted: 10/05/2020] [Indexed: 02/07/2023]
Abstract
Redox dysfunctions and neuro-oxidative stress play a major role in the pathophysiology and progression of Parkinson's disease (PD). Glutathione (GSH) and the reduced/oxidized glutathione (GSH/GSSG) ratio are lowered in oxidative stress conditions and may lead to increased oxidative toxicity. GSH is involved not only in neuro-immune and neuro-oxidative processes, including thiol redox signaling, but also in cell proliferation and differentiation and in the regulation of cell death, including apoptotic pathways. Lowered GSH metabolism and a low GSH/GSSG ratio following oxidative stress are associated with mitochondrial dysfunctions and constitute a critical factor in the neuroinflammatory and neurodegenerative processes accompanying PD. This review provides indirect evidence that GSH redox signaling is associated with the pathophysiology of PD. Nevertheless, it has not been delineated whether GSH redox imbalances are a causative factor in PD or whether PD-associated pathways cause the GSH redox imbalances in PD. The results show that antioxidant approaches, including neuroprotective and anti-neuroinflammatory agents, which neutralize reactive oxygen species, may have therapeutic efficacy in the treatment of PD and its progression.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine (CONEM), Mo i Rana, Norway.
| | | | - Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Impact Research Center, Deakin University, Geelong, Australia
| | - Maryam Dadar
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Beatrice Severin
- Faculty of Medicine, Ovidius University of Constanta, Constanta, Romania
| |
Collapse
|
24
|
Egerton A, Grace AA, Stone J, Bossong MG, Sand M, McGuire P. Glutamate in schizophrenia: Neurodevelopmental perspectives and drug development. Schizophr Res 2020; 223:59-70. [PMID: 33071070 DOI: 10.1016/j.schres.2020.09.013] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 08/12/2020] [Accepted: 09/20/2020] [Indexed: 12/14/2022]
Abstract
Research into the neurobiological processes that may lead to the onset of schizophrenia places growing emphasis on the glutamatergic system and brain development. Preclinical studies have shown that neurodevelopmental, genetic, and environmental factors contribute to glutamatergic dysfunction and schizophrenia-related phenotypes. Clinical research has suggested that altered brain glutamate levels may be present before the onset of psychosis and relate to outcome in those at clinical high risk. After psychosis onset, glutamate dysfunction may also relate to the degree of antipsychotic response and clinical outcome. These findings support ongoing efforts to develop pharmacological interventions that target the glutamate system and could suggest that glutamatergic compounds may be more effective in specific patient subgroups or illness stages. In this review, we consider the updated glutamate hypothesis of schizophrenia, from a neurodevelopmental perspective, by reviewing recent preclinical and clinical evidence, and discuss the potential implications for novel therapeutics.
Collapse
Affiliation(s)
- Alice Egerton
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| | - Anthony A Grace
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA, USA
| | - James Stone
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Matthijs G Bossong
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Michael Sand
- Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, USA
| | - Philip McGuire
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
25
|
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: 57] [Impact Index Per Article: 14.3] [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
|
26
|
Di Pietro V, Yakoub KM, Caruso G, Lazzarino G, Signoretti S, Barbey AK, Tavazzi B, Lazzarino G, Belli A, Amorini AM. Antioxidant Therapies in Traumatic Brain Injury. Antioxidants (Basel) 2020; 9:antiox9030260. [PMID: 32235799 PMCID: PMC7139349 DOI: 10.3390/antiox9030260] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/14/2020] [Accepted: 03/20/2020] [Indexed: 02/08/2023] Open
Abstract
Due to a multiplicity of causes provoking traumatic brain injury (TBI), TBI is a highly heterogeneous pathology, characterized by high mortality and disability rates. TBI is an acute neurodegenerative event, potentially and unpredictably evolving into sub-chronic and chronic neurodegenerative events, with transient or permanent neurologic, cognitive, and motor deficits, for which no valid standardized therapies are available. A vast body of literature demonstrates that TBI-induced oxidative/nitrosative stress is involved in the development of both acute and chronic neurodegenerative disorders. Cellular defenses against this phenomenon are largely dependent on low molecular weight antioxidants, most of which are consumed with diet or as nutraceutical supplements. A large number of studies have evaluated the efficacy of antioxidant administration to decrease TBI-associated damage in various animal TBI models and in a limited number of clinical trials. Points of weakness of preclinical studies are represented by the large variability in the TBI model adopted, in the antioxidant tested, in the timing, dosages, and routes of administration used, and in the variety of molecular and/or neurocognitive parameters evaluated. The analysis of the very few clinical studies does not allow strong conclusions to be drawn on the real effectiveness of antioxidant administration to TBI patients. Standardizing TBI models and different experimental conditions, as well as testing the efficacy of administration of a cocktail of antioxidants rather than only one, should be mandatory. According to some promising clinical results, it appears that sports-related concussion is probably the best type of TBI to test the benefits of antioxidant administration.
Collapse
Affiliation(s)
- Valentina Di Pietro
- Neurotrauma and Ophthalmology Research Group, Institute of Inflammation and Aging, University of Birmingham, Birmingham B15 2TT, UK; (V.D.P.); (K.M.Y.)
- NIHR Surgical Reconstruction and Microbiology Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2TT, UK
- The Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana Champaign, Champaign, IL 61801, USA;
| | - Kamal M. Yakoub
- Neurotrauma and Ophthalmology Research Group, Institute of Inflammation and Aging, University of Birmingham, Birmingham B15 2TT, UK; (V.D.P.); (K.M.Y.)
- NIHR Surgical Reconstruction and Microbiology Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2TT, UK
| | - Giuseppe Caruso
- Department of Laboratories, Oasi Research Institute – IRCCS, Via Conte Ruggero 73, 94018 Troina (EN), Italy;
| | - Giacomo Lazzarino
- UniCamillus, Saint Camillus International University of Health Sciences, Via di Sant’Alessandro 8, 00131 Rome, Italy;
| | - Stefano Signoretti
- UOC Neurochirurgia, ASL Roma2, S. Eugenio Hospital, Piazzale dell’Umanesimo 10, 00144 Rome, Italy;
| | - Aron K. Barbey
- The Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana Champaign, Champaign, IL 61801, USA;
| | - Barbara Tavazzi
- Institute of Biochemistry and Clinical Biochemistry, Catholic University of Rome, Largo F.Vito 1, 00168 Rome, Italy
- Department of Scienze di laboratorio e infettivologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
- Correspondence: (B.T.); (G.L.); (A.B.)
| | - Giuseppe Lazzarino
- Department of Biomedical and Biotechnological Sciences, Division of Medical Biochemistry, University of Catania, Via S.Sofia 97, 95123 Catania, Italy;
- Correspondence: (B.T.); (G.L.); (A.B.)
| | - Antonio Belli
- Neurotrauma and Ophthalmology Research Group, Institute of Inflammation and Aging, University of Birmingham, Birmingham B15 2TT, UK; (V.D.P.); (K.M.Y.)
- NIHR Surgical Reconstruction and Microbiology Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2TT, UK
- Correspondence: (B.T.); (G.L.); (A.B.)
| | - Angela Maria Amorini
- Department of Biomedical and Biotechnological Sciences, Division of Medical Biochemistry, University of Catania, Via S.Sofia 97, 95123 Catania, Italy;
| |
Collapse
|
27
|
Kumar J, Liddle EB, Fernandes CC, Palaniyappan L, Hall EL, Robson SE, Simmonite M, Fiesal J, Katshu MZ, Qureshi A, Skelton M, Christodoulou NG, Brookes MJ, Morris PG, Liddle PF. Glutathione and glutamate in schizophrenia: a 7T MRS study. Mol Psychiatry 2020; 25:873-882. [PMID: 29934548 PMCID: PMC7156342 DOI: 10.1038/s41380-018-0104-7] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Revised: 05/04/2018] [Accepted: 05/14/2018] [Indexed: 12/21/2022]
Abstract
In schizophrenia, abnormal neural metabolite concentrations may arise from cortical damage following neuroinflammatory processes implicated in acute episodes. Inflammation is associated with increased glutamate, whereas the antioxidant glutathione may protect against inflammation-induced oxidative stress. We hypothesized that patients with stable schizophrenia would exhibit a reduction in glutathione, glutamate, and/or glutamine in the cerebral cortex, consistent with a post-inflammatory response, and that this reduction would be most marked in patients with "residual schizophrenia", in whom an early stage with positive psychotic symptoms has progressed to a late stage characterized by long-term negative symptoms and impairments. We recruited 28 patients with stable schizophrenia and 45 healthy participants matched for age, gender, and parental socio-economic status. We measured glutathione, glutamate and glutamine concentrations in the anterior cingulate cortex (ACC), left insula, and visual cortex using 7T proton magnetic resonance spectroscopy (MRS). Glutathione and glutamate were significantly correlated in all three voxels. Glutamine concentrations across the three voxels were significantly correlated with each other. Principal components analysis (PCA) produced three clear components: an ACC glutathione-glutamate component; an insula-visual glutathione-glutamate component; and a glutamine component. Patients with stable schizophrenia had significantly lower scores on the ACC glutathione-glutamate component, an effect almost entirely leveraged by the sub-group of patients with residual schizophrenia. All three metabolite concentration values in the ACC were significantly reduced in this group. These findings are consistent with the hypothesis that excitotoxicity during the acute phase of illness leads to reduced glutathione and glutamate in the residual phase of the illness.
Collapse
Affiliation(s)
- Jyothika Kumar
- 0000 0004 1936 8868grid.4563.4Division of Psychiatry and Applied Psychology, University of Nottingham, Nottingham, UK
| | - Elizabeth B. Liddle
- 0000 0004 1936 8868grid.4563.4Division of Psychiatry and Applied Psychology, University of Nottingham, Nottingham, UK
| | - Carolina C. Fernandes
- 0000 0004 1936 8868grid.4563.4Sir Peter Mansfield Imaging Centre, University of Nottingham, Nottingham, UK
| | - Lena Palaniyappan
- 0000 0004 1936 8884grid.39381.30Departments of Psychiatry, Medical Biophysics and Neuroscience, Western University, London, ON Canada ,Lawson Research, Brain and Mind & Robarts Research Institutes, London, ON Canada
| | - Emma L. Hall
- 0000 0004 1936 8868grid.4563.4Sir Peter Mansfield Imaging Centre, University of Nottingham, Nottingham, UK
| | - Siân E. Robson
- 0000 0000 8610 2323grid.482042.8Healthcare Improvement Scotland, Gyle Square, Edinburgh, UK
| | - Molly Simmonite
- 0000000086837370grid.214458.eDepartment of Psychology, University of Michigan, Ann Arbor, MI USA
| | - Jan Fiesal
- grid.500956.fSouth Staffordshire and Shropshire Healthcare NHS Foundation Trust, Stafford, UK
| | - Mohammad Z. Katshu
- 0000 0004 1936 8868grid.4563.4Division of Psychiatry and Applied Psychology, University of Nottingham, Nottingham, UK ,0000 0001 1514 761Xgrid.439378.2Nottinghamshire Healthcare NHS Foundation Trust, Nottingham, UK
| | - Ayaz Qureshi
- 0000 0004 0430 6955grid.450837.dGreater Manchester West Mental Health NHS Foundation Trust, Manchester, UK
| | - Michael Skelton
- 0000 0004 0396 1667grid.418388.eDerbyshire Healthcare NHS Foundation Trust, Derby, UK
| | - Nikolaos G. Christodoulou
- 0000 0004 1936 8868grid.4563.4Division of Psychiatry and Applied Psychology, University of Nottingham, Nottingham, UK ,0000 0001 1514 761Xgrid.439378.2Nottinghamshire Healthcare NHS Foundation Trust, Nottingham, UK
| | - Matthew J. Brookes
- 0000 0004 1936 8868grid.4563.4Sir Peter Mansfield Imaging Centre, University of Nottingham, Nottingham, UK
| | - Peter G. Morris
- 0000 0004 1936 8868grid.4563.4Sir Peter Mansfield Imaging Centre, University of Nottingham, Nottingham, UK
| | - Peter F. Liddle
- 0000 0004 1936 8868grid.4563.4Division of Psychiatry and Applied Psychology, University of Nottingham, Nottingham, UK
| |
Collapse
|
28
|
The Role of Parvalbumin-positive Interneurons in Auditory Steady-State Response Deficits in Schizophrenia. Sci Rep 2019; 9:18525. [PMID: 31811155 PMCID: PMC6898379 DOI: 10.1038/s41598-019-53682-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 10/12/2019] [Indexed: 12/19/2022] Open
Abstract
Despite an increasing body of evidence demonstrating subcellular alterations in parvalbumin-positive (PV+) interneurons in schizophrenia, their functional consequences remain elusive. Since PV+ interneurons are involved in the generation of fast cortical rhythms, these changes have been hypothesized to contribute to well-established alterations of beta and gamma range oscillations in patients suffering from schizophrenia. However, the precise role of these alterations and the role of different subtypes of PV+ interneurons is still unclear. Here we used a computational model of auditory steady-state response (ASSR) deficits in schizophrenia. We investigated the differential effects of decelerated synaptic dynamics, caused by subcellular alterations at two subtypes of PV+ interneurons: basket cells and chandelier cells. Our simulations suggest that subcellular alterations at basket cell synapses rather than chandelier cell synapses are the main contributor to these deficits. Particularly, basket cells might serve as target for innovative therapeutic interventions aiming at reversing the oscillatory deficits.
Collapse
|
29
|
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: 128] [Impact Index Per Article: 25.6] [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
|
30
|
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: 8.6] [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
|
31
|
Kim Y, Vadodaria KC, Lenkei Z, Kato T, Gage FH, Marchetto MC, Santos R. Mitochondria, Metabolism, and Redox Mechanisms in Psychiatric Disorders. Antioxid Redox Signal 2019; 31:275-317. [PMID: 30585734 PMCID: PMC6602118 DOI: 10.1089/ars.2018.7606] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 12/21/2018] [Accepted: 12/23/2018] [Indexed: 12/17/2022]
Abstract
Significance: Our current knowledge of the pathophysiology and molecular mechanisms causing psychiatric disorders is modest, but genetic susceptibility and environmental factors are central to the etiology of these conditions. Autism, schizophrenia, bipolar disorder and major depressive disorder show genetic gene risk overlap and share symptoms and metabolic comorbidities. The identification of such common features may provide insights into the development of these disorders. Recent Advances: Multiple pieces of evidence suggest that brain energy metabolism, mitochondrial functions and redox balance are impaired to various degrees in psychiatric disorders. Since mitochondrial metabolism and redox signaling can integrate genetic and environmental environmental factors affecting the brain, it is possible that they are implicated in the etiology and progression of psychiatric disorders. Critical Issue: Evidence for direct links between cellular mitochondrial dysfunction and disease features are missing. Future Directions: A better understanding of the mitochondrial biology and its intracellular connections to the nuclear genome, the endoplasmic reticulum and signaling pathways, as well as its role in intercellular communication in the organism, is still needed. This review focuses on the findings that implicate mitochondrial dysfunction, the resultant metabolic changes and oxidative stress as important etiological factors in the context of psychiatric disorders. We also propose a model where specific pathophysiologies of psychiatric disorders depend on circuit-specific impairments of mitochondrial dysfunction and redox signaling at specific developmental stages.
Collapse
Affiliation(s)
- Yeni Kim
- Department of Child and Adolescent Psychiatry, National Center for Mental Health, Seoul, South Korea
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, California
| | - Krishna C. Vadodaria
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, California
| | - Zsolt Lenkei
- Laboratory of Dynamic of Neuronal Structure in Health and Disease, Institute of Psychiatry and Neuroscience of Paris (UMR_S1266 INSERM, University Paris Descartes), Paris, France
| | - Tadafumi Kato
- Laboratory for Molecular Dynamics of Mental Disorders, RIKEN Center for Brain Science, Wako, Japan
| | - Fred H. Gage
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, California
| | - Maria C. Marchetto
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, California
| | - Renata Santos
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, California
- Laboratory of Dynamic of Neuronal Structure in Health and Disease, Institute of Psychiatry and Neuroscience of Paris (UMR_S1266 INSERM, University Paris Descartes), Paris, France
| |
Collapse
|
32
|
de la Salle S, Shah D, Choueiry J, Bowers H, McIntosh J, Ilivitsky V, Knott V. NMDA Receptor Antagonist Effects on Speech-Related Mismatch Negativity and Its Underlying Oscillatory and Source Activity in Healthy Humans. Front Pharmacol 2019; 10:455. [PMID: 31139075 PMCID: PMC6517681 DOI: 10.3389/fphar.2019.00455] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 04/11/2019] [Indexed: 11/18/2022] Open
Abstract
Background: Previous studies in schizophrenia have consistently shown that deficits in the generation of the auditory mismatch negativity (MMN) – a pre-attentive, event-related potential (ERP) typically elicited by changes to simple sound features – are linked to N-methyl-D-aspartate (NMDA) receptor hypofunction. Concomitant with extensive language dysfunction in schizophrenia, patients also exhibit MMN deficits to changes in speech but their relationship to NMDA-mediated neurotransmission is not clear. Accordingly, our study aimed to investigate speech MMNs in healthy humans and their underlying electrophysiological mechanisms in response to NMDA antagonist treatment. We also evaluated the relationship between baseline MMN/electrocortical activity and emergent schizophrenia-like symptoms associated with NMDA receptor blockade. Methods: In a sample of 18 healthy volunteers, a multi-feature Finnish language paradigm incorporating changes in syllables, vowels and consonant stimuli was used to assess the acute effects of the NMDA receptor antagonist ketamine and placebo on the MMN. Further, measures of underlying neural activity, including evoked theta power, theta phase locking and source-localized current density in cortical regions of interest were assessed. Subjective symptoms were assessed with the Clinician Administered Dissociative States Scale (CADSS). Results: Participants exhibited significant ketamine-induced increases in psychosis-like symptoms and depending on temporal or frontal recording region, co-occurred with reductions in MMN generation in response to syllable frequency/intensity, vowel duration, across vowel and consonant deviants. MMN attenuation was associated with decreases in evoked theta power, theta phase locking and diminished current density in auditory and inferior frontal (language-related cortical) regions. Baseline (placebo) MMN and underlying electrophysiological features associated with the processing of changes in syllable intensity correlated with the degree of psychotomimetic response to ketamine. Conclusion: Ketamine-induced impairments in healthy human speech MMNs and their underlying electrocortical mechanisms closely resemble those observed in schizophrenia and support a model of dysfunctional NMDA receptor-mediated neurotransmission of language processing deficits in schizophrenia.
Collapse
Affiliation(s)
| | - Dhrasti Shah
- School of Psychology, University of Ottawa, Ottawa, ON, Canada
| | - Joelle Choueiry
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Hayley Bowers
- Department of Psychology, University of Guelph, Guelph, ON, Canada
| | - Judy McIntosh
- The Royal's Institute of Mental Health Research, Ottawa, ON, Canada
| | | | - Verner Knott
- School of Psychology, University of Ottawa, Ottawa, ON, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.,The Royal's Institute of Mental Health Research, Ottawa, ON, Canada.,Royal Ottawa Mental Health Centre, Ottawa, ON, Canada
| |
Collapse
|
33
|
Girgis RR, Baker S, Mao X, Gil R, Javitt DC, Kantrowitz JT, Gu M, Spielman DM, Ojeil N, Xu X, Abi-Dargham A, Shungu DC, Kegeles LS. Effects of acute N-acetylcysteine challenge on cortical glutathione and glutamate in schizophrenia: A pilot in vivo proton magnetic resonance spectroscopy study. Psychiatry Res 2019; 275:78-85. [PMID: 30884334 PMCID: PMC6515541 DOI: 10.1016/j.psychres.2019.03.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 03/11/2019] [Accepted: 03/11/2019] [Indexed: 12/11/2022]
Abstract
Findings from in vivo brain proton magnetic resonance spectroscopy (1H MRS) and preclinical studies have suggested region- and medication status-dependent increases in glutamate (Glu) levels and deficiencies in glutathione (GSH) levels in schizophrenia. N-acetylcysteine (NAC), a GSH synthesis precursor, has demonstrated modest clinical benefit in schizophrenia. The objective of this study was to examine the effects of acute administration of NAC on GSH and Glu levels measured with 1H MRS in 19 patients with schizophrenia and 20 healthy control subjects. Levels of GSH were acquired in dorsal anterior cingulate cortex (dACC), and those of Glu in dACC and medial prefrontal cortex (mPFC), at baseline and 60 min following acute oral administration of 2400 mg of NAC. No differences in the levels of GSH or Glu were found at baseline or following NAC administration between patients with schizophrenia and control subjects in either of the targeted brain regions. Future studies measuring GSH levels in brain regions previously found to exhibit glutamatergic abnormalities or using genetic polymorphisms, while controlling for the age and medication status of the cohorts, are warranted to better identify groups of patients more likely to respond to NAC and its mode of action and mechanisms.
Collapse
Affiliation(s)
- Ragy R Girgis
- Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York State Psychiatric Institute, 1051 Riverside Drive, Unit 31, New York, NY 10032, USA.
| | - Seth Baker
- Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York State Psychiatric Institute, 1051 Riverside Drive, Unit 31, New York, NY 10032, USA
| | - Xiangling Mao
- Department of Radiology, Weill Cornell Medical College, New York, USA
| | - Roberto Gil
- Department of Psychiatry, Stony Brook University School of Medicine, Stony Brook, USA
| | - Daniel C Javitt
- Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York State Psychiatric Institute, 1051 Riverside Drive, Unit 31, New York, NY 10032, USA
| | - Joshua T Kantrowitz
- Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York State Psychiatric Institute, 1051 Riverside Drive, Unit 31, New York, NY 10032, USA
| | - Meng Gu
- Department of Radiology, Stanford University, Stanford, CA, USA
| | | | - Najate Ojeil
- Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York State Psychiatric Institute, 1051 Riverside Drive, Unit 31, New York, NY 10032, USA
| | - Xiaoyan Xu
- Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York State Psychiatric Institute, 1051 Riverside Drive, Unit 31, New York, NY 10032, USA
| | - Anissa Abi-Dargham
- Department of Psychiatry, Stony Brook University School of Medicine, Stony Brook, USA
| | - Dikoma C Shungu
- Department of Radiology, Weill Cornell Medical College, New York, USA
| | - Lawrence S Kegeles
- Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York State Psychiatric Institute, 1051 Riverside Drive, Unit 31, New York, NY 10032, USA
| |
Collapse
|
34
|
Morris G, Berk M, Maes M, Puri BK. Could Alzheimer's Disease Originate in the Periphery and If So How So? Mol Neurobiol 2019; 56:406-434. [PMID: 29705945 PMCID: PMC6372984 DOI: 10.1007/s12035-018-1092-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 04/17/2018] [Indexed: 12/11/2022]
Abstract
The classical amyloid cascade model for Alzheimer's disease (AD) has been challenged by several findings. Here, an alternative molecular neurobiological model is proposed. It is shown that the presence of the APOE ε4 allele, altered miRNA expression and epigenetic dysregulation in the promoter region and exon 1 of TREM2, as well as ANK1 hypermethylation and altered levels of histone post-translational methylation leading to increased transcription of TNFA, could variously explain increased levels of peripheral and central inflammation found in AD. In particular, as a result of increased activity of triggering receptor expressed on myeloid cells 2 (TREM-2), the presence of the apolipoprotein E4 (ApoE4) isoform, and changes in ANK1 expression, with subsequent changes in miR-486 leading to altered levels of protein kinase B (Akt), mechanistic (previously mammalian) target of rapamycin (mTOR) and signal transducer and activator of transcription 3 (STAT3), all of which play major roles in microglial activation, proliferation and survival, there is activation of microglia, leading to the subsequent (further) production of cytokines, chemokines, nitric oxide, prostaglandins, reactive oxygen species, inducible nitric oxide synthase and cyclooxygenase-2, and other mediators of inflammation and neurotoxicity. These changes are associated with the development of amyloid and tau pathology, mitochondrial dysfunction (including impaired activity of the electron transport chain, depleted basal mitochondrial potential and oxidative damage to key tricarboxylic acid enzymes), synaptic dysfunction, altered glycogen synthase kinase-3 (GSK-3) activity, mTOR activation, impairment of autophagy, compromised ubiquitin-proteasome system, iron dyshomeostasis, changes in APP translation, amyloid plaque formation, tau hyperphosphorylation and neurofibrillary tangle formation.
Collapse
Affiliation(s)
- Gerwyn Morris
- IMPACT Strategic Research Centre, School of Medicine, Barwon Health, Deakin University, P.O. Box 291, Geelong, Victoria, Australia
| | - Michael Berk
- IMPACT Strategic Research Centre, School of Medicine, Barwon Health, Deakin University, P.O. Box 291, Geelong, Victoria, Australia
- Department of Psychiatry, Level 1 North, Main Block, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia
- Florey Institute for Neuroscience and Mental Health, Kenneth Myer Building, University of Melbourne, 30 Royal Parade, Parkville, Victoria, Australia
- Orygen, The National Centre of Excellence in Youth Mental Health, 35 Poplar Rd, Parkville, Victoria, Australia
| | - Michael Maes
- IMPACT Strategic Research Centre, School of Medicine, Barwon Health, Deakin University, P.O. Box 291, Geelong, Victoria, Australia
- Department of Psychiatry, Chulalongkorn University, Bangkok, Thailand
| | - Basant K Puri
- Department of Medicine, Hammersmith Hospital, Imperial College London, London, UK.
| |
Collapse
|
35
|
Tardiolo G, Bramanti P, Mazzon E. Overview on the Effects of N-Acetylcysteine in Neurodegenerative Diseases. Molecules 2018; 23:molecules23123305. [PMID: 30551603 PMCID: PMC6320789 DOI: 10.3390/molecules23123305] [Citation(s) in RCA: 148] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 12/07/2018] [Accepted: 12/10/2018] [Indexed: 02/06/2023] Open
Abstract
N-acetylcysteine (NAC), which is an acetylated cysteine compound, has aroused scientific interest for decades due to its important medical applications. It also represents a nutritional supplement in the human diet. NAC is a glutathione precursor and shows antioxidant and anti-inflammatory activities. In addition to the uses quoted in the literature, NAC may be considered helpful in therapies to counteract neurodegenerative and mental health diseases. Furthermore, this compound has been evaluated for its neuroprotective potential in the prevention of cognitive aging dementia. NAC is inexpensive, commercially available and no relevant side effects were observed after its administration. The purpose of this paper is to give an overview on the effects and applications of NAC in Parkinson's and Alzheimer's disorders and in neuropathic pain and stroke.
Collapse
Affiliation(s)
- Giuseppe Tardiolo
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy.
| | - Placido Bramanti
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy.
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy.
| |
Collapse
|
36
|
Effect of N-acetyl cysteine (NAC) supplementation on positive and negative syndrome scale in schizophrenia: a systematic review and meta-analysis of randomised controlled trials. Eur J Clin Pharmacol 2018; 75:289-301. [PMID: 30446769 DOI: 10.1007/s00228-018-2595-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Accepted: 10/31/2018] [Indexed: 10/27/2022]
Abstract
OBJECTIVE To conduct a systematic review and meta-analysis of published randomised controlled trials on the efficacy of NAC supplementation on positive and negative syndrome scale in schizophrenia. METHODS A meta-analysis was conducted, and studies were identified by a search of electronic databases from inception to May 2018. Combined and stratified analyses were used. RESULTS Seven trials were identified, and data from n = 447 participants were included. Pooled analysis showed improvement of positive and negative syndrome scale following NAC treatment compared with placebo, for total (SMB = - 0.96) [95% CI - 1.69, - 0.24; P = 0.009], general (SMB = - 1.04) [95% CI - 1.80, - 0.27; P = 0.008] and negative (SMB = - 0.73) [95% CI - 1.29, - 0.17; P = 0.01] scores, respectively. Significant heterogeneity was found, and subgroup analysis showed significant reductions in studies with a treatment duration of ≤ 24 weeks, with a considerable effect size on total, general, and negative scores (Total SMD = - 0.83; General SMD = - 0.67; Negative SMD = - 1.09) following NAC. CONCLUSIONS NAC improved all aspects of positive and negative syndrome scale in schizophrenic populations and may be more efficacious with treatment durations up to 24 weeks.
Collapse
|
37
|
NMDA-receptor inhibition and oxidative stress during hippocampal maturation differentially alter parvalbumin expression and gamma-band activity. Sci Rep 2018; 8:9545. [PMID: 29934499 PMCID: PMC6015018 DOI: 10.1038/s41598-018-27830-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 06/11/2018] [Indexed: 02/06/2023] Open
Abstract
Dysfunction of parvalbumin (PV)-expressing interneurons is thought to underlie the alterations of gamma-band oscillations observed in schizophrenia. Although the pathomechanisms of this disease remain unclear, oxidative stress induced by NMDA receptor (NMDAR) hypofunction and decreased glutathione (GSH) synthesizing capacity have been shown to lead to PV-loss and aberrant oscillatory activity. However, the individual contributions of NMDAR-inhibition and GSH-depletion to the developmental alterations observed in schizophrenia are largely unknown. We therefore investigated each condition in isolation using hippocampal slice cultures wherein interneuron maturation occurs entirely in vitro. Although both treatments caused oxidative stress, NMDAR-inhibition led to an immediate reduction in gamma oscillation frequency and a delayed loss of PV. In contrast, GSH-depletion immediately decreased PV expression and increased power, without affecting frequency. Hence, although disturbances of PV-expression and gamma oscillations coexist in schizophrenia, they can arise from separate pathological processes.
Collapse
|
38
|
Benvenutti R, Marcon M, Reis CG, Nery LR, Miguel C, Herrmann AP, Vianna MRM, Piato A. N-acetylcysteine protects against motor, optomotor and morphological deficits induced by 6-OHDA in zebrafish larvae. PeerJ 2018; 6:e4957. [PMID: 29868300 PMCID: PMC5985760 DOI: 10.7717/peerj.4957] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 05/22/2018] [Indexed: 12/13/2022] Open
Abstract
Background Parkinson's disease (PD) is the second most common neurodegenerative disorder. In addition to its highly debilitating motor symptoms, non-motor symptoms may precede their motor counterparts by many years, which may characterize a prodromal phase of PD. A potential pharmacological strategy is to introduce neuroprotective agents at an earlier stage in order to prevent further neuronal death. N-acetylcysteine (NAC) has been used against paracetamol overdose hepatotoxicity by restoring hepatic concentrations of glutathione (GSH), and as a mucolytic in chronic obstructive pulmonary disease by reducing disulfide bonds in mucoproteins. It has been shown to be safe for humans at high doses. More recently, several studies have evidenced that NAC has a multifaceted mechanism of action, presenting indirect antioxidant effect by acting as a GSH precursor, besides its anti-inflammatory and neurotrophic effects. Moreover, NAC modulates glutamate release through activation of the cystine-glutamate antiporter in extra-synaptic astrocytes. Its therapeutic benefits have been demonstrated in clinical trials for several neuropsychiatric conditions but has not been tested in PD models yet. Methods In this study, we evaluated the potential of NAC to prevent the damage induced by 6-hydroxydopamine (6-OHDA) on motor, optomotor and morphological parameters in a PD model in larval zebrafish. Results NAC was able to prevent the motor deficits (total distance, mean speed, maximum acceleration, absolute turn angle and immobility time), optomotor response impairment and morphological alterations (total length and head length) caused by exposure to 6-OHDA, which reinforce and broaden the relevance of its neuroprotective effects. Discussion NAC acts in different targets relevant to PD pathophysiology. Further studies and clinical trials are needed to assess this agent as a candidate for prevention and adjunctive treatment of PD.
Collapse
Affiliation(s)
- Radharani Benvenutti
- Programa de Pós-graduação em Neurociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Matheus Marcon
- Programa de Pós-graduação em Neurociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Carlos G Reis
- Programa de Pós-graduação em Neurociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Laura R Nery
- Programa de Pós-graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Camila Miguel
- Programa de Pós-graduação em Zoologia, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Ana P Herrmann
- Programa de Pós-graduação em Farmacologia e Terapêutica, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Monica R M Vianna
- Programa de Pós-graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.,Programa de Pós-graduação em Zoologia, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Angelo Piato
- Programa de Pós-graduação em Neurociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.,Programa de Pós-graduação em Farmacologia e Terapêutica, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
39
|
Sepehrmanesh Z, Heidary M, Akasheh N, Akbari H, Heidary M. Therapeutic effect of adjunctive N-acetyl cysteine (NAC) on symptoms of chronic schizophrenia: A double-blind, randomized clinical trial. Prog Neuropsychopharmacol Biol Psychiatry 2018; 82:289-296. [PMID: 29126981 DOI: 10.1016/j.pnpbp.2017.11.001] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Revised: 11/03/2017] [Accepted: 11/06/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND Schizophrenia is one of the most disabling psychiatric syndromes with the prevalence of 1% in the general population. Despite availability of various antipsychotics, negative symptoms and cognitive impairment are difficult to treat. In addition antipsychotic monotherapy is not effective in most of these patients. Current evidence indicates the roles of glutamatergic system in this disorder. N-acetyl cysteine (NAC) also increases extracellular glutamate. This study was conducted to evaluate the clinical effects of oral NAC as an add-on to maintenance medication for the treatment of chronic schizophrenia. MATERIALS AND METHODS This 12-week, double-blind, randomized, placebo-controlled, clinical trial was performed to determine the effectiveness of 1200mg N-acetyl cysteine as an adjunctive treatment with conventional antipsychotic medications in 84 patients with chronic schizophrenia. The subjects were evaluated with the Positive and Negative Syndrome Scale (PANSS), Mini-Mental State Examination (MMSE), and a standard neuropsychological screening test. Data were analyzed with SPSS-16 software. RESULTS NAC-treated patients showed significantly improvement in the positive (F=5.47, P=0.02) and negative (F=0.20, df=1) PANSS subscale. Also the general and total PANSS score of NAC group declined over times whilst it was increased for placebo group. Regarding cognitive functions, improvement was observed in some explored areas, such as attention, short-term and working memory, executive functioning and speed of processing. There was no significant difference between the 2 groups in the frequency of adverse effects. CONCLUSION The present study detected improvement in positive, negative, general and total psychopathology symptoms as well as cognitive performance with NAC treatment. It is also well-tolerated, safe and easy-to-use agent as an effective therapeutic strategy to improve outcome in schizophrenia treatment.
Collapse
Affiliation(s)
- Zahra Sepehrmanesh
- Department of psychiatry, School of Medicine, Kashan University of Medical Science, Kashan, Iran
| | - Mahsa Heidary
- Department of psychiatry, School of Medicine, Kashan University of Medical Science, Kashan, Iran.
| | - Negar Akasheh
- School of Medicine, Qazvin University of Medical Science, Qazvin, Iran
| | - Hossein Akbari
- Department of Public Health, Kashan University Of Medical Sciences, kashan, Iran
| | - Mahshid Heidary
- Department of Clinical Psychology, Qom Islamic Azad University, Qom, Iran.
| |
Collapse
|
40
|
Maclean KN, Jiang H, Aivazidis S, Kim E, Shearn CT, Harris PS, Petersen DR, Allen RH, Stabler SP, Roede JR. Taurine treatment prevents derangement of the hepatic γ-glutamyl cycle and methylglyoxal metabolism in a mouse model of classical homocystinuria: regulatory crosstalk between thiol and sulfinic acid metabolism. FASEB J 2018; 32:1265-1280. [PMID: 29101223 DOI: 10.1096/fj.201700586r] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Cystathionine β-synthase-deficient homocystinuria (HCU) is a poorly understood, life-threatening inborn error of sulfur metabolism. Analysis of hepatic glutathione (GSH) metabolism in a mouse model of HCU demonstrated significant depletion of cysteine, GSH, and GSH disulfide independent of the block in trans-sulfuration compared with wild-type controls. HCU induced the expression of the catalytic and regulatory subunits of γ-glutamyl ligase, GSH synthase (GS), γ-glutamyl transpeptidase 1, 5-oxoprolinase (OPLAH), and the GSH-dependent methylglyoxal detoxification enzyme, glyoxalase-1. Multiple components of the transcription factor nuclear factor (erythroid-derived 2)-like 2 (Nrf2)-mediated antioxidant-response regulatory axis were induced without any detectable activation of Nrf2. Metabolomic analysis revealed the accumulation of multiple γ-glutamyl amino acids and that plasma ophthalmate levels could serve as a noninvasive marker for hepatic redox stress. Neither cysteine, nor betaine treatment was able to reverse the observed enzyme inductions. Taurine treatment normalized the expression levels of γ-glutamyl ligase C/M, GS, OPLAH, and glyoxalase-1, and reversed HCU-induced deficits in protein glutathionylation by acting to double GSH levels relative to controls. Collectively, our data indicate that the perturbation of the γ-glutamyl cycle could contribute to multiple sequelae in HCU and that taurine has significant therapeutic potential for both HCU and other diseases for which GSH depletion is a critical pathogenic factor.-Maclean, K. N., Jiang, H., Aivazidis, S., Kim, E., Shearn, C. T., Harris, P. S., Petersen, D. R., Allen, R. H., Stabler, S. P., Roede, J. R. Taurine treatment prevents derangement of the hepatic γ-glutamyl cycle and methylglyoxal metabolism in a mouse model of classical homocystinuria: regulatory crosstalk between thiol and sulfinic acid metabolism.
Collapse
Affiliation(s)
- Kenneth N Maclean
- Department of Pediatrics, University of Colorado Health Sciences Center, Aurora, Colorado, USA
| | - Hua Jiang
- Department of Pediatrics, University of Colorado Health Sciences Center, Aurora, Colorado, USA
| | - Stefanos Aivazidis
- Department of Pharmaceutical Sciences, University of Colorado Health Sciences Center, Aurora, Colorado, USA
| | - Eugene Kim
- Department of Pediatrics, University of Colorado Health Sciences Center, Aurora, Colorado, USA
| | - Colin T Shearn
- Department of Pharmaceutical Sciences, University of Colorado Health Sciences Center, Aurora, Colorado, USA
| | - Peter S Harris
- Department of Pharmaceutical Sciences, University of Colorado Health Sciences Center, Aurora, Colorado, USA
| | - Dennis R Petersen
- Department of Pharmaceutical Sciences, University of Colorado Health Sciences Center, Aurora, Colorado, USA
| | - Robert H Allen
- Department of Medicine, University of Colorado Health Sciences Center, Aurora, Colorado, USA
| | - Sally P Stabler
- Department of Medicine, University of Colorado Health Sciences Center, Aurora, Colorado, USA
| | - James R Roede
- Department of Pharmaceutical Sciences, University of Colorado Health Sciences Center, Aurora, Colorado, USA
| |
Collapse
|
41
|
Retsa C, Knebel JF, Geiser E, Ferrari C, Jenni R, Fournier M, Alameda L, Baumann PS, Clarke S, Conus P, Do KQ, Murray MM. Treatment in early psychosis with N-acetyl-cysteine for 6months improves low-level auditory processing: Pilot study. Schizophr Res 2018; 191:80-86. [PMID: 28711476 DOI: 10.1016/j.schres.2017.07.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 06/27/2017] [Accepted: 07/03/2017] [Indexed: 02/03/2023]
Abstract
Sensory impairments constitute core dysfunctions in schizophrenia. In the auditory modality, impaired mismatch negativity (MMN) has been observed in chronic schizophrenia and may reflect N-methyl-d-aspartate (NMDA) hypo-function, consistent with models of schizophrenia based on oxidative stress. Moreover, a recent study demonstrated deficits in the N100 component of the auditory evoked potential (AEP) in early psychosis patients. Previous work has shown that add-on administration of the glutathione precursor N-acetyl-cysteine (NAC) improves the MMN and clinical symptoms in chronic schizophrenia. To date, it remains unknown whether NAC also improves general low-level auditory processing and if its efficacy would extend to early-phase psychosis. We addressed these issues with a randomized, double-blind study of a small sample (N=15) of early psychosis (EP) patients and 18 healthy controls from whom AEPs were recorded during an active, auditory oddball task. Patients were recorded twice: once prior to NAC/placebo administration and once after six months of treatment. The N100 component was significantly smaller in patients before NAC administration versus controls. Critically, NAC administration improved this AEP deficit. Source estimations revealed increased activity in the left temporo-parietal lobe in patients after NAC administration. Overall, the data from this pilot study, which call for replication in a larger sample, indicate that NAC improves low-level auditory processing in early psychosis.
Collapse
Affiliation(s)
- Chrysa Retsa
- The LINE (Laboratory for Investigative Neurophysiology), Radiodiagnostic Service, University Hospital Center and University of Lausanne, 1011 Lausanne, Switzerland; Neuropsychology and Neurorehabilitation Service and Radiodiagnostic Service, University Hospital Center and University of Lausanne, 1011 Lausanne, Switzerland
| | - Jean-François Knebel
- The LINE (Laboratory for Investigative Neurophysiology), Radiodiagnostic Service, University Hospital Center and University of Lausanne, 1011 Lausanne, Switzerland; Neuropsychology and Neurorehabilitation Service and Radiodiagnostic Service, University Hospital Center and University of Lausanne, 1011 Lausanne, Switzerland; The EEG Brain Mapping Core, Center for Biomedical Imaging (CIBM), University Hospital Center and University of Lausanne, 1011 Lausanne, Switzerland
| | - Eveline Geiser
- The LINE (Laboratory for Investigative Neurophysiology), Radiodiagnostic Service, University Hospital Center and University of Lausanne, 1011 Lausanne, Switzerland; Neuropsychology and Neurorehabilitation Service and Radiodiagnostic Service, University Hospital Center and University of Lausanne, 1011 Lausanne, Switzerland
| | - Carina Ferrari
- Center for Psychiatric Neuroscience, Department of Psychiatry, University Hospital Center and University of Lausanne, Prilly-Lausanne, Switzerland; Service of General Psychiatry, Department of Psychiatry, University Hospital Center and University of Lausanne, Prilly-Lausanne, Switzerland
| | - Raoul Jenni
- Center for Psychiatric Neuroscience, Department of Psychiatry, University Hospital Center and University of Lausanne, Prilly-Lausanne, Switzerland; Service of General Psychiatry, Department of Psychiatry, University Hospital Center and University of Lausanne, Prilly-Lausanne, Switzerland
| | - Margot Fournier
- Center for Psychiatric Neuroscience, Department of Psychiatry, University Hospital Center and University of Lausanne, Prilly-Lausanne, Switzerland
| | - Luis Alameda
- Center for Psychiatric Neuroscience, Department of Psychiatry, University Hospital Center and University of Lausanne, Prilly-Lausanne, Switzerland; Service of General Psychiatry, Department of Psychiatry, University Hospital Center and University of Lausanne, Prilly-Lausanne, Switzerland; Psychiatric Liaison Service, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Philipp S Baumann
- Center for Psychiatric Neuroscience, Department of Psychiatry, University Hospital Center and University of Lausanne, Prilly-Lausanne, Switzerland; Service of General Psychiatry, Department of Psychiatry, University Hospital Center and University of Lausanne, Prilly-Lausanne, Switzerland
| | - Stephanie Clarke
- The LINE (Laboratory for Investigative Neurophysiology), Radiodiagnostic Service, University Hospital Center and University of Lausanne, 1011 Lausanne, Switzerland; Neuropsychology and Neurorehabilitation Service and Radiodiagnostic Service, University Hospital Center and University of Lausanne, 1011 Lausanne, Switzerland
| | - Philippe Conus
- Service of General Psychiatry, Department of Psychiatry, University Hospital Center and University of Lausanne, Prilly-Lausanne, Switzerland
| | - Kim Q Do
- Center for Psychiatric Neuroscience, Department of Psychiatry, University Hospital Center and University of Lausanne, Prilly-Lausanne, Switzerland
| | - Micah M Murray
- The LINE (Laboratory for Investigative Neurophysiology), Radiodiagnostic Service, University Hospital Center and University of Lausanne, 1011 Lausanne, Switzerland; Neuropsychology and Neurorehabilitation Service and Radiodiagnostic Service, University Hospital Center and University of Lausanne, 1011 Lausanne, Switzerland; The EEG Brain Mapping Core, Center for Biomedical Imaging (CIBM), University Hospital Center and University of Lausanne, 1011 Lausanne, Switzerland; Department of Ophthalmology, University of Lausanne, Fondation Asile des Aveugles, Lausanne, Switzerland; Department of Hearing and Speech Sciences, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
42
|
Geiser E, Retsa C, Knebel JF, Ferrari C, Jenni R, Fournier M, Alameda L, Baumann PS, Clarke S, Conus P, Do KQ, Murray MM. The coupling of low-level auditory dysfunction and oxidative stress in psychosis patients. Schizophr Res 2017; 190:52-59. [PMID: 28189532 DOI: 10.1016/j.schres.2017.02.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 01/30/2017] [Accepted: 02/01/2017] [Indexed: 02/08/2023]
Abstract
Patients diagnosed with schizophrenia often present with low-level sensory deficits. It is an open question whether there is a functional link between these deficits and the pathophysiology of the disease, e.g. oxidative stress and glutathione (GSH) metabolism dysregulation. Auditory evoked potentials (AEPs) were recorded from 21 psychosis disorder patients and 30 healthy controls performing an active, auditory oddball task. AEPs to standard sounds were analyzed within an electrical neuroimaging framework. A peripheral measure of participants' redox balance, the ratio of glutathione peroxidase and glutathione reductase activities (GPx/GR), was correlated with the AEP data. Patients displayed significantly decreased AEPs over the time window of the P50/N100 complex resulting from significantly weaker responses in the left temporo-parietal lobe. The GPx/GR ratio significantly correlated with patients' brain activity during the time window of the P50/N100 in the medial frontal lobe. We show for the first time a direct coupling between electrophysiological indices of AEPs and peripheral redox dysregulation in psychosis patients. This coupling is limited to stages of auditory processing that are impaired relative to healthy controls and suggests a link between biochemical and sensory dysfunction. The data highlight the potential of low-level sensory processing as a trait-marker of psychosis.
Collapse
Affiliation(s)
- Eveline Geiser
- Neuropsychology and Neurorehabilitation Service, University Hospital Center and University of Lausanne, 1011 Lausanne, Switzerland; Radiodiagnostic Service, University Hospital Center and University of Lausanne, 1011 Lausanne, Switzerland
| | - Chrysa Retsa
- Neuropsychology and Neurorehabilitation Service, University Hospital Center and University of Lausanne, 1011 Lausanne, Switzerland
| | - Jean-François Knebel
- Neuropsychology and Neurorehabilitation Service, University Hospital Center and University of Lausanne, 1011 Lausanne, Switzerland; Radiodiagnostic Service, University Hospital Center and University of Lausanne, 1011 Lausanne, Switzerland; The EEG Brain Mapping Core, Center for Biomedical Imaging (CIBM), University Hospital Center and University of Lausanne, 1011 Lausanne, Switzerland
| | - Carina Ferrari
- Center for Psychiatric Neuroscience, Department of Psychiatry, University Hospital Center and University of Lausanne, Prilly-Lausanne, Switzerland; Service of General Psychiatry, Department of Psychiatry, University Hospital Center and University of Lausanne, Prilly-Lausanne, Switzerland
| | - Raoul Jenni
- Center for Psychiatric Neuroscience, Department of Psychiatry, University Hospital Center and University of Lausanne, Prilly-Lausanne, Switzerland; Service of General Psychiatry, Department of Psychiatry, University Hospital Center and University of Lausanne, Prilly-Lausanne, Switzerland
| | - Margot Fournier
- Center for Psychiatric Neuroscience, Department of Psychiatry, University Hospital Center and University of Lausanne, Prilly-Lausanne, Switzerland
| | - Luis Alameda
- Center for Psychiatric Neuroscience, Department of Psychiatry, University Hospital Center and University of Lausanne, Prilly-Lausanne, Switzerland; Service of General Psychiatry, Department of Psychiatry, University Hospital Center and University of Lausanne, Prilly-Lausanne, Switzerland; Psychiatric Liaison Service, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Philipp S Baumann
- Center for Psychiatric Neuroscience, Department of Psychiatry, University Hospital Center and University of Lausanne, Prilly-Lausanne, Switzerland; Service of General Psychiatry, Department of Psychiatry, University Hospital Center and University of Lausanne, Prilly-Lausanne, Switzerland
| | - Stephanie Clarke
- Neuropsychology and Neurorehabilitation Service, University Hospital Center and University of Lausanne, 1011 Lausanne, Switzerland
| | - Philippe Conus
- Service of General Psychiatry, Department of Psychiatry, University Hospital Center and University of Lausanne, Prilly-Lausanne, Switzerland
| | - Kim Q Do
- Center for Psychiatric Neuroscience, Department of Psychiatry, University Hospital Center and University of Lausanne, Prilly-Lausanne, Switzerland
| | - Micah M Murray
- Neuropsychology and Neurorehabilitation Service, University Hospital Center and University of Lausanne, 1011 Lausanne, Switzerland; Radiodiagnostic Service, University Hospital Center and University of Lausanne, 1011 Lausanne, Switzerland; The EEG Brain Mapping Core, Center for Biomedical Imaging (CIBM), University Hospital Center and University of Lausanne, 1011 Lausanne, Switzerland; Psychiatric Liaison Service, Lausanne University Hospital (CHUV), Lausanne, Switzerland; Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Lausanne, Switzerland; Department of Hearing and Speech Sciences, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
43
|
Demro C, Rowland L, Wijtenburg SA, Waltz J, Gold J, Kline E, Thompson E, Reeves G, Hong LE, Schiffman J. Glutamatergic metabolites among adolescents at risk for psychosis. Psychiatry Res 2017; 257:179-185. [PMID: 28772136 PMCID: PMC8970349 DOI: 10.1016/j.psychres.2017.07.040] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 06/22/2017] [Accepted: 07/23/2017] [Indexed: 02/02/2023]
Abstract
Proton-Magnetic Resonance Spectroscopy (1H-MRS) may serve as an important tool for identifying biomarkers that aid the understanding of early psychosis, as development of this condition may be associated with metabolite concentration changes that reflect an alteration in glutamatergic mechanisms. The current study explored 1H-MRS metabolite concentrations in the striatum and anterior cingulate cortex (ACC) as potential biomarkers of psychosis-risk symptom severity. In a sample of 12 adolescents at clinical high-risk for psychosis, the subclinical symptom of grandiosity significantly correlated with glutamate in the ACC. Striatal glutathione, a marker of oxidative stress linked to the glutamatergic system, significantly correlated with grandiosity. Anterior cingulate glutathione significantly correlated with grandiosity and disorganized communication. These findings suggest that within a small sample of young people at clinical high-risk, glutamatergic metabolites are correlated with symptomatology generally predictive of conversion to psychosis. These mechanisms may serve as relevant biomarkers for facilitating prediction of symptom severity and providing insight into the etiology of early psychosis.
Collapse
Affiliation(s)
- Caroline Demro
- University of Maryland, Baltimore County, Department of Psychology, 1000 Hilltop Circle, Baltimore, MD 21250, USA
| | - Laura Rowland
- University of Maryland School of Medicine, Department of Psychiatry of Maryland Psychiatric Research Center, 55 Wade Avenue, Catonsville, MD 21228, USA
| | - S. Andrea Wijtenburg
- University of Maryland School of Medicine, Department of Psychiatry of Maryland Psychiatric Research Center, 55 Wade Avenue, Catonsville, MD 21228, USA
| | - James Waltz
- University of Maryland School of Medicine, Department of Psychiatry of Maryland Psychiatric Research Center, 55 Wade Avenue, Catonsville, MD 21228, USA
| | - James Gold
- University of Maryland School of Medicine, Department of Psychiatry of Maryland Psychiatric Research Center, 55 Wade Avenue, Catonsville, MD 21228, USA
| | - Emily Kline
- Harvard Medical School, Massachusetts Mental Health Center Department of Psychiatry of the Beth Israel Deaconess Medical Center, 75 Fenwood Road, Boston, MA 02115, USA
| | - Elizabeth Thompson
- University of Maryland, Baltimore County, Department of Psychology, 1000 Hilltop Circle, Baltimore, MD 21250, USA
| | - Gloria Reeves
- University of Maryland, Baltimore, 701 W. Pratt Street, Baltimore, MD 21201, USA
| | - L. Elliot Hong
- University of Maryland School of Medicine, Department of Psychiatry of Maryland Psychiatric Research Center, 55 Wade Avenue, Catonsville, MD 21228, USA
| | - Jason Schiffman
- University of Maryland, Baltimore County, Department of Psychology, 1000 Hilltop Circle, Baltimore, MD 21250, USA.
| |
Collapse
|
44
|
Ginsenoside Re protects against phencyclidine-induced behavioral changes and mitochondrial dysfunction via interactive modulation of glutathione peroxidase-1 and NADPH oxidase in the dorsolateral cortex of mice. Food Chem Toxicol 2017; 110:300-315. [PMID: 29037473 DOI: 10.1016/j.fct.2017.10.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 09/21/2017] [Accepted: 10/11/2017] [Indexed: 12/15/2022]
Abstract
We investigated whether ginsenoside Re (Re) modulates phencyclidine (PCP)-induced sociability deficits and recognition memory impairments to extend our recent finding. We examined the role of GPx-1 gene in the pharmacological activity of Re against mitochondrial dysfunction induced by PCP in the dorsolateral cortex of mice. Since mitochondrial oxidative stress activates NADPH oxidase (PHOX), we applied PHOX inhibitor apocynin for evaluating interactive modulation between GPx-1 and PHOX against PCP neurotoxicity. Sociability deficits and recognition memory impairments induced by PCP were more pronounced in GPx-1 knockout (KO) than in wild type (WT) mice. PCP-induced mitochondrial oxidative stress, mitochondrial dysfunction, and membrane translocation of p47phox were more evident in GPx-1 KO than in WT. Re treatment significantly attenuated PCP-induced neurotoxic changes. Re also significantly attenuated PCP-induced sociability deficits and recognition memory impairments. The attenuation by Re was comparable to that by apocynin. The attenuation was more obvious in GPx-1 KO than in WT. Importantly, apocynin did not show any additional positive effects on the neuroprotective activity of Re, indicating that PHOX is a molecular target for therapeutic activity of Re. Our results suggest that Re requires interactive modulation between GPx activity and PHOX (p47phox) to exhibit neuroprotective potentials against PCP insult.
Collapse
|
45
|
Barron H, Hafizi S, Mizrahi R. Towards an Integrated View of Early Molecular Changes Underlying Vulnerability to Social Stress in Psychosis. MODERN TRENDS IN PHARMACOPSYCHIATRY 2017; 31:96-106. [PMID: 28738349 DOI: 10.1159/000470810] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Psychotic disorders are heterogeneous and complex, involving many putative causal factors interacting along the course of disease development. Many of the factors implicated in the pathogenesis of psychosis also appear to be involved in disease onset and subsequent neuroprogression. Herein, we highlight the pertinent literature implicating inflammation and oxidative stress in the pathogenesis of psychosis, and the potential contribution of N-methyl-D-aspartate receptors (NMDARs). We also emphasize the role of peripubertal social stress in psychosis, and the ways in which hippocampal dysfunction can mediate dysregulation of the hypothalamic-pituitary-adrenal axis and cortisol release. Finally, we propose a model wherein inflammation and oxidative stress act as a first hit, producing altered parvalbumin interneuron development, NMDAR hypofunction, microglial priming, and sensitivity to a second hit of peripubertal social stress. With a greater understanding of how these factors interact, it may be possible to detect, prevent, and treat psychosis more effectively.
Collapse
|
46
|
Kim YK, Na KS. Neuroprotection in Schizophrenia and Its Therapeutic Implications. Psychiatry Investig 2017; 14:383-391. [PMID: 28845163 PMCID: PMC5561394 DOI: 10.4306/pi.2017.14.4.383] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 07/04/2016] [Accepted: 07/06/2016] [Indexed: 12/21/2022] Open
Abstract
Schizophrenia is a chronic and debilitating mental disorder. The persisting negative and cognitive symptoms that are unresponsive to pharmacotherapy reveal the impairment of neuroprotective aspects of schizophrenia. In this review, of the several neuroprotective factors, we mainly focused on neuroinflammation, neurogenesis, and oxidative stress. We conducted a narrative and selective review. Neuroinflammation is mainly mediated by pro-inflammatory cytokines and microglia. Unlike peripheral inflammatory responses, neuroinflammation has a role in various neuronal activities such as neurotransmission neurogenesis. The cross-talk between neuroinflammation and neurogenesis usually has beneficial effects in the CNS under physiological conditions. However, uncontrolled and chronic neuroinflammation exert detrimental effects such as neuronal loss, inhibited neurogenesis, and excessive oxidative stress. Neurogenesis is also a major component of neuroprotection. Adult neurogenesis mainly occurs in the hippocampal region, which has an important role in memory formation and processing. Impaired neurogenesis and an ineffective response to antipsychotics may be thought to indicate a deteriorating course of schizophrenia. Oxidative stress and excessive dopaminergic neurotransmission may create a vicious cycle and consequently disturb NMDA receptor-mediated glutamatergic neurotransmission. Based on the current evidences, several neuroprotective therapeutic approaches have been reported to be efficacious for improving psychopathology, but further longitudinal and large-sample based studies are needed.
Collapse
Affiliation(s)
- Yong-Ku Kim
- Department of Psychiatry, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Kyoung-Sae Na
- Department of Psychiatry, Gachon University Gil Medical Center, Incheon, Republic of Korea
| |
Collapse
|
47
|
Perkovic MN, Erjavec GN, Strac DS, Uzun S, Kozumplik O, Pivac N. Theranostic Biomarkers for Schizophrenia. Int J Mol Sci 2017; 18:E733. [PMID: 28358316 PMCID: PMC5412319 DOI: 10.3390/ijms18040733] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 03/23/2017] [Accepted: 03/27/2017] [Indexed: 12/14/2022] Open
Abstract
Schizophrenia is a highly heritable, chronic, severe, disabling neurodevelopmental brain disorder with a heterogeneous genetic and neurobiological background, which is still poorly understood. To allow better diagnostic procedures and therapeutic strategies in schizophrenia patients, use of easy accessible biomarkers is suggested. The most frequently used biomarkers in schizophrenia are those associated with the neuroimmune and neuroendocrine system, metabolism, different neurotransmitter systems and neurotrophic factors. However, there are still no validated and reliable biomarkers in clinical use for schizophrenia. This review will address potential biomarkers in schizophrenia. It will discuss biomarkers in schizophrenia and propose the use of specific blood-based panels that will include a set of markers associated with immune processes, metabolic disorders, and neuroendocrine/neurotrophin/neurotransmitter alterations. The combination of different markers, or complex multi-marker panels, might help in the discrimination of patients with different underlying pathologies and in the better classification of the more homogenous groups. Therefore, the development of the diagnostic, prognostic and theranostic biomarkers is an urgent and an unmet need in psychiatry, with the aim of improving diagnosis, therapy monitoring, prediction of treatment outcome and focus on the personal medicine approach in order to improve the quality of life in patients with schizophrenia and decrease health costs worldwide.
Collapse
Affiliation(s)
| | | | - Dubravka Svob Strac
- Rudjer Boskovic Institute, Division of Molecular Medicine, 10000 Zagreb, Croatia.
| | - Suzana Uzun
- Clinic for Psychiatry Vrapce, 10090 Zagreb, Croatia.
| | | | - Nela Pivac
- Rudjer Boskovic Institute, Division of Molecular Medicine, 10000 Zagreb, Croatia.
| |
Collapse
|
48
|
Neuroinflammation and Oxidative Stress in Psychosis and Psychosis Risk. Int J Mol Sci 2017; 18:ijms18030651. [PMID: 28304340 PMCID: PMC5372663 DOI: 10.3390/ijms18030651] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 03/07/2017] [Accepted: 03/15/2017] [Indexed: 12/18/2022] Open
Abstract
Although our understanding of psychotic disorders has advanced substantially in the past few decades, very little has changed in the standard of care for these illnesses since the development of atypical anti-psychotics in the 1990s. Here, we integrate new insights into the pathophysiology with the increasing interest in early detection and prevention. First, we explore the role of N-methyl-d-aspartate receptors in a subpopulation of cortical parvalbumin-containing interneurons (PVIs). Postmortem and preclinical data has implicated these neurons in the positive and negative symptoms, as well as the cognitive dysfunction present in schizophrenia. These neurons also appear to be sensitive to inflammation and oxidative stress during the perinatal and peripubertal periods, which may be mediated in large part by aberrant synaptic pruning. After exploring some of the molecular mechanisms through which neuroinflammation and oxidative stress are thought to exert their effects, we highlight the progress that has been made in identifying psychosis prior to onset through the identification of individuals at clinical high risk for psychosis (CHR). By combining our understanding of psychosis pathogenesis with the increasing characterization of endophenotypes that precede frank psychosis, it may be possible to identify patients before they present with psychosis and intervene to reduce the burden of the disease to both patients and families.
Collapse
|
49
|
Nakazawa K, Jeevakumar V, Nakao K. Spatial and temporal boundaries of NMDA receptor hypofunction leading to schizophrenia. NPJ SCHIZOPHRENIA 2017; 3:7. [PMID: 28560253 PMCID: PMC5441533 DOI: 10.1038/s41537-016-0003-3] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Revised: 11/17/2016] [Accepted: 11/22/2016] [Indexed: 12/21/2022]
Abstract
The N-methyl-d-aspartate receptor hypofunction is one of the most prevalent models of schizophrenia. For example, healthy subjects treated with uncompetitive N-methyl-d-aspartate receptor antagonists elicit positive, negative, and cognitive-like symptoms of schizophrenia. Patients with anti-N-methyl-d-aspartate receptor encephalitis, which is likely caused by autoantibody-mediated down-regulation of cell surface N-methyl-d-aspartate receptors, often experience psychiatric symptoms similar to schizophrenia initially. However, where and when N-methyl-d-aspartate receptor hypofunction occurs in the brain of schizophrenic patients is poorly understood. Here we review the findings from N-methyl-d-aspartate receptor antagonist and autoantibody models, postmortem studies on N-methyl-d-aspartate receptor subunits, as well as the global and cell-type-specific knockout mouse models of subunit GluN1. We compare various conditional GluN1 knockout mouse strains, focusing on the onset of N-methyl-d-aspartate receptor deletion and on the cortical cell-types. Based on these results, we hypothesize that N-methyl-d-aspartate receptor hypofunction initially occurs in cortical GABAergic neurons during early postnatal development. The resulting GABA neuron maturation deficit may cause reduction of intrinsic excitability and GABA release, leading to disinhibition of pyramidal neurons. The cortical disinhibition in turn could elicit glutamate spillover and subsequent homeostatic down regulation of N-methyl-d-aspartate receptor function in pyramidal neurons in prodromal stage. These two temporally-distinct N-methyl-d-aspartate receptor hypofunctions may be complimentary, as neither alone may not be able to fully explain the entire schizophrenia pathophysiology. Potential underlying mechanisms for N-methyl-d-aspartate receptor hypofunction in cortical GABA neurons are also discussed, based on studies of naturally-occurring N-methyl-d-aspartate receptor antagonists, neuregulin/ErbB4 signaling pathway, and theoretical analysis of excitatory/inhibitory balance.
Collapse
Affiliation(s)
- Kazu Nakazawa
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL USA
| | - Vivek Jeevakumar
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL USA
| | - Kazuhito Nakao
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL USA
| |
Collapse
|
50
|
Ben-Azu B, Aderibigbe AO, Ajayi AM, Iwalewa EO. Neuroprotective effects of the ethanol stem bark extracts of Terminalia ivorensis in ketamine-induced schizophrenia-like behaviors and oxidative damage in mice. PHARMACEUTICAL BIOLOGY 2016; 54:2871-2879. [PMID: 27250524 DOI: 10.1080/13880209.2016.1190382] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 02/18/2016] [Accepted: 05/12/2016] [Indexed: 06/05/2023]
Abstract
CONTEXT Schizophrenia is a heterogenous neurological disorder, which has been hypothetically linked to oxidative imbalance and associated behavioral perturbations. Preliminary evidence from animal models predictive of human psychosis suggests that Terminalia ivorensis A. Chev. (Combretaceae) has antipsychotic-like activity in mice. OBJECTIVE This study investigates the neuroprotective property of the ethanol stem bark extracts of T. ivorensis (EETI) in reversal treatment of ketamine-induced schizophrenia-like behaviors and oxidative alteration in adult male Swiss albino mice. MATERIALS AND METHODS Animals were divided into six treatment groups (n = 5). Animals received distilled water or ketamine (20 mg/kg) once daily intraperitoneally (i.p.) for 14 days, and from the 8th to the 14th day, they were treated with EETI (125, 250 or 500 mg/kg), risperidone (RIS) or vehicle orally once daily. Behaviors related to positive (locomotor activity) and cognitive (Y maze) symptoms of schizophrenia were assessed. Glutathione (GSH) levels, superoxide dismutase (SOD) and catalase (CAT) activities, including malondialdehyde (MDA) concentration were measured in mice whole brains. RESULT The LD50 of EETI was 2236.06 mg/kg, p.o. body weight. EETI (125, 250 or 500 mg/kg, p.o.) demonstrated significant (p < 0.05) inhibition of ketamine-induced hyperlocomotion and cognitive dysfunction. The extract decreased MDA concentration (39.0, 62.6 and 67.5%) in a dose-dependent manner. Moreover, EETI significantly (p < 0.05) reversed the depletion of GSH, and increased activities of SOD and CAT in brain tissues. DISCUSSION AND CONCLUSION These findings suggest that EETI probably exert its antipsychotic-like activity, via a neuroprotective compensatory mechanism of action, and as such, could be relevant in the management of schizophrenia.
Collapse
Affiliation(s)
- Benneth Ben-Azu
- a Department of Pharmacology and Therapeutics, College of Medicine , University of Ibadan , Ibadan , Oyo State , Nigeria
| | - Adegbuyi Oladele Aderibigbe
- a Department of Pharmacology and Therapeutics, College of Medicine , University of Ibadan , Ibadan , Oyo State , Nigeria
| | - Abayomi Mayowa Ajayi
- a Department of Pharmacology and Therapeutics, College of Medicine , University of Ibadan , Ibadan , Oyo State , Nigeria
| | - Ezekiel Oluwagbenga Iwalewa
- a Department of Pharmacology and Therapeutics, College of Medicine , University of Ibadan , Ibadan , Oyo State , Nigeria
| |
Collapse
|