1
|
Duncan LE, Li T, Salem M, Li W, Mortazavi L, Senturk H, Shahverdizadeh N, Vesuna S, Shen H, Yoon J, Wang G, Ballon J, Tan L, Pruett BS, Knutson B, Deisseroth K, Giardino WJ. Mapping the cellular etiology of schizophrenia and complex brain phenotypes. Nat Neurosci 2025; 28:248-258. [PMID: 39833308 PMCID: PMC11802450 DOI: 10.1038/s41593-024-01834-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 10/29/2024] [Indexed: 01/22/2025]
Abstract
Psychiatric disorders are multifactorial and effective treatments are lacking. Probable contributing factors to the challenges in therapeutic development include the complexity of the human brain and the high polygenicity of psychiatric disorders. Combining well-powered genome-wide and brain-wide genetics and transcriptomics analyses can deepen our understanding of the etiology of psychiatric disorders. Here, we leverage two landmark resources to infer the cell types involved in the etiology of schizophrenia, other psychiatric disorders and informative comparison of brain phenotypes. We found both cortical and subcortical neuronal associations for schizophrenia, bipolar disorder and depression. These cell types included somatostatin interneurons, excitatory neurons from the retrosplenial cortex and eccentric medium spiny-like neurons from the amygdala. In contrast we found T cell and B cell associations with multiple sclerosis and microglial associations with Alzheimer's disease. We provide a framework for a cell-type-based classification system that can lead to drug repurposing or development opportunities and personalized treatments. This work formalizes a data-driven, cellular and molecular model of complex brain disorders.
Collapse
Affiliation(s)
- Laramie E Duncan
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
| | - Tayden Li
- Department of Statistics, Stanford University, Stanford, CA, USA
| | - Madeleine Salem
- Vice Provost for Undergraduate Education, Stanford University, Stanford, CA, USA
| | - Will Li
- Vice Provost for Undergraduate Education, Stanford University, Stanford, CA, USA
| | - Leili Mortazavi
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Psychology, Stanford University, Stanford, CA, USA
| | - Hazal Senturk
- Department of Computer Science, University of San Francisco, San Francisco, CA, USA
| | - Naghmeh Shahverdizadeh
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Sam Vesuna
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Hanyang Shen
- Department of Epidemiology, Stanford University, Stanford, CA, USA
| | - Jong Yoon
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Gordon Wang
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Jacob Ballon
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Longzhi Tan
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Neurobiology, Stanford University, Stanford, CA, USA
| | | | - Brian Knutson
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Psychology, Stanford University, Stanford, CA, USA
| | - Karl Deisseroth
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - William J Giardino
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| |
Collapse
|
2
|
Hou PS, Lin SF, Zhu JD, Chung CY, Tsai SJ, Yang AC. Local cortical structure pattern and genetic links in schizophrenia: An MRI and CRISPR/Cas9 study. Prog Neuropsychopharmacol Biol Psychiatry 2025; 137:111270. [PMID: 39862915 DOI: 10.1016/j.pnpbp.2025.111270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 01/05/2025] [Accepted: 01/21/2025] [Indexed: 01/27/2025]
Abstract
While the etiology of schizophrenia (SZ) remains elusive, its diverse phenotypes suggest the involvement of distinct functional cortical areas, and the heritability of SZ implies the underlying genetic factors. This study aimed to integrate imaging and molecular analyses to elucidate the genetic underpinnings of SZ. We investigated the local cortical structural pattern changes in Brodmann areas (BAs) by calculating the cortical structural pattern index (SPI) using magnetic resonance imaging analysis from 194 individuals with SZ and 330 controls. Significant local structural changes were detected in certain Brodmann areas in symmetric or asymmetric patterns, such as symmetric changes in the BA4 primary motor area and BA23 part of posterior cingulate cortex, and asymmetric changes in the BA13 insula, BA11 inferior orbitofrontal area, and BA 24, and BA 31 cingulate cortex. Following genome-wide association tests, we found genetic variants and SNP-mapped genes and verified the areal preferential expression profiles in the developing human and mouse neocortex. Finally, we performed a loss-of-function analysis using the CRISPR/Cas9 system to investigate the effects of disrupting the SZ-related SNP-mapped Morf4l1, Reep3, or Tmed3 gene on cortical cell fate to understand their roles in generating appropriate composition of cortical neurons. This study outlines a pipeline for identifying local structural changes, associated genetic causes, and potential molecular mechanisms underlying mental disorders. Additionally, these data shed light on establishing a structurally integral cerebral cortex for higher cognitive functions.
Collapse
Affiliation(s)
- Pei-Shan Hou
- Institute of Anatomy and Cell Biology, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; Institute of Brain Science, National Yang-Ming Chiao Tung University, Taipei 11221, Taiwan; Brain Research Center, National Yang-Ming Chiao Tung University, Taipei 11221, Taiwan.
| | - Shu-Fei Lin
- Institute of Anatomy and Cell Biology, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Jun-Ding Zhu
- Department of Occupational Therapy, College of Medical Science and Technology, Chung Shan Medical University, Taichung 40201, Taiwan; Occupational Therapy Room, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Chih-Yun Chung
- Institute of Anatomy and Cell Biology, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Shih-Jen Tsai
- Institute of Brain Science, National Yang-Ming Chiao Tung University, Taipei 11221, Taiwan; Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Albert C Yang
- Institute of Brain Science, National Yang-Ming Chiao Tung University, Taipei 11221, Taiwan; Brain Research Center, National Yang-Ming Chiao Tung University, Taipei 11221, Taiwan; Digital Medicine and Smart Healthcare Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
3
|
Segal A, Tiego J, Parkes L, Holmes AJ, Marquand AF, Fornito A. Embracing variability in the search for biological mechanisms of psychiatric illness. Trends Cogn Sci 2025; 29:85-99. [PMID: 39510933 PMCID: PMC11742270 DOI: 10.1016/j.tics.2024.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 11/15/2024]
Abstract
Despite decades of research, we lack objective diagnostic or prognostic biomarkers of mental health problems. A key reason for this limited progress is a reliance on the traditional case-control paradigm, which assumes that each disorder has a single cause that can be uncovered by comparing average phenotypic values of patient and control samples. Here, we discuss the problematic assumptions on which this paradigm is based and highlight recent efforts that seek to characterize, rather than minimize, the inherent clinical and biological variability that underpins psychiatric populations. Embracing such variability is necessary to understand pathophysiological mechanisms and develop more targeted and effective treatments.
Collapse
Affiliation(s)
- Ashlea Segal
- Wu-Tsai Institute, and Department of Neuroscience, School of Medicine, Yale University, New Haven, CT 06520, USA; School of Psychological Sciences, Turner Institute for Brain and Mental Health, and Monash Biomedical Imaging, Monash University, Melbourne 3800, Australia.
| | - Jeggan Tiego
- School of Psychological Sciences, Turner Institute for Brain and Mental Health, and Monash Biomedical Imaging, Monash University, Melbourne 3800, Australia
| | - Linden Parkes
- Brain Health Institute, Department of Psychiatry, Rutgers University, Piscataway, NJ 08854, USA
| | - Avram J Holmes
- Brain Health Institute, Department of Psychiatry, Rutgers University, Piscataway, NJ 08854, USA
| | - Andre F Marquand
- Department of Cognitive Neuroscience, Radboud UMC, 6500 HB Nijmegen, The Netherlands; Donders Institute for Cognition, Brain and Behavior, 6525 EN Nijmegen, The Netherlands
| | - Alex Fornito
- School of Psychological Sciences, Turner Institute for Brain and Mental Health, and Monash Biomedical Imaging, Monash University, Melbourne 3800, Australia
| |
Collapse
|
4
|
Ikeda Y, Nakashima M, Yoshikawa S, Taniguchi K, Suga N, Matsuda S. Behavioral Test Scores Could Be Linked to the Protein Expression Values of p62 and GLAST in the Brains of Mice with Neuropsychiatric Disorder-Related Behaviors. BIOLOGY 2024; 13:1039. [PMID: 39765706 PMCID: PMC11672909 DOI: 10.3390/biology13121039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/23/2024] [Accepted: 10/23/2024] [Indexed: 01/11/2025]
Abstract
Neuropsychiatric disorders are a public health concern, in which diagnosis and prognosis may be based on clinical symptoms that might often diverge across individuals. Schizophrenia is a major neuropsychiatric disorder, which may affect millions worldwide. However, the biochemical alterations of this disorder have not been comprehensively distinguished. In addition, there is less confidence in finding specific biomarkers for neuropsychiatric disorders, including schizophrenia, but rather a specific characteristic behavioral pattern. In general, maternal immune activation is considered to be one of the important factors in the development of neuropsychiatric disorders. Here, a mouse model of neuropsychiatric disorders was created, in which poly I:C, sodium dextran sulfate (DSS), and κ-carrageenan (CGN) were utilized for maternal immune activation during the pregnancies of mother mice. Subsequently, we examined the link between biochemical changes in p62 and/or glutamate aspartate transporter (GLAST) in the brains of offspring mice and the alteration in their experimental behavior scores. Furthermore, a therapeutic study was conducted on these neuropsychiatric disorder model mice using butyric acid, piceid, and metformin. It was found that some molecules could effectively improve the behavioral scores of neuropsychiatric model mice. Importantly, significant correlations between certain behavioral scores and p62 protein expression, as well as between the scores and GLAST expression, were recognized. This is the first report of a significant correlation between pathological behaviors and biochemical alterations in neuropsychiatric disorder model animals. This concept could contribute to the development of innovative treatments to at least ameliorate the symptoms of several psychiatric disorders.
Collapse
Affiliation(s)
| | | | | | | | | | - Satoru Matsuda
- Department of Food Science and Nutrition, Nara Women’s University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| |
Collapse
|
5
|
Horan WP, Targum SD, Claxton A, Kaul I, Yohn SE, Marder SR, Miller AC, Brannan SK. Efficacy of KarXT on negative symptoms in acute schizophrenia: A post hoc analysis of pooled data from 3 trials. Schizophr Res 2024; 274:57-65. [PMID: 39260339 DOI: 10.1016/j.schres.2024.08.001] [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: 02/09/2024] [Revised: 06/06/2024] [Accepted: 08/08/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND Currently approved antipsychotics do not adequately treat negative symptoms (NS), which are a major determinant of functional disability in schizophrenia. KarXT, an M1 /M4 preferring muscarinic receptor agonist, has shown efficacy as a broad-spectrum monotherapy for the treatment of schizophrenia in participants with acute psychosis. Post hoc analyses evaluated the possibility that NS improve independently of positive symptoms with KarXT in a subgroup of participants with moderate-to-severe NS and no predominance of positive symptoms. METHODS Data were pooled from the three pivotal trials of KarXT monotherapy in people with schizophrenia with an acute exacerbation of psychosis. All 3 studies used similar 5-week randomized, double-blind, placebo-controlled designs (modified intention-to-treat sample N = 640). PANSS criteria proposed in the literature identified a subset of study participants (n = 64) with prominent NS. RESULTS KarXT was significantly better than placebo on PANSS Marder Negative Factor Scores in the full sample (p < .001; Cohen's d = 0.42) and more so in the prominent NS subgroup (p < .001; Cohen's d = 1.18). Further, the KarXT effect in the NS subgroup remained significant after accounting for changes in positive symptoms, depression/anxiety, disorganization, and hostility. CONCLUSIONS Participants with prominent NS revealed greater improvement of NS following KarXT therapy than the full sample that persisted after accounting for positive and other symptoms. While these findings must be interpreted with caution, they are consistent with the possibility that NS improvements associated with KarXT are not secondary to improvements in other symptom domains and support further investigation in larger, stable outpatient studies.
Collapse
Affiliation(s)
- William P Horan
- Bristol Myers Squibb, Princeton, NJ, USA; University of California, Los Angeles, CA, USA.
| | | | | | - Inder Kaul
- Bristol Myers Squibb, Princeton, NJ, USA
| | | | | | | | | |
Collapse
|
6
|
Muhtaseb AW, Duan J. Modeling common and rare genetic risk factors of neuropsychiatric disorders in human induced pluripotent stem cells. Schizophr Res 2024; 273:39-61. [PMID: 35459617 PMCID: PMC9735430 DOI: 10.1016/j.schres.2022.04.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/05/2022] [Accepted: 04/07/2022] [Indexed: 12/13/2022]
Abstract
Recent genome-wide association studies (GWAS) and whole-exome sequencing of neuropsychiatric disorders, especially schizophrenia, have identified a plethora of common and rare disease risk variants/genes. Translating the mounting human genetic discoveries into novel disease biology and more tailored clinical treatments is tied to our ability to causally connect genetic risk variants to molecular and cellular phenotypes. When combined with the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/CRISPR-associated (Cas) nuclease-mediated genome editing system, human induced pluripotent stem cell (hiPSC)-derived neural cultures (both 2D and 3D organoids) provide a promising tractable cellular model for bridging the gap between genetic findings and disease biology. In this review, we first conceptualize the advances in understanding the disease polygenicity and convergence from the past decade of iPSC modeling of different types of genetic risk factors of neuropsychiatric disorders. We then discuss the major cell types and cellular phenotypes that are most relevant to neuropsychiatric disorders in iPSC modeling. Finally, we critically review the limitations of iPSC modeling of neuropsychiatric disorders and outline the need for implementing and developing novel methods to scale up the number of iPSC lines and disease risk variants in a systematic manner. Sufficiently scaled-up iPSC modeling and a better functional interpretation of genetic risk variants, in combination with cutting-edge CRISPR/Cas9 gene editing and single-cell multi-omics methods, will enable the field to identify the specific and convergent molecular and cellular phenotypes in precision for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Abdurrahman W Muhtaseb
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, United States of America; Department of Human Genetics, The University of Chicago, Chicago, IL 60637, United States of America
| | - Jubao Duan
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, United States of America; Department of Psychiatry and Behavioral Neuroscience, The University of Chicago, Chicago, IL 60637, United States of America.
| |
Collapse
|
7
|
Hughes H, Brady LJ, Schoonover KE. GABAergic dysfunction in postmortem dorsolateral prefrontal cortex: implications for cognitive deficits in schizophrenia and affective disorders. Front Cell Neurosci 2024; 18:1440834. [PMID: 39381500 PMCID: PMC11458443 DOI: 10.3389/fncel.2024.1440834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 09/05/2024] [Indexed: 10/10/2024] Open
Abstract
The microcircuitry within superficial layers of the dorsolateral prefrontal cortex (DLPFC), composed of excitatory pyramidal neurons and inhibitory GABAergic interneurons, has been suggested as the neural substrate of working memory performance. In schizophrenia, working memory impairments are thought to result from alterations of microcircuitry within the DLPFC. GABAergic interneurons, in particular, are crucially involved in synchronizing neural activity at gamma frequency, the power of which increases with working memory load. Alterations of GABAergic interneurons, particularly parvalbumin (PV) and somatostatin (SST) subtypes, are frequently observed in schizophrenia. Abnormalities of GABAergic neurotransmission, such as deficiencies in the 67 kDA isoform of GABA synthesis enzyme (GAD67), vesicular GABA transporter (vGAT), and GABA reuptake transporter 1 (GAT1) in presynaptic boutons, as well as postsynaptic alterations in GABA A receptor subunits further contribute to impaired inhibition. This review explores GABAergic abnormalities of the postmortem DLPFC in schizophrenia, with a focus on the roles of interneuron subtypes involved in cognition, and GABAergic neurotransmission within presynaptic boutons and postsynaptic alterations. Where available, comparisons between schizophrenia and affective disorders that share cognitive pathology such as bipolar disorder and major depressive disorder will be made. Challenges in directly measuring GABA levels are addressed, emphasizing the need for innovative techniques. Understanding GABAergic abnormalities and their implications for neural circuit dysfunction in schizophrenia is crucial for developing targeted therapies.
Collapse
Affiliation(s)
- Hannah Hughes
- Graduate Biomedical Sciences Program, School of Medicine, University of Alabama at Birmingham, Tuskegee, AL, United States
| | - Lillian J. Brady
- Department of Psychiatry, School of Medicine, University of Alabama at Birmingham, Tuskegee, AL, United States
- Comprehensive Neuroscience Center, University of Alabama at Birmingham, Tuskegee, AL, United States
| | - Kirsten E. Schoonover
- Department of Psychiatry, School of Medicine, University of Alabama at Birmingham, Tuskegee, AL, United States
- Comprehensive Neuroscience Center, University of Alabama at Birmingham, Tuskegee, AL, United States
- Department of Psychology and Sociology, College of Arts and Sciences, Tuskegee University, Tuskegee, AL, United States
| |
Collapse
|
8
|
Asad Z, Fakheir Y, Abukhaled Y, Khalil R. Implications of altered pyramidal cell morphology on clinical symptoms of neurodevelopmental disorders. Eur J Neurosci 2024; 60:4877-4892. [PMID: 39054743 DOI: 10.1111/ejn.16484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 05/26/2024] [Accepted: 07/13/2024] [Indexed: 07/27/2024]
Abstract
The prevalence of pyramidal cells (PCs) in the mammalian cerebral cortex underscore their value as they play a crucial role in various brain functions, ranging from cognition, sensory processing, to motor output. PC morphology significantly influences brain connectivity and plays a critical role in maintaining normal brain function. Pathological alterations to PC morphology are thought to contribute to the aetiology of neurodevelopmental disorders such as autism spectrum disorder (ASD) and schizophrenia. This review explores the relationship between abnormalities in PC morphology in key cortical areas and the clinical manifestations in schizophrenia and ASD. We focus largely on human postmortem studies and provide evidence that dendritic segment length, complexity and spine density are differentially affected in these disorders. These morphological alterations can lead to disruptions in cortical connectivity, potentially contributing to the cognitive and behavioural deficits observed in these disorders. Furthermore, we highlight the importance of investigating the functional and structural characteristics of PCs in these disorders to illuminate the underlying pathogenesis and stimulate further research in this area.
Collapse
Affiliation(s)
- Zummar Asad
- School of Medicine, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland
| | - Yara Fakheir
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Yara Abukhaled
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Reem Khalil
- Department of Biology, Chemistry and Environmental Sciences, American University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
9
|
Dutra-Tavares AC, Couto LA, Souza TP, Bandeira-Martins A, Silva JO, Filgueiras CC, Ribeiro-Carvalho A, Manhães AC, Abreu-Villaça Y. Nicotine's Effects on Schizophrenia-like Symptoms in a Mice Model: Time Matters. Brain Sci 2024; 14:855. [PMID: 39335351 PMCID: PMC11430416 DOI: 10.3390/brainsci14090855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 08/12/2024] [Accepted: 08/18/2024] [Indexed: 09/30/2024] Open
Abstract
Tobacco consumption in schizophrenia (SCHZ) patients is highly prevalent. Data support the occurrence of sequential events during comorbidity establishment, and both smoking first, SCHZ second and SCHZ first, smoking second sequences have been proposed. To investigate whether these two possibilities lead to distinct outcomes of comorbidity, we used a phencyclidine-induced SCHZ model and nicotine exposure as a surrogate of smoking. C57Bl/6 mice were submitted to a protocol that either began with 4 days of phencyclidine exposure or 4 days of nicotine exposure. This period was followed by 5 days of combined phencyclidine + nicotine exposure. Locomotor sensitization and pre-pulse inhibition (PPI) were assessed due to their well-known associations with SCHZ as opposed to rearing, an unrelated behavior. Nicotine priming potentiated phencyclidine-evoked sensitization. However, nicotine exposure after SCHZ modeling did not interfere with phencyclidine's effects. In the PPI test, nicotine after SCHZ modeling worsened the phencyclidine-evoked deficiency in males. In contrast, nicotine priming had no effects. Regarding rearing, nicotine priming failed to interfere with phencyclidine-mediated inhibition. Similarly, phencyclidine priming did not modify nicotine-mediated inhibition. The present results indicate that the sequence, either SCHZ-first or nicotine-first, differentially impacts comorbidity outcomes, a finding that is relevant for the identification of mechanisms of nicotine interference in the neurobiology of SCHZ.
Collapse
Affiliation(s)
- Ana Carolina Dutra-Tavares
- Departamento de Ciências Biomédicas e Saúde, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Cabo Frio 28905-320, RJ, Brazil;
| | - Luciana Araújo Couto
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro UERJ, Rio de Janeiro 20550-170, RJ, Brazil (T.P.S.); (A.B.-M.); (J.O.S.); (C.C.F.); (Y.A.-V.)
| | - Thainá P. Souza
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro UERJ, Rio de Janeiro 20550-170, RJ, Brazil (T.P.S.); (A.B.-M.); (J.O.S.); (C.C.F.); (Y.A.-V.)
| | - Anais Bandeira-Martins
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro UERJ, Rio de Janeiro 20550-170, RJ, Brazil (T.P.S.); (A.B.-M.); (J.O.S.); (C.C.F.); (Y.A.-V.)
| | - Juliana Oliveira Silva
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro UERJ, Rio de Janeiro 20550-170, RJ, Brazil (T.P.S.); (A.B.-M.); (J.O.S.); (C.C.F.); (Y.A.-V.)
| | - Claudio C. Filgueiras
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro UERJ, Rio de Janeiro 20550-170, RJ, Brazil (T.P.S.); (A.B.-M.); (J.O.S.); (C.C.F.); (Y.A.-V.)
| | - Anderson Ribeiro-Carvalho
- Departamento de Ciências, Faculdade de Formação de Professores, UERJ, São Gonçalo 24435-005, RJ, Brazil;
| | - Alex C. Manhães
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro UERJ, Rio de Janeiro 20550-170, RJ, Brazil (T.P.S.); (A.B.-M.); (J.O.S.); (C.C.F.); (Y.A.-V.)
| | - Yael Abreu-Villaça
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro UERJ, Rio de Janeiro 20550-170, RJ, Brazil (T.P.S.); (A.B.-M.); (J.O.S.); (C.C.F.); (Y.A.-V.)
| |
Collapse
|
10
|
Mana L, Schwartz-Pallejà M, Vila-Vidal M, Deco G. Overview on cognitive impairment in psychotic disorders: From impaired microcircuits to dysconnectivity. Schizophr Res 2024; 269:132-143. [PMID: 38788432 DOI: 10.1016/j.schres.2024.05.008] [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: 01/15/2024] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024]
Abstract
Schizophrenia's cognitive deficits, often overshadowed by positive symptoms, significantly contribute to the disorder's morbidity. Increasing attention highlights these deficits as reflections of neural circuit dysfunction across various cortical regions. Numerous connectivity alterations linked to cognitive symptoms in psychotic disorders have been reported, both at the macroscopic and microscopic level, emphasizing the potential role of plasticity and microcircuits impairment during development and later stages. However, the heterogeneous clinical presentation of cognitive impairment and diverse connectivity findings pose challenges in summarizing them into a cohesive picture. This review aims to synthesize major cognitive alterations, recent insights into network structural and functional connectivity changes and proposed mechanisms and microcircuit alterations underpinning these symptoms, particularly focusing on neurodevelopmental impairment, E/I balance, and sleep disturbances. Finally, we will also comment on some of the most recent and promising therapeutic approaches that aim to target these mechanisms to address cognitive symptoms. Through this comprehensive exploration, we strive to provide an updated and nuanced overview of the multiscale connectivity impairment underlying cognitive impairment in psychotic disorders.
Collapse
Affiliation(s)
- L Mana
- Center for Brain and Cognition, Computational Neuroscience Group, Department of Information and Communication Technologies, Universitat Pompeu Fabra, Roc Boronat 138, Barcelona 08018, Spain.
| | - M Schwartz-Pallejà
- Center for Brain and Cognition, Computational Neuroscience Group, Department of Information and Communication Technologies, Universitat Pompeu Fabra, Roc Boronat 138, Barcelona 08018, Spain; Department of Experimental and Health Science, Universitat Pompeu Fabra, Roc Boronat 138, Barcelona 08018, Spain; Eurecat, Technology Center of Catalonia, Multimedia Technologies, Barcelona, Spain.
| | - M Vila-Vidal
- Center for Brain and Cognition, Computational Neuroscience Group, Department of Information and Communication Technologies, Universitat Pompeu Fabra, Roc Boronat 138, Barcelona 08018, Spain; Computational Biology and Complex Systems Group, Department of Physics, Universitat Politècnica de Catalunya, Barcelona, Spain.
| | - G Deco
- Center for Brain and Cognition, Computational Neuroscience Group, Department of Information and Communication Technologies, Universitat Pompeu Fabra, Roc Boronat 138, Barcelona 08018, Spain; Institució Catalana de la Recerca i Estudis Avançats (ICREA), Passeig Lluís Companys 23, Barcelona 08010, Spain.
| |
Collapse
|
11
|
Nesbit MO, Ahn S, Zou H, Floresco SB, Phillips AG. Potentiation of prefrontal cortex dopamine function by the novel cognitive enhancer d-govadine. Neuropharmacology 2024; 246:109849. [PMID: 38244888 DOI: 10.1016/j.neuropharm.2024.109849] [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: 10/09/2023] [Revised: 12/13/2023] [Accepted: 01/16/2024] [Indexed: 01/22/2024]
Abstract
Cognitive impairment is a debilitating feature of psychiatric disorders including schizophrenia, mood disorders and substance use disorders for which there is a substantial lack of effective therapies. d-Govadine (d-GOV) is a tetrahydroprotoberberine recently shown to significantly enhance working memory and behavioural flexibility in several prefrontal cortex (PFC)-dependent rodent tasks. d-GOV potentiates dopamine (DA) efflux in the mPFC and not the nucleus accumbens, a unique pharmacology that sets it apart from many dopaminergic drugs and likely contributes to its effects on cognitive function. However, specific mechanisms involved in the preferential effects of d-GOV on mPFC DA function remain to be determined. The present study employs brain dialysis in male rats to deliver d-GOV into the mPFC or ventral tegmental area (VTA), while simultaneously sampling DA and norepinephrine (NE) efflux in the mPFC. Intra-PFC delivery or systemic administration of d-GOV preferentially potentiated medial prefrontal DA vs NE efflux. This differential effect of d-GOV on the primary catecholamines known to affect mPFC function further underscores its specificity for the mPFC DA system. Importantly, the potentiating effect of d-GOV on mPFC DA was disrupted when glutamatergic transmission was blocked in either the mPFC or the VTA. We hypothesize that d-GOV acts in the mPFC to engage the mesocortical feedback loop through which prefrontal glutamatergic projections activate a population of VTA DA neurons that specifically project back to the PFC. The activation of a PFC-VTA feedback loop to elevate PFC DA efflux without affecting mesolimbic DA release represents a novel approach to developing pro-cognitive drugs.
Collapse
Affiliation(s)
- Maya O Nesbit
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Soyon Ahn
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Haiyan Zou
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Stan B Floresco
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Anthony G Phillips
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
12
|
Spark DL, Ma S, Nowell CJ, Langmead CJ, Stewart GD, Nithianantharajah J. Sex-Dependent Attentional Impairments in a Subchronic Ketamine Mouse Model for Schizophrenia. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2024; 4:229-239. [PMID: 38298794 PMCID: PMC10829638 DOI: 10.1016/j.bpsgos.2023.05.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 05/16/2023] [Accepted: 05/18/2023] [Indexed: 02/02/2024] Open
Abstract
Background The development of more effective treatments for schizophrenia targeting cognitive and negative symptoms has been limited, partly due to a disconnect between rodent models and human illness. Ketamine administration is widely used to model symptoms of schizophrenia in both humans and rodents. In mice, subchronic ketamine treatment reproduces key dopamine and glutamate dysfunction; however, it is unclear how this translates into behavioral changes reflecting positive, negative, and cognitive symptoms. Methods In male and female mice treated with either subchronic ketamine or saline, we assessed spontaneous and amphetamine-induced locomotor activity to measure behaviors relevant to positive symptoms, and used a touchscreen-based progressive ratio task of motivation and the rodent continuous performance test of attention to capture specific negative and cognitive symptoms, respectively. To explore neuronal changes underlying the behavioral effects of subchronic ketamine treatment, we quantified expression of the immediate early gene product, c-Fos, in key corticostriatal regions using immunofluorescence. Results We showed that spontaneous locomotor activity was unchanged in male and female subchronic ketamine-treated animals, and amphetamine-induced locomotor response was reduced. Subchronic ketamine treatment did not alter motivation in either male or female mice. In contrast, we identified a sex-specific effect of subchronic ketamine on attentional processing wherein female mice performed worse than control mice due to increased nonselective responding. Finally, we showed that subchronic ketamine treatment increased c-Fos expression in prefrontal cortical and striatal regions, consistent with a mechanism of widespread disinhibition of neuronal activity. Conclusions Our results highlight that the subchronic ketamine mouse model reproduces a subset of behavioral symptoms that are relevant for schizophrenia.
Collapse
Affiliation(s)
- Daisy L. Spark
- Drug Discovery Biology Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Neuroscience & Mental Health Therapeutic Program Area, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Neuromedicines Discovery Centre, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Sherie Ma
- Drug Discovery Biology Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Cameron J. Nowell
- Drug Discovery Biology Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Christopher J. Langmead
- Drug Discovery Biology Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Neuroscience & Mental Health Therapeutic Program Area, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Neuromedicines Discovery Centre, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Gregory D. Stewart
- Drug Discovery Biology Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Neuroscience & Mental Health Therapeutic Program Area, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Neuromedicines Discovery Centre, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Jess Nithianantharajah
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
13
|
Dutra-Tavares AC, Souza TP, Silva JO, Semeão KA, Mello FF, Filgueiras CC, Ribeiro-Carvalho A, Manhães AC, Abreu-Villaça Y. Neonatal phencyclidine as a model of sex-biased schizophrenia symptomatology in adolescent mice. Psychopharmacology (Berl) 2023; 240:2111-2129. [PMID: 37530885 DOI: 10.1007/s00213-023-06434-3] [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: 04/22/2023] [Accepted: 07/22/2023] [Indexed: 08/03/2023]
Abstract
Sex-biased differences in schizophrenia are evident in several features of the disease, including symptomatology and response to pharmacological treatments. As a neurodevelopmental disorder, these differences might originate early in life and emerge later during adolescence. Considering that the disruption of the glutamatergic system during development is known to contribute to schizophrenia, we hypothesized that the neonatal phencyclidine model could induce sex-dependent behavioral and neurochemical changes associated with this disorder during adolescence. C57BL/6 mice received either saline or phencyclidine (5, 10, or 20 mg/kg) on postnatal days (PN) 7, 9, and 11. Behavioral assessment occurred in late adolescence (PN48-50), when mice were submitted to the open field, social interaction, and prepulse inhibition tests. Either olanzapine or saline was administered before each test. The NMDAR obligatory GluN1 subunit and the postsynaptic density protein 95 (PSD-95) were evaluated in the frontal cortex and hippocampus at early (PN30) and late (PN50) adolescence. Neonatal phencyclidine evoked dose-dependent deficits in all analyzed behaviors and males were more susceptible. Males also had reduced GluN1 expression in the frontal cortex at PN30. There were late-emergent effects at PN50. Cortical GluN1 was increased in both sexes, while phencyclidine increased cortical and decreased hippocampal PSD-95 in females. Olanzapine failed to mitigate most phencyclidine-evoked alterations. In some instances, this antipsychotic aggravated the deficits or potentiated subthreshold effects. These results lend support to the use of neonatal phencyclidine as a sex-biased neurodevelopmental preclinical model of schizophrenia. Olanzapine null effects and deleterious outcomes suggest that its use during adolescence should be further evaluated.
Collapse
Affiliation(s)
- Ana Carolina Dutra-Tavares
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Av. Prof. Manuel de Abreu 444, 5 andar, Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil
| | - Thainá P Souza
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Av. Prof. Manuel de Abreu 444, 5 andar, Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil
| | - Juliana O Silva
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Av. Prof. Manuel de Abreu 444, 5 andar, Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil
| | - Keila A Semeão
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Av. Prof. Manuel de Abreu 444, 5 andar, Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil
| | - Felipe F Mello
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Av. Prof. Manuel de Abreu 444, 5 andar, Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil
| | - Claudio C Filgueiras
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Av. Prof. Manuel de Abreu 444, 5 andar, Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil
| | - Anderson Ribeiro-Carvalho
- Departamento de Ciências, Faculdade de Formação de Professores da Universidade do Estado do Rio de Janeiro (UERJ), RJ, São Gonçalo, Brazil
| | - Alex C Manhães
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Av. Prof. Manuel de Abreu 444, 5 andar, Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil
| | - Yael Abreu-Villaça
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Av. Prof. Manuel de Abreu 444, 5 andar, Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil.
| |
Collapse
|
14
|
Zhang WJ, Shi HZ, Guo MN, Xu LF, Zhai HR, Liu ZZ, Zhu YQ, Zhang WN, Wang J. PGC-1α regulates critical period onset/closure, mediating cortical plasticity. Front Mol Neurosci 2023; 16:1149906. [PMID: 37822967 PMCID: PMC10563514 DOI: 10.3389/fnmol.2023.1149906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 08/29/2023] [Indexed: 10/13/2023] Open
Abstract
Peroxisome proliferator-activated receptor PPARγ coactivator-α (PGC-1α) is concentrated in inhibitory interneurons and plays a vital role in neuropsychiatric diseases. We previously reported some characteristic features of schizophrenia (SZ) in GABAergic neuron-specific Pgc-1alpha knockout (KO) mice (Dlx5/6-Cre: Pgc-1alphaf/f). However, there is a fundamental gap in the molecular mechanism by which the Pgc-1alpha gene is involved in the neurobehavioral abnormalities of SZ. The loss of critical period (CP) triggers-maturations of parvalbumin interneurons (PVIs) and brakes-and the formation of perineuronal nets (PNNs) implicates mistimed trajectories during adult brain development. In this study, using the Pgc-1alpha KO mouse line, we investigated the association of Pgc-1alpha gene deletion with SZ-like behavioral deficits, PVI maturation, PNN integrity and synaptic ultrastructure. These findings suggest that Pgc-1alpha gene deletion resulted in a failure of CP onset and closure, thereby prolonging cortical plasticity timing. To determine whether the manipulation of the PNN structure is a potential method of altering neuronal plasticity, GM6001, a broad-spectrum matrix metalloproteinase (MMP)-inhibitor was applied. Here we confirmed that the treatment could effectively correct the CP plasticity window and ameliorate the synaptic ultrastructure in the Pgc-1alpha KO brain. Moreover, the intervention effect on neuronal plasticity was followed by the rescue of short-term habituation deficits and the mitigation of aberrant salience, which are some characteristic features of SZ. Taken collectively, these findings suggest that the role of PGC-1α in regulating cortical plasticity is mediated, at least partially, through the regulation of CP onset/closure. Strategically introduced reinforcement of molecular brakes may be a novel preventive therapy for psychiatric disorders associated with PGC-1α dysregulation.
Collapse
Affiliation(s)
- Wei-Jun Zhang
- The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Hou-Zhen Shi
- The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Mei-Na Guo
- The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Long-Fei Xu
- The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Hong-Ru Zhai
- The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Zi-Zhong Liu
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yong-Qiang Zhu
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Wei-Ning Zhang
- The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Jia Wang
- The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
- Zhenjiang Jieshengrui Biotechnology Co., Ltd., Zhenjiang, Jiangsu, China
| |
Collapse
|
15
|
Hughes BW, Siemsen BM, Tsvetkov E, Berto S, Kumar J, Cornbrooks RG, Akiki RM, Cho JY, Carter JS, Snyder KK, Assali A, Scofield MD, Cowan CW, Taniguchi M. NPAS4 in the medial prefrontal cortex mediates chronic social defeat stress-induced anhedonia-like behavior and reductions in excitatory synapses. eLife 2023; 12:e75631. [PMID: 36780219 PMCID: PMC9925055 DOI: 10.7554/elife.75631] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/29/2023] [Indexed: 02/14/2023] Open
Abstract
Chronic stress can produce reward system deficits (i.e., anhedonia) and other common symptoms associated with depressive disorders, as well as neural circuit hypofunction in the medial prefrontal cortex (mPFC). However, the molecular mechanisms by which chronic stress promotes depressive-like behavior and hypofrontality remain unclear. We show here that the neuronal activity-regulated transcription factor, NPAS4, in the mPFC is regulated by chronic social defeat stress (CSDS), and it is required in this brain region for CSDS-induced changes in sucrose preference and natural reward motivation in the mice. Interestingly, NPAS4 is not required for CSDS-induced social avoidance or anxiety-like behavior. We also find that mPFC NPAS4 is required for CSDS-induced reductions in pyramidal neuron dendritic spine density, excitatory synaptic transmission, and presynaptic function, revealing a relationship between perturbation in excitatory synaptic transmission and the expression of anhedonia-like behavior in the mice. Finally, analysis of the mice mPFC tissues revealed that NPAS4 regulates the expression of numerous genes linked to glutamatergic synapses and ribosomal function, the expression of upregulated genes in CSDS-susceptible animals, and differentially expressed genes in postmortem human brains of patients with common neuropsychiatric disorders, including depression. Together, our findings position NPAS4 as a key mediator of chronic stress-induced hypofrontal states and anhedonia-like behavior.
Collapse
Affiliation(s)
- Brandon W Hughes
- Department of Neuroscience, Medical University of South CarolinaCharlestonUnited States
| | - Benjamin M Siemsen
- Department of Neuroscience, Medical University of South CarolinaCharlestonUnited States
- Department of Anesthesiology, Medical University of South CarolinaCharlestonUnited States
| | - Evgeny Tsvetkov
- Department of Neuroscience, Medical University of South CarolinaCharlestonUnited States
| | - Stefano Berto
- Department of Neuroscience, Medical University of South CarolinaCharlestonUnited States
| | - Jaswinder Kumar
- Department of Psychiatry, Harvard Medical SchoolBelmontUnited States
- Neuroscience Graduate Program, University of Texas Southwestern Medical CenterDallasUnited States
| | - Rebecca G Cornbrooks
- Department of Neuroscience, Medical University of South CarolinaCharlestonUnited States
| | - Rose Marie Akiki
- Department of Neuroscience, Medical University of South CarolinaCharlestonUnited States
| | - Jennifer Y Cho
- Department of Neuroscience, Medical University of South CarolinaCharlestonUnited States
| | - Jordan S Carter
- Department of Neuroscience, Medical University of South CarolinaCharlestonUnited States
| | - Kirsten K Snyder
- Department of Neuroscience, Medical University of South CarolinaCharlestonUnited States
| | - Ahlem Assali
- Department of Neuroscience, Medical University of South CarolinaCharlestonUnited States
| | - Michael D Scofield
- Department of Neuroscience, Medical University of South CarolinaCharlestonUnited States
- Department of Anesthesiology, Medical University of South CarolinaCharlestonUnited States
| | - Christopher W Cowan
- Department of Neuroscience, Medical University of South CarolinaCharlestonUnited States
- Department of Psychiatry, Harvard Medical SchoolBelmontUnited States
- Neuroscience Graduate Program, University of Texas Southwestern Medical CenterDallasUnited States
| | - Makoto Taniguchi
- Department of Neuroscience, Medical University of South CarolinaCharlestonUnited States
- Department of Psychiatry, Harvard Medical SchoolBelmontUnited States
| |
Collapse
|
16
|
Omeiza NA, Bakre A, Ben-Azu B, Sowunmi AA, Abdulrahim HA, Chimezie J, Lawal SO, Adebayo OG, Alagbonsi AI, Akinola O, Abolaji AO, Aderibigbe AO. Mechanisms underpinning Carpolobia lutea G. Don ethanol extract's neurorestorative and antipsychotic-like activities in an NMDA receptor antagonist model of schizophrenia. JOURNAL OF ETHNOPHARMACOLOGY 2023; 301:115767. [PMID: 36206872 DOI: 10.1016/j.jep.2022.115767] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 09/11/2022] [Accepted: 09/25/2022] [Indexed: 06/16/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Persistent ketamine insults to the central nervous system block NMDA receptors and disrupt putative neurotransmission, oxido-nitrosative, and inflammatory pathways, resulting in schizophrenia-like symptoms in animals. Previously, the ethnomedicinal benefits of Carpolobia lutea against insomnia, migraine headache, and insanity has been documented, but the mechanisms of action remain incomplete. AIM OF THE STUDY Presently, we explored the neuro-therapeutic role of Carpolobia lutea ethanol extract (C. lutea) in ketamine-induced schizophrenia-like symptoms in mice. MATERIALS AND METHODS Sixty-four male Swiss (22 ± 2 g) mice were randomly assigned into eight groups (n = 8/group) and exposed to a reversal ketamine model of schizophrenia. For 14 days, either distilled water (10 mL/kg; p.o.) or ketamine (20 mg/kg; i.p.) was administered, following possible reversal treatments with C. lutea (100, 200, 400, and 800 mg/kg; p.o.), haloperidol (1 mg/kg, p.o.), or clozapine (5 mg/kg; p.o.) beginning on days 8-14. During the experiment, a battery of behavioral characterizations defining schizophrenia-like symptoms were obtained using ANY-maze software, followed by neurochemical, oxido-inflammatory and histological assessments in the mice brains. RESULTS A 7-day reversal treatment with C. lutea reversed predictors of positive, negative and cognitive symptoms of schizophrenia. C. lutea also mitigated ketamine-induced neurochemical derangements as evidenced by modulations of dopamine, glutamate, norepinephrine and serotonin neurotransmission. Also, the increased acetylcholinesterase activity, malondialdehyde nitrite, interleukin-6 and tumor necrosis-factor-α concentrations were reversed by C. lutea accompanied with elevated levels of catalase, superoxide dismutase and reduced glutathione. Furthermore, C. lutea reversed ketamine-induced neuronal alterations in the prefrontal cortex, hippocampus and cerebellum sections of the brain. CONCLUSION These findings suggest that C. lutea reverses the cardinal symptoms of ketamine-induced schizophrenia in a dose-dependent fashion by modulating the oxido-inflammatory and neurotransmitter-related mechanisms.
Collapse
Affiliation(s)
- Noah A Omeiza
- Department of Pharmacology and Therapeutics, Neuropharmacology Unit, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria.
| | - Adewale Bakre
- Department of Pharmacology and Therapeutics, Neuropharmacology Unit, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Benneth Ben-Azu
- Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Nigeria
| | - Abimbola A Sowunmi
- Department of Pharmacology and Therapeutics, Neuropharmacology Unit, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Halimat A Abdulrahim
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Joseph Chimezie
- Department of Physiology, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Sodiq O Lawal
- Department of Physiology, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Olusegun G Adebayo
- Department of Physiology, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria; Department of Physiology, Neurophysiology Unit, Faculty of Basic Medical Sciences, PAMO University of Medical Sciences, Port-Harcourt, Nigeria
| | - Abdullateef I Alagbonsi
- Department of Clinical Biology (Physiology), School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Huye, Southern Province, Rwanda
| | - Olugbenga Akinola
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, College of Medicine, University of Ibadan, Nigeria
| | - Amos O Abolaji
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Adegbuyi O Aderibigbe
- Department of Pharmacology and Therapeutics, Neuropharmacology Unit, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
17
|
Sibilska S, Mofleh R, Kocsis B. Development of network oscillations through adolescence in male and female rats. Front Cell Neurosci 2023; 17:1135154. [PMID: 37213214 PMCID: PMC10196069 DOI: 10.3389/fncel.2023.1135154] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 04/19/2023] [Indexed: 05/23/2023] Open
Abstract
The primary aim of this research was to study the developmental trajectory of oscillatory synchronization in neural networks of normal healthy rats during adolescence, corresponding to the vulnerable age of schizophrenia prodrome in human. To monitor the development of oscillatory networks through adolescence we used a "pseudo-longitudinal" design. Recordings were performed in terminal experiments under urethane anesthesia, every day from PN32 to PN52 using rats-siblings from the same mother, to reduce individual innate differences between subjects. We found that hippocampal theta power decreased and delta power in prefrontal cortex increased through adolescence, indicating that the oscillations in the two different frequency bands follow distinct developmental trajectories to reach the characteristic oscillatory activity found in adults. Perhaps even more importantly, theta rhythm showed age-dependent stabilization toward late adolescence. Furthermore, sex differences was found in both networks, more prominent in the prefrontal cortex compared with hippocampus. Delta increase was stronger in females and theta stabilization was completed earlier in females, in postnatal days PN41-47, while in males it was only completed in late adolescence. Our finding of a protracted maturation of theta-generating networks in late adolescence is overall consistent with the findings of longitudinal studies in human adolescents, in which oscillatory networks demonstrated a similar pattern of maturation.
Collapse
|
18
|
Synaptic plasticity and mental health: methods, challenges and opportunities. Neuropsychopharmacology 2023; 48:113-120. [PMID: 35810199 PMCID: PMC9700665 DOI: 10.1038/s41386-022-01370-w] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/13/2022] [Accepted: 06/20/2022] [Indexed: 12/14/2022]
Abstract
Activity-dependent synaptic plasticity is a ubiquitous property of the nervous system that allows neurons to communicate and change their connections as a function of past experiences. Through reweighting of synaptic strengths, the nervous system can remodel itself, giving rise to durable memories that create the biological basis for mental function. In healthy individuals, synaptic plasticity undergoes characteristic developmental and aging trajectories. Dysfunctional plasticity, in turn, underlies a wide spectrum of neuropsychiatric disorders including depression, schizophrenia, addiction, and posttraumatic stress disorder. From a mechanistic standpoint, synaptic plasticity spans the gamut of spatial and temporal scales, from microseconds to the lifespan, from microns to the entire nervous system. With the numbers and strengths of synapses changing on such wide scales, there is an important need to develop measurement techniques with complimentary sensitivities and a growing number of approaches are now being harnessed for this purpose. Through hemodynamic measures, structural and tracer imaging, and noninvasive neuromodulation, it is possible to image structural and functional changes that underlie synaptic plasticity and associated behavioral learning. Here we review the mechanisms of neural plasticity and the historical and future trends in techniques that allow imaging of synaptic changes that accompany psychiatric disorders, highlighting emerging therapeutics and the challenges and opportunities accompanying this burgeoning area of study.
Collapse
|
19
|
Renner J, Rasia-Filho AA. Morphological Features of Human Dendritic Spines. ADVANCES IN NEUROBIOLOGY 2023; 34:367-496. [PMID: 37962801 DOI: 10.1007/978-3-031-36159-3_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Dendritic spine features in human neurons follow the up-to-date knowledge presented in the previous chapters of this book. Human dendrites are notable for their heterogeneity in branching patterns and spatial distribution. These data relate to circuits and specialized functions. Spines enhance neuronal connectivity, modulate and integrate synaptic inputs, and provide additional plastic functions to microcircuits and large-scale networks. Spines present a continuum of shapes and sizes, whose number and distribution along the dendritic length are diverse in neurons and different areas. Indeed, human neurons vary from aspiny or "relatively aspiny" cells to neurons covered with a high density of intermingled pleomorphic spines on very long dendrites. In this chapter, we discuss the phylogenetic and ontogenetic development of human spines and describe the heterogeneous features of human spiny neurons along the spinal cord, brainstem, cerebellum, thalamus, basal ganglia, amygdala, hippocampal regions, and neocortical areas. Three-dimensional reconstructions of Golgi-impregnated dendritic spines and data from fluorescence microscopy are reviewed with ultrastructural findings to address the complex possibilities for synaptic processing and integration in humans. Pathological changes are also presented, for example, in Alzheimer's disease and schizophrenia. Basic morphological data can be linked to current techniques, and perspectives in this research field include the characterization of spines in human neurons with specific transcriptome features, molecular classification of cellular diversity, and electrophysiological identification of coexisting subpopulations of cells. These data would enlighten how cellular attributes determine neuron type-specific connectivity and brain wiring for our diverse aptitudes and behavior.
Collapse
Affiliation(s)
- Josué Renner
- Department of Basic Sciences/Physiology and Graduate Program in Biosciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
| | - Alberto A Rasia-Filho
- Department of Basic Sciences/Physiology and Graduate Program in Biosciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
- Graduate Program in Neuroscience, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
20
|
Kubota M, Takahata K, Matsuoka K, Sano Y, Yamamoto Y, Tagai K, Tarumi R, Suzuki H, Kurose S, Nakajima S, Shiwaku H, Seki C, Kawamura K, Zhang MR, Takahashi H, Takado Y, Higuchi M. Positron Emission Tomography Assessments of Phosphodiesterase 10A in Patients With Schizophrenia. Schizophr Bull 2022; 49:688-696. [PMID: 36458958 PMCID: PMC10154699 DOI: 10.1093/schbul/sbac181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
BACKGROUND AND HYPOTHESIS Phosphodiesterase 10A (PDE10A) is a highly expressed enzyme in the basal ganglia, where cortical glutamatergic and midbrain dopaminergic inputs are integrated. Therapeutic PDE10A inhibition effects on schizophrenia have been reported previously, but the status of this molecule in the living patients with schizophrenia remains elusive. Therefore, this study aimed to investigate the central PDE10A status in patients with schizophrenia and examine its relationship with psychopathology, cognition, and corticostriatal glutamate levels. STUDY DESIGN This study included 27 patients with schizophrenia, with 5 antipsychotic-free cases, and 27 healthy controls. Positron emission tomography with [18F]MNI-659, a specific PDE10A radioligand, was employed to quantify PDE10A availability by measuring non-displaceable binding potential (BPND) of the ligand in the limbic, executive, and sensorimotor striatal functional subregions, and in the pallidum. BPND estimates were compared between patients and controls while controlling for age and gender. BPND correlations were examined with behavioral and clinical measures, along with regional glutamate levels quantified by the magnetic resonance spectroscopy. STUDY RESULTS Multivariate analysis of covariance demonstrated a significant main effect of diagnosis on BPND (p = .03). A posthoc test showed a trend-level higher sensorimotor striatal BPND in patients, although it did not survive multiple comparison corrections. BPND in controls in this subregion was significantly and negatively correlated with the Tower of London scores, a cognitive subtest. Striatal or dorsolateral prefrontal glutamate levels did not correlate significantly with BPND in either group. CONCLUSIONS The results suggest altered striatal PDE10A availability and associated local neural dysfunctions in patients with schizophrenia.
Collapse
Affiliation(s)
- Manabu Kubota
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Inage-ku, Chiba, Japan.,Department of Psychiatry, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
| | - Keisuke Takahata
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Inage-ku, Chiba, Japan.,Department of Neuropsychiatry, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Kiwamu Matsuoka
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Inage-ku, Chiba, Japan
| | - Yasunori Sano
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Inage-ku, Chiba, Japan.,Department of Neuropsychiatry, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Yasuharu Yamamoto
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Inage-ku, Chiba, Japan.,Department of Neuropsychiatry, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Kenji Tagai
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Inage-ku, Chiba, Japan.,Department of Psychiatry, The Jikei University Graduate School of Medicine, Minato-ku, Tokyo, Japan
| | - Ryosuke Tarumi
- Department of Neuropsychiatry, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Hisaomi Suzuki
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Inage-ku, Chiba, Japan.,National Hospital Organization Shimofusa Psychiatric Medical Center, Midori-ku, Chiba, Japan
| | - Shin Kurose
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Inage-ku, Chiba, Japan.,Department of Neuropsychiatry, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Shinichiro Nakajima
- Department of Neuropsychiatry, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Hiroki Shiwaku
- Department of Psychiatry and Behavioral Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Chie Seki
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Inage-ku, Chiba, Japan
| | - Kazunori Kawamura
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Inage-ku, Chiba, Japan
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Inage-ku, Chiba, Japan
| | - Hidehiko Takahashi
- Department of Psychiatry and Behavioral Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Yuhei Takado
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Inage-ku, Chiba, Japan
| | - Makoto Higuchi
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Inage-ku, Chiba, Japan
| |
Collapse
|
21
|
Peters KZ, Naneix F. The role of dopamine and endocannabinoid systems in prefrontal cortex development: Adolescence as a critical period. Front Neural Circuits 2022; 16:939235. [PMID: 36389180 PMCID: PMC9663658 DOI: 10.3389/fncir.2022.939235] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 10/14/2022] [Indexed: 01/07/2023] Open
Abstract
The prefrontal cortex plays a central role in the control of complex cognitive processes including action control and decision making. It also shows a specific pattern of delayed maturation related to unique behavioral changes during adolescence and allows the development of adult cognitive processes. The adolescent brain is extremely plastic and critically vulnerable to external insults. Related to this vulnerability, adolescence is also associated with the emergence of numerous neuropsychiatric disorders involving alterations of prefrontal functions. Within prefrontal microcircuits, the dopamine and the endocannabinoid systems have widespread effects on adolescent-specific ontogenetic processes. In this review, we highlight recent advances in our understanding of the maturation of the dopamine system and the endocannabinoid system in the prefrontal cortex during adolescence. We discuss how they interact with GABA and glutamate neurons to modulate prefrontal circuits and how they can be altered by different environmental events leading to long-term neurobiological and behavioral changes at adulthood. Finally, we aim to identify several future research directions to help highlight gaps in our current knowledge on the maturation of these microcircuits.
Collapse
Affiliation(s)
- Kate Zara Peters
- Sussex Neuroscience, School of Psychology, University of Sussex, Falmer, United Kingdom
| | - Fabien Naneix
- The Rowett Institute, University of Aberdeen, Aberdeen, United Kingdom,*Correspondence: Fabien Naneix
| |
Collapse
|
22
|
Liu X, Hua F, Yang D, Lin Y, Zhang L, Ying J, Sheng H, Wang X. Roles of neuroligins in central nervous system development: focus on glial neuroligins and neuron neuroligins. Lab Invest 2022; 20:418. [PMID: 36088343 PMCID: PMC9463862 DOI: 10.1186/s12967-022-03625-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/01/2022] [Indexed: 11/10/2022]
Abstract
Neuroligins are postsynaptic cell adhesion molecules that are relevant to many neurodevelopmental disorders. They are differentially enriched at the postsynapse and interact with their presynaptic ligands, neurexins, whose differential binding to neuroligins has been shown to regulate synaptogenesis, transmission, and other synaptic properties. The proper functioning of functional networks in the brain depends on the proper connection between neuronal synapses. Impaired synaptogenesis or synaptic transmission results in synaptic dysfunction, and these synaptic pathologies are the basis for many neurodevelopmental disorders. Deletions or mutations in the neuroligins genes have been found in patients with both autism and schizophrenia. It is because of the important role of neuroligins in synaptic connectivity and synaptic dysfunction that studies on neuroligins in the past have mainly focused on their expression in neurons. As studies on the expression of genes specific to various cells of the central nervous system deepened, neuroligins were found to be expressed in non-neuronal cells as well. In the central nervous system, glial cells are the most representative non-neuronal cells, which can also express neuroligins in large amounts, especially astrocytes and oligodendrocytes, and they are involved in the regulation of synaptic function, as are neuronal neuroligins. This review examines the mechanisms of neuron neuroligins and non-neuronal neuroligins in the central nervous system and also discusses the important role of neuroligins in the development of the central nervous system and neurodevelopmental disorders from the perspective of neuronal neuroligins and glial neuroligins.
Collapse
|
23
|
Zhang W, Chen Y, Qin J, Lu J, Fan Y, Shi Z, Song X, Li C, Zhao T. Prolonged sevoflurane exposure causes abnormal synapse development and dysregulates beta-neurexin and neuroligins in the hippocampus in neonatal rats. J Affect Disord 2022; 312:22-29. [PMID: 35691415 DOI: 10.1016/j.jad.2022.05.115] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 05/17/2022] [Accepted: 05/20/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND The underlying molecular mechanisms of the excitatory/inhibitory (E/I) imbalance induced by sevoflurane exposure to neonates remain poorly understood. This study aimed to investigate the long-term effects of prolonged sevoflurane exposure to neonatal rats during the peak period of synaptogenesis on the changes of trans-synaptic neurexin-neuroligin interactions, synaptic ultrastructure in the hippocampus and cognition. METHODS A total of 30 rat pups at postnatal day (P) 7 was randomly divided into two groups: the control group (exposed to 30 % oxygen balanced with nitrogen) and the sevoflurane group (exposed to 2.5 % sevoflurane plus 30 % oxygen balanced with nitrogen) for 6 h. Neurocognitive behaviors were assessed with the Open field test at P23-25 and the Morris water maze test at P26-30. The expression of β-neurexin (β-NRX), N-methyl-d-aspartate receptor 2 subunit (NR2A and NR2B), neuroligin-1 (NLG-1), neuroligin-2 (NLG-2), postsynaptic density protein-95 (PSD-95), α1-subunit of the γ-aminobutyric acid A receptor (GABAAα1) and gephyrin in the hippocampus at P30 were measured by Western blot. The ultrastructure of synapses was examined under electron microscope. RESULTS Prolonged sevoflurane exposure at P7 resulted in cognitive deficiency in adolescence, as well as the downregulation of β-NRX, NR2A, NR2B, NLG-1, and PSD-95, and the upregulation of GABAAα1, NLG-2, and gephyrin in the hippocampal CA3 region. Sevoflurane anesthesia also increased the number of symmetric synapses in the hippocampus. CONCLUSIONS Prolonged sevoflurane exposure during the brain development leads to cognitive deficiency and disproportion of excitatory/inhibitory synapses which may be caused by dysregulated expression of synaptic adhesion molecules of β-NRX and neuroligins.
Collapse
Affiliation(s)
- Wenhua Zhang
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China; Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Yanxin Chen
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China; Department of Anesthesiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510623, China
| | - Jingwen Qin
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Junming Lu
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China; Department of Anesthesiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510623, China
| | - Yanting Fan
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Ziwen Shi
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Xingrong Song
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Chuanxiang Li
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China; Department of Anesthesiology, Pinghu Hospital of Shenzhen University, Shenzhen 518111, China.
| | - Tianyun Zhao
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China.
| |
Collapse
|
24
|
Sarpal DK, Tarcijonas G, Calabro FJ, Foran W, Haas GL, Luna B, Murty VP. Context-specific abnormalities of the central executive network in first-episode psychosis: relationship with cognition. Psychol Med 2022; 52:2299-2308. [PMID: 33222723 PMCID: PMC9805803 DOI: 10.1017/s0033291720004201] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Cognitive impairments, which contribute to the profound functional deficits observed in psychotic disorders, have found to be associated with abnormalities in trial-level cognitive control. However, neural tasks operate within the context of sustained cognitive states, which can be assessed with 'background connectivity' following the removal of task effects. To date, little is known about the integrity of brain processes supporting the maintenance of a cognitive state in individuals with psychotic disorders. Thus, here we examine background connectivity during executive processing in a cohort of participants with first-episode psychosis (FEP). METHODS The following fMRI study examined background connectivity of the dorsolateral prefrontal cortex (DLPFC), during working memory engagement in a group of 43 patients with FEP, relative to 35 healthy controls (HC). Findings were also examined in relation to measures of executive function. RESULTS The FEP group relative to HC showed significantly lower background DLPFC connectivity with bilateral superior parietal lobule (SPL) and left inferior parietal lobule. Background connectivity between DLPFC and SPL was also positively associated with overall cognition across all subjects and in our FEP group. In comparison, resting-state frontoparietal connectivity did not differ between groups and was not significantly associated with overall cognition, suggesting that psychosis-related alterations in executive networks only emerged during states of goal-oriented behavior. CONCLUSIONS These results provide novel evidence indicating while frontoparietal connectivity at rest appears intact in psychosis, when engaged during a cognitive state, it is impaired possibly undermining cognitive control capacities in FEP.
Collapse
Affiliation(s)
- Deepak K. Sarpal
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Goda Tarcijonas
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Finnegan J. Calabro
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - William Foran
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Gretchen L. Haas
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Beatriz Luna
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Vishnu P. Murty
- Department of Psychology, Temple University, Philadelphia, PA, USA
| |
Collapse
|
25
|
Kozlova A, Zhang S, Kotlar AV, Jamison B, Zhang H, Shi S, Forrest MP, McDaid J, Cutler DJ, Epstein MP, Zwick ME, Pang ZP, Sanders AR, Warren ST, Gejman PV, Mulle JG, Duan J. Loss of function of OTUD7A in the schizophrenia- associated 15q13.3 deletion impairs synapse development and function in human neurons. Am J Hum Genet 2022; 109:1500-1519. [PMID: 35931052 PMCID: PMC9388388 DOI: 10.1016/j.ajhg.2022.07.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 06/27/2022] [Indexed: 02/06/2023] Open
Abstract
Identifying causative gene(s) within disease-associated large genomic regions of copy-number variants (CNVs) is challenging. Here, by targeted sequencing of genes within schizophrenia (SZ)-associated CNVs in 1,779 SZ cases and 1,418 controls, we identified three rare putative loss-of-function (LoF) mutations in OTU deubiquitinase 7A (OTUD7A) within the 15q13.3 deletion in cases but none in controls. To tie OTUD7A LoF with any SZ-relevant cellular phenotypes, we modeled the OTUD7A LoF mutation, rs757148409, in human induced pluripotent stem cell (hiPSC)-derived induced excitatory neurons (iNs) by CRISPR-Cas9 engineering. The mutant iNs showed a ∼50% decrease in OTUD7A expression without undergoing nonsense-mediated mRNA decay. The mutant iNs also exhibited marked reduction of dendritic complexity, density of synaptic proteins GluA1 and PSD-95, and neuronal network activity. Congruent with the neuronal phenotypes in mutant iNs, our transcriptomic analysis showed that the set of OTUD7A LoF-downregulated genes was enriched for those relating to synapse development and function and was associated with SZ and other neuropsychiatric disorders. These results suggest that OTUD7A LoF impairs synapse development and neuronal function in human neurons, providing mechanistic insight into the possible role of OTUD7A in driving neuropsychiatric phenotypes associated with the 15q13.3 deletion.
Collapse
Affiliation(s)
- Alena Kozlova
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA
| | - Siwei Zhang
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA; Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL 60637, USA
| | - Alex V Kotlar
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA; Pillar Biosciences Inc., Natick, MA 01760, USA
| | - Brendan Jamison
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA
| | - Hanwen Zhang
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA
| | - Serena Shi
- Winston Churchill High School, Potomac, MD 20854, USA
| | - Marc P Forrest
- Department of Neuroscience, Northwestern University, Chicago, IL 60611, USA; Center for Autism and Neurodevelopment, Northwestern University, Chicago, IL 60611, USA
| | - John McDaid
- Department of Neurosurgery, NorthShore University HealthSystem, Evanston, IL 60201, USA
| | - David J Cutler
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Michael P Epstein
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Michael E Zwick
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA; Senior Vice President for Research, Rutgers University, New Brunswick, NJ 08901, USA
| | - Zhiping P Pang
- Department of Neuroscience and Cell Biology, Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Alan R Sanders
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA; Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL 60637, USA
| | - Stephen T Warren
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Pablo V Gejman
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA; Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL 60637, USA
| | - Jennifer G Mulle
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA; Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA
| | - Jubao Duan
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA; Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
26
|
Cardoso-Cruz H, Laranjeira I, Monteiro C, Galhardo V. Altered prefrontal-striatal theta-band oscillatory dynamics underlie working memory deficits in neuropathic pain rats. Eur J Pain 2022; 26:1546-1568. [PMID: 35603472 DOI: 10.1002/ejp.1982] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 04/21/2022] [Accepted: 05/16/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND Prelimbic medial prefrontal cortex (PL-mPFC) and nucleus accumbens core region (NAcc) play an important role in supporting several executive cognitive mechanisms, such as spatial working-memory (WM). Recently, this circuit has been also associated with both sensory and affective components of pain. However, it is still unclear whether this circuit is endogenously engaged in neuropathic pain-related cognitive dysfunctions. METHODS To answer this question, we induced the expression of halorhodopsin in local PL-mPFC neurons projecting to NAcc, and then selectively inhibited the terminals of these neurons in the NAcc while recording neural activity during the performance of a delayed non-match to sample (DNMS) spatial WM task. Within-subject behavioral performance and PL-mPFC to NAcc circuit neural activity was assessed after the onset of a persistent rodent neuropathic pain model - spared nerve injury (SNI). RESULTS Our results revealed that the induction of the neuropathy reduced WM performance, and altered the interplay between PL-mPFC and NAcc neurons namely in increasing the functional connectivity from NAcc to PL-mPFC, particularly in the theta-band spontaneous oscillations; in addition, these behavioral and functional perturbations were partially reversed by selective optogenetic inhibition of PL-mPFC neuron terminals into the NAcc during the DNMS task delay-period, without significant antinociceptive effects. CONCLUSIONS Altogether, these results strongly suggest that the PL-mPFC excitatory output into the NAcc plays an important role in the deregulation of WM under pain conditions. SIGNIFICANCE Selective optogenetic inhibition of prefrontal-striatal microcircuit reverses pain-related working memory deficits, but has no significant impact on pain responses. Neuropathic pain underlies an increase of functional connectivity between the nucleus accumbens core area and the prelimbic medial prefrontal cortex mediated by theta-band activity.
Collapse
Affiliation(s)
- Helder Cardoso-Cruz
- Instituto de Investigação e Inovação em Saúde (i3S), Pain Neurobiology Group; Universidade do Porto, 4200-135, Porto, Portugal.,Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, 4200-135, Porto, Portugal.,Faculdade de Medicina (FMUP), Departamento de Biomedicina - Unidade de Biologia Experimental; Universidade do Porto, 4200-319, Porto, Portugal
| | - Inês Laranjeira
- Instituto de Investigação e Inovação em Saúde (i3S), Pain Neurobiology Group; Universidade do Porto, 4200-135, Porto, Portugal.,Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, 4200-135, Porto, Portugal.,Faculdade de Medicina (FMUP), Departamento de Biomedicina - Unidade de Biologia Experimental; Universidade do Porto, 4200-319, Porto, Portugal.,Mestrado em Neurobiologia da Faculdade de Medicina da Universidade do Porto. 4200-319, Porto, Portugal
| | - Clara Monteiro
- Instituto de Investigação e Inovação em Saúde (i3S), Pain Neurobiology Group; Universidade do Porto, 4200-135, Porto, Portugal.,Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, 4200-135, Porto, Portugal.,Faculdade de Medicina (FMUP), Departamento de Biomedicina - Unidade de Biologia Experimental; Universidade do Porto, 4200-319, Porto, Portugal
| | - Vasco Galhardo
- Instituto de Investigação e Inovação em Saúde (i3S), Pain Neurobiology Group; Universidade do Porto, 4200-135, Porto, Portugal.,Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, 4200-135, Porto, Portugal.,Faculdade de Medicina (FMUP), Departamento de Biomedicina - Unidade de Biologia Experimental; Universidade do Porto, 4200-319, Porto, Portugal
| |
Collapse
|
27
|
Sarpal DK, Blazer A, Wilson JD, Calabro FJ, Foran W, Kahn CE, Luna B, Chengappa KNR. Relationship between plasma clozapine/N-desmethylclozapine and changes in basal forebrain-dorsolateral prefrontal cortex coupling in treatment-resistant schizophrenia. Schizophr Res 2022; 243:170-177. [PMID: 35381515 PMCID: PMC9189030 DOI: 10.1016/j.schres.2022.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 03/07/2022] [Accepted: 03/27/2022] [Indexed: 10/18/2022]
Abstract
Clozapine (CLZ) demonstrates a unique clinical efficacy relative to other antipsychotic drugs. Previous work has linked the plasma ratio of CLZ and its major metabolite, N-desmethylclozapine (NDMC), to an inverse relationship with cognition via putative action on the cholinergic system. However, neuroimaging correlates of CLZ/NDMC remain unknown. Here, we examined changes in basal forebrain functional connectivity with the dorsolateral prefrontal cortex, and secondly, cognition in relation to the CLZ/NDMC ratio. A cohort of nineteen chronically ill participants with treatment-resistant schizophrenia (TRS) undergoing 12 weeks of CLZ treatment were included. Measures of cognition and plasma CLZ/NDMC ratios were obtained in addition to resting-state functional neuroimaging scans, captured at baseline and after 12 weeks of CLZ treatment. We observed a significant correlation between basal forebrain-DLPFC connectivity and CLZ/NDMC ratios across CLZ treatment (p = 0.02). Consistent with previous findings, we also demonstrate a positive relationship between CLZ/NDMC ratio and working memory (p = 0.03). These findings may reflect the action of CLZ and NDMC on the muscarinic cholinergic system, highlighting a possible neural correlate of cognition across treatment.
Collapse
Affiliation(s)
- Deepak K. Sarpal
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Annie Blazer
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - James D. Wilson
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Finnegan J. Calabro
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA,Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - William Foran
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Charles E. Kahn
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Beatriz Luna
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA,Department of Psychology, University of Pittsburgh, Pittsburgh, PA, USA,Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA
| | - KN Roy Chengappa
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
28
|
Myint AM, Halaris A. Imbalances in Kynurenines as Potential Biomarkers in the Diagnosis and Treatment of Psychiatric Disorders. Front Psychiatry 2022; 13:913303. [PMID: 35836656 PMCID: PMC9275364 DOI: 10.3389/fpsyt.2022.913303] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 05/27/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Aye-Mu Myint
- Department of Psychiatry, Loyola University School of Medicine and Loyola University Medical Center, Maywood, IL, United States
| | - Angelos Halaris
- Department of Psychiatry, Loyola University School of Medicine and Loyola University Medical Center, Maywood, IL, United States
| |
Collapse
|
29
|
Vafadari B. Stress and the Role of the Gut-Brain Axis in the Pathogenesis of Schizophrenia: A Literature Review. Int J Mol Sci 2021; 22:ijms22189747. [PMID: 34575911 PMCID: PMC8471971 DOI: 10.3390/ijms22189747] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/19/2021] [Accepted: 08/31/2021] [Indexed: 12/21/2022] Open
Abstract
Schizophrenia is a severe neuropsychiatric disorder, and its etiology remains largely unknown. Environmental factors have been reported to play roles in the pathogenesis of schizophrenia, and one of the major environmental factors identified for this disorder is psychosocial stress. Several studies have suggested that stressful life events, as well as the chronic social stress associated with city life, may lead to the development of schizophrenia. The other factor is the gut–brain axis. The composition of the gut microbiome and alterations thereof may affect the brain and may lead to schizophrenia. The main interest of this review article is in overviewing the major recent findings on the effects of stress and the gut–brain axis, as well as their possible bidirectional effects, in the pathogenesis of schizophrenia.
Collapse
Affiliation(s)
- Behnam Vafadari
- Clinic for Anesthesiology, University Medical Center Göttingen, Georg-August-University, 37073 Göttingen, Germany
| |
Collapse
|
30
|
Peters KZ, Zlebnik NE, Cheer JF. Cannabis exposure during adolescence: A uniquely sensitive period for neurobiological effects. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 161:95-120. [PMID: 34801175 DOI: 10.1016/bs.irn.2021.07.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Adolescence is a crucial developmental period where neural circuits are refined and the brain is especially vulnerable to external insults. The endocannabinoid (eCB) system undergoes changes during adolescence which affect the way in which it modulates the development of other systems, in particular dopamine circuits, which show protracted development into adolescence. Given the rise of cannabis use by adolescents and young people, as well as variants containing increasingly higher concentrations of THC, it is now crucial to understand the unique effects of adolescent exposure to cannabis on the developing brain and it might shape future adult vulnerabilities to conditions such as psychosis, schizophrenia, addiction and more. Here we discuss the development of the eCB system across the lifespan, how CB1 receptors modulate dopamine release and potential neurobiological and behavioral effects of adolescent THC exposure on the developing brain such as alterations in excitatory/inhibitory balance during this developmental period.
Collapse
Affiliation(s)
- K Z Peters
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States; Sussex Neuroscience, School of Psychology, University of Sussex, Falmer, United Kingdom.
| | - N E Zlebnik
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - J F Cheer
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States; Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States; Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
31
|
Fish KN, Rocco BR, DeDionisio AM, Dienel SJ, Sweet RA, Lewis DA. Altered Parvalbumin Basket Cell Terminals in the Cortical Visuospatial Working Memory Network in Schizophrenia. Biol Psychiatry 2021; 90:47-57. [PMID: 33892915 PMCID: PMC8243491 DOI: 10.1016/j.biopsych.2021.02.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 01/21/2021] [Accepted: 02/11/2021] [Indexed: 12/28/2022]
Abstract
BACKGROUND Visuospatial working memory (vsWM), which is commonly impaired in schizophrenia, involves information processing across the primary visual cortex, association visual cortex, posterior parietal cortex, and dorsolateral prefrontal cortex (DLPFC). Within these regions, vsWM requires inhibition from parvalbumin-expressing basket cells (PVBCs). Here, we analyzed indices of PVBC axon terminals across regions of the vsWM network in schizophrenia. METHODS For 20 matched pairs of subjects with schizophrenia and unaffected comparison subjects, tissue sections from the primary visual cortex, association visual cortex, posterior parietal cortex, and DLPFC were immunolabeled for PV, the 65- and 67-kDa isoforms of glutamic acid decarboxylase (GAD65 and GAD67) that synthesize GABA (gamma-aminobutyric acid), and the vesicular GABA transporter. The density of PVBC terminals and of protein levels per terminal was quantified in layer 3 of each cortical region using fluorescence confocal microscopy. RESULTS In comparison subjects, all measures, except for GAD65 levels, exhibited a caudal-to-rostral decline across the vsWM network. In subjects with schizophrenia, the density of detectable PVBC terminals was significantly lower in all regions except the DLPFC, whereas PVBC terminal levels of PV, GAD67, and GAD65 proteins were lower in all regions. A composite measure of inhibitory strength was lower in subjects with schizophrenia, although the magnitude of the diagnosis effect was greater in the primary visual, association visual, and posterior parietal cortices than in the DLPFC. CONCLUSIONS In schizophrenia, alterations in PVBC terminals across the vsWM network suggest the presence of a shared substrate for cortical dysfunction during vsWM tasks. However, regional differences in the magnitude of the disease effect on an index of PVBC inhibitory strength suggest region-specific alterations in information processing during vsWM tasks.
Collapse
Affiliation(s)
- Kenneth N Fish
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania.
| | - Brad R Rocco
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Adam M DeDionisio
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Samuel J Dienel
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Robert A Sweet
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - David A Lewis
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
32
|
Fan L, Duan Q, Luo S. Symphony of Well-Being: Harmony Between Neural Variability and Self-Construal. Front Hum Neurosci 2021; 15:679086. [PMID: 34276327 PMCID: PMC8278332 DOI: 10.3389/fnhum.2021.679086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 06/07/2021] [Indexed: 01/07/2023] Open
Abstract
Both neural activities and psychological processes vary over time. Individuals with interdependent self-construal tend to define themselves and adjust their behaviors to social contexts and others. The current research tested the hypothesis that the coordination between interdependent self-construal and neural variability could predict life satisfaction changes in university freshmen. We integrated resting-state functional magnetic resonance imaging scanning and self-construal assessment to estimate self-dependent neural variability (SDNV). In the whole-brain prediction, SDNV successfully predicted individuals’ life satisfaction changes over 2 years. Interdependent individuals with higher neural variability and independent individuals with lower neural variability became more satisfied with their lives. In the network-based prediction, the predictive effects were significant in the default mode, frontoparietal control, visual and salience networks. The important nodes that contributed to the predictive models were more related to psychological constructs associated with the social and self-oriented functions. The current research sheds light on the neural and psychological mechanisms of the subjective well-being of individuals from a dynamic perspective.
Collapse
Affiliation(s)
- Leyi Fan
- Department of Psychology, Guangdong Key Laboratory of Social Cognitive Neuroscience and Mental Health, Sun Yat-sen University, Guangzhou, China.,Department of Psychology, Guangdong Provincial Key Laboratory of Brain Function and Disease, Sun Yat-sen University, Guangzhou, China
| | - Qin Duan
- Department of Psychology, Guangdong Key Laboratory of Social Cognitive Neuroscience and Mental Health, Sun Yat-sen University, Guangzhou, China.,Department of Psychology, Guangdong Provincial Key Laboratory of Brain Function and Disease, Sun Yat-sen University, Guangzhou, China
| | - Siyang Luo
- Department of Psychology, Guangdong Key Laboratory of Social Cognitive Neuroscience and Mental Health, Sun Yat-sen University, Guangzhou, China.,Department of Psychology, Guangdong Provincial Key Laboratory of Brain Function and Disease, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
33
|
Opposite alterations of 5-HT 2A receptor brain density in subjects with schizophrenia: relevance of radiotracers pharmacological profile. Transl Psychiatry 2021; 11:302. [PMID: 34016955 PMCID: PMC8137947 DOI: 10.1038/s41398-021-01430-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 04/26/2021] [Accepted: 05/05/2021] [Indexed: 02/04/2023] Open
Abstract
The status of serotonin 5-HT2A receptors (5-HT2ARs) in schizophrenia has been controversial. In vivo positron emission tomography neuroimaging and in vitro post-mortem binding studies have reported conflicting results about 5-HT2AR density. Radiotracers bind different receptor conformations depending on their agonist, antagonist or inverse agonist properties. This study investigates 5-HT2AR density in the post-mortem prefrontal cortex from subjects with schizophrenia and controls using three radiotracers with a different pharmacological profile. The specific binding parameters of the inverse agonist [18F]altanserin, the agonist [3H]lysergic acid diethylamide (LSD) and the antagonist [3H]MDL100907 to brain cortex membranes from 20 subjects with schizophrenia and 20 individually matched controls were evaluated under similar methodological conditions. Ten schizophrenia subjects were antipsychotic-free at death. Saturation curve analyses were performed by non-linear regression to obtain a maximal density of binding sites (Bmax) and the affinity of the respective radiotracers (Kd). In schizophrenia subjects, 5-HT2AR density was decreased when quantified by [18F]altanserin binding, whereas increased when evaluated by [3H]LSD binding. However, [3H]MDL100907 binding was unaltered. A slight loss of affinity (higher Kd) was observed exclusively in [3H]LSD binding. The findings were more evident in antipsychotic-free subjects than in antipsychotic-treated subjects. In conclusion, a higher proportion of the 5-HT2AR-active functional conformation, which is rather identified by agonist radiotracers, was observed in schizophrenia patients. A consequent reduction of the inactive 5-HT2AR conformation, which is preferentially identified by inverse agonist radiotracers, was also obtained. Antagonist radiotracers do not distinguish between molecular conformations of the receptor, and accordingly, the absence of changes was shown. These results are compatible with the proposed increased functional activity of brain cortical 5-HT2ARs in schizophrenia.
Collapse
|
34
|
Jahangir M, Zhou JS, Lang B, Wang XP. GABAergic System Dysfunction and Challenges in Schizophrenia Research. Front Cell Dev Biol 2021; 9:663854. [PMID: 34055795 PMCID: PMC8160111 DOI: 10.3389/fcell.2021.663854] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 04/06/2021] [Indexed: 12/13/2022] Open
Abstract
Despite strenuous studies since the last century, the precise cause and pathology of schizophrenia are still largely unclear and arguably controversial. Although many hypotheses have been proposed to explain the etiology of schizophrenia, the definitive genes or core pathological mechanism remains absent. Among these hypotheses, however, GABAergic dysfunction stands out as a common feature consistently reported in schizophrenia, albeit a satisfactory mechanism that could be exploited for therapeutic purpose has not been developed yet. This review is focusing on the progress made to date in the field in terms of understanding the mechanisms involving dysfunctional GABAergic system and loops identified in schizophrenia research.
Collapse
Affiliation(s)
- Muhammad Jahangir
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jian-Song Zhou
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Bing Lang
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Xiao-Ping Wang
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
35
|
Jiang X, Wu F, Zhang Y, Li H, Kang J, Kong L, Wang F, Tang Y. Gender differences of amplitude of low-frequency fluctuations in bipolar disorder: A resting state fMRI study. J Affect Disord 2021; 280:189-196. [PMID: 33217701 DOI: 10.1016/j.jad.2020.11.087] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 09/27/2020] [Accepted: 11/08/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND The clinical and epidemiological features of bipolar disorder (BD) between females and males have many differences. The association between brain function and gender in BD is unknown. This research aimed to investigate the association between brain function and gender in BD by using amplitude of low-frequency fluctuations (ALFFs). METHODS Ninety-eight patients (49 females and 49 males) with BD and 171 matched healthy controls (HCs, 89 females and 82 males) were recruited for resting-state functional magnetic resonance imaging. ALFF was used to estimate brain function. RESULTS A main effect of diagnosis in ALFF was observed in the dorsal lateral prefrontal cortex (DLPFC), ventral prefrontal cortex (VPFC), caudate and occipital lobe. A main effect of gender in ALFF was found in the right VPFC, DLPFC, thalamus, and occipital lobe. A main effect of diagnosis gender interaction in ALFF was observed in the left DLPFC. Analyses of two-sample t-test indicated that male patients with BD had increased ALFF in the right hippocampus, right amygdala, left caudate, and left DLPFC, and decreased ALFF in the occipital lobe compared with male HC. Female patients with BD demonstrated increased ALFF in the right VPFC and right DLPFC compared with female HC. Male patients with BD exhibited increased ALFF in the right VPFC and left DLPFC and decreased ALFF in the occipital lobe compared with female patients with BD. LIMITATIONS This study did not consider the effect of medications and emotional states on brain activity. CONCLUSIONS Results suggested gender differences in the dysfunctions of the cortico-limbic neural system in BD.
Collapse
Affiliation(s)
- Xuejun Jiang
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China; Department of Gerontology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Feng Wu
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yifan Zhang
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Huizi Li
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jiahui Kang
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Lingtao Kong
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Fei Wang
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China; Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yanqing Tang
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China; Department of Gerontology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
36
|
Kaminski J, Mascarell-Maricic L, Fukuda Y, Katthagen T, Heinz A, Schlagenhauf F. Glutamate in the Dorsolateral Prefrontal Cortex in Patients With Schizophrenia: A Meta-analysis of 1H-Magnetic Resonance Spectroscopy Studies. Biol Psychiatry 2021; 89:270-277. [PMID: 33129486 DOI: 10.1016/j.biopsych.2020.09.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 08/28/2020] [Accepted: 09/01/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND To date, there is no systematic overview of glutamate in the dorsolateral prefrontal cortex (DLPFC) of patients with schizophrenia. Here, we meta-analyzed case-control studies of high-field proton magnetic resonance spectroscopy (1H-MRS) investigating glutamate in DLPFC. Additionally, we estimated variance ratios to investigate homo/heterogeneity. METHODS Preregistration of the study was performed on September 20, 2019. The predefined literature search on PubMed comprised articles with search terms (magnetic resonance spectroscopy OR MRS) AND (glutamate OR glut∗ OR GLX) AND (schizophrenia OR psychosis OR schizophren∗). Meta-analyses with a fixed- and random-effects model with inverse variance method, DerSimonian-Laird estimator for τ2, and Cohen's d were calculated. For differences in variability, we calculated a random-effects model for measures of variance ratios. The primary study outcome was the difference in glutamate in the DLPFC in cases versus controls. Secondary outcomes were differences in variability. RESULTS The quantitative analysis comprised 429 cases and 365 controls. Overall, we found no group difference (d = 0.03 [95% confidence interval (CI), -0.20 to 0.26], z = 0.28, p = .78). Sensitivity analysis revealed an effect for medication status (Q = 8.35, p = .039), i.e., increased glutamate in antipsychotic-naïve patients (d = 0.46 [95% CI, 0.08 to 0.84], z = 2.37, p = .018). Concerning variance ratios, we found an effect of medication status (Q = 16.95, p < .001) due to lower coefficient of variation ratio (CVR) in medication-naïve patients (logCVR = -0.49 [95% CI, -0.78 to -0.20], z = -3.33, p < .001). In studies with medicated patients, we found higher CVR (logCVR = 0.22 [95% CI, 0.06 to 0.39], z = 2.67; p = .008). CONCLUSIONS We carefully interpret the higher levels and lower variability in cortical glutamate in antipsychotic-naïve patients as a possible key factor resulting from a putative allostatic mechanism. We conclude that care has to be taken when evaluating metabolite levels in clinical samples in which medication might confound findings.
Collapse
Affiliation(s)
- Jakob Kaminski
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Psychiatry and Psychotherapy CCM, Berlin, Germany; Berlin Institute of Health, Berlin, Germany.
| | - Lea Mascarell-Maricic
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Psychiatry and Psychotherapy CCM, Berlin, Germany
| | - Yu Fukuda
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Psychiatry and Psychotherapy CCM, Berlin, Germany
| | - Teresa Katthagen
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Psychiatry and Psychotherapy CCM, Berlin, Germany
| | - Andreas Heinz
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Psychiatry and Psychotherapy CCM, Berlin, Germany; Bernstein Center for Computational Neuroscience, Berlin, Germany
| | - Florian Schlagenhauf
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Psychiatry and Psychotherapy CCM, Berlin, Germany; Berlin Institute of Health, Berlin, Germany; Bernstein Center for Computational Neuroscience, Berlin, Germany; Department of Neurology, Max Planck Institute for Human Cognitive and Brain Science, Leipzig, Germany
| |
Collapse
|
37
|
Chiola S, Napan KL, Wang Y, Lazarenko RM, Armstrong CJ, Cui J, Shcheglovitov A. Defective AMPA-mediated synaptic transmission and morphology in human neurons with hemizygous SHANK3 deletion engrafted in mouse prefrontal cortex. Mol Psychiatry 2021; 26:4670-4686. [PMID: 33558651 PMCID: PMC8349370 DOI: 10.1038/s41380-021-01023-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 12/18/2020] [Accepted: 01/08/2021] [Indexed: 01/12/2023]
Abstract
Genetic abnormalities in synaptic proteins are common in individuals with autism; however, our understanding of the cellular and molecular mechanisms disrupted by these abnormalities is limited. SHANK3 is a postsynaptic scaffolding protein of excitatory synapses that has been found mutated or deleted in most patients with 22q13 deletion syndrome and about 2% of individuals with idiopathic autism and intellectual disability. Here, we generated CRISPR/Cas9-engineered human pluripotent stem cells (PSCs) with complete hemizygous SHANK3 deletion (SHANK3+/-), which is the most common genetic abnormality in patients, and investigated the synaptic and morphological properties of SHANK3-deficient PSC-derived cortical neurons engrafted in the mouse prefrontal cortex. We show that human PSC-derived neurons integrate into the mouse cortex by acquiring appropriate cortical layer identities and by receiving and sending anatomical projections from/to multiple different brain regions. We also demonstrate that SHANK3-deficient human neurons have reduced AMPA-, but not NMDA- or GABA-mediated synaptic transmission and exhibit impaired dendritic arbors and spines, as compared to isogenic control neurons co-engrafted in the same brain region. Together, this study reveals specific synaptic and morphological deficits caused by SHANK3 hemizygosity in human cortical neurons at different developmental stages under physiological conditions and validates the use of co-engrafted control and mutant human neurons as a new platform for studying connectivity deficits in genetic neurodevelopmental disorders associated with autism.
Collapse
Affiliation(s)
- Simone Chiola
- grid.223827.e0000 0001 2193 0096Departments of Neurobiology, University of Utah, Salt Lake City, UT USA
| | - Kandy L. Napan
- grid.223827.e0000 0001 2193 0096Departments of Neurobiology, University of Utah, Salt Lake City, UT USA
| | - Yueqi Wang
- grid.223827.e0000 0001 2193 0096Departments of Neurobiology, University of Utah, Salt Lake City, UT USA ,grid.223827.e0000 0001 2193 0096Neuroscience Graduate Program, University of Utah, Salt Lake City, UT USA
| | - Roman M. Lazarenko
- grid.152326.10000 0001 2264 7217Departments of Pharmacology, Vanderbilt University, Nashville, TN USA
| | - Celeste J. Armstrong
- grid.223827.e0000 0001 2193 0096Departments of Neurobiology, University of Utah, Salt Lake City, UT USA
| | - Jun Cui
- grid.41891.350000 0001 2156 6108Department of Cell Biology and Neurosciences, Montana State University, Bozeman, MT USA
| | - Aleksandr Shcheglovitov
- Departments of Neurobiology, University of Utah, Salt Lake City, UT, USA. .,Neuroscience Graduate Program, University of Utah, Salt Lake City, UT, USA. .,Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA. .,Department of Adult Psychiatry, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
38
|
Tendilla-Beltrán H, Sanchez-Islas NDC, Marina-Ramos M, Leza JC, Flores G. The prefrontal cortex as a target for atypical antipsychotics in schizophrenia, lessons of neurodevelopmental animal models. Prog Neurobiol 2020; 199:101967. [PMID: 33271238 DOI: 10.1016/j.pneurobio.2020.101967] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 10/10/2020] [Accepted: 11/22/2020] [Indexed: 02/06/2023]
Abstract
Prefrontal cortex (PFC) inflammatory imbalance, oxidative/nitrosative stress (O/NS) and impaired neuroplasticity in schizophrenia are thought to have neurodevelopmental origins. Animal models are not only useful to test this hypothesis, they are also effective to establish a relationship among brain disturbances and behavior with the atypical antipsychotics (AAPs) effects. Here we review data of PFC post-mortem and in vivo neuroimaging, human induced pluripotent stem cells (hiPSC), and peripheral blood studies of inflammatory, O/NS, and neuroplasticity alterations in the disease as well as about their modulation by AAPs. Moreover, we reviewed the PFC alterations and the AAP mechanisms beyond their canonical antipsychotic action in four neurodevelopmental animal models relevant to the study of schizophrenia with a distinct approach in the generation of schizophrenia-like phenotypes, but all converge in O/NS and altered neuroplasticity in the PFC. These animal models not only reinforce the neurodevelopmental risk factor model of schizophrenia but also arouse some novel potential therapeutic targets for the disease including the reestablishment of the antioxidant response by the perineuronal nets (PNNs) and the nuclear factor erythroid 2-related factor (Nrf2) pathway, as well as the dendritic spine dynamics in the PFC pyramidal cells.
Collapse
Affiliation(s)
- Hiram Tendilla-Beltrán
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico; Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional (IPN), CDMX, Mexico
| | | | - Mauricio Marina-Ramos
- Departamento de Ciencias de la Salud, Universidad Popular Autónoma del Estado de Puebla, Puebla, Mexico
| | - Juan C Leza
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Instituto Universitario de Investigación en Neuroquímica (IUIN), UCM. Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Investigación Sanitaria Hospital, 12 de Octubre (Imas12), Madrid, Spain
| | - Gonzalo Flores
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico.
| |
Collapse
|
39
|
Kubota M, Moriguchi S, Takahata K, Nakajima S, Horita N. Treatment effects on neurometabolite levels in schizophrenia: A systematic review and meta-analysis of proton magnetic resonance spectroscopy studies. Schizophr Res 2020; 222:122-132. [PMID: 32505446 DOI: 10.1016/j.schres.2020.03.069] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 01/29/2020] [Accepted: 03/29/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Although there is growing evidence of alterations in the neurometabolite status associated with the pathophysiology of schizophrenia, how treatments influence these metabolite levels in patients with schizophrenia remains poorly studied. METHODS We conducted a literature search using Embase, Medline, and PsycINFO to identify proton magnetic resonance spectroscopy studies that compared neurometabolite levels before and after treatment in patients with schizophrenia. Six neurometabolites (glutamate, glutamine, glutamate + glutamine, gamma-aminobutyric acid, N-acetylaspartate, myo-inositol) and six regions of interest (frontal cortex, temporal cortex, parieto-occipital cortex, thalamus, basal ganglia, hippocampus) were investigated. RESULTS Thirty-two studies (n = 773 at follow-up) were included in our meta-analysis. Our results demonstrated that the frontal glutamate + glutamine level was significantly decreased (14 groups; n = 292 at follow-up; effect size = -0.35, P = 0.0003; I2 = 22%) and the thalamic N-acetylaspartate level was significantly increased (7 groups; n = 184 at follow-up; effect size = 0.47, P < 0.00001; I2 = 0%) after treatment in schizophrenia patients. No significant associations were found between neurometabolite changes and age, gender, duration of illness, duration of treatment, or baseline symptom severity. CONCLUSIONS The current results suggest that glutamatergic neurometabolite levels in the frontal cortex and neuronal integrity in the thalamus in schizophrenia might be modified following treatment.
Collapse
Affiliation(s)
- Manabu Kubota
- Department of Functional Brain Imaging, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan; Department of Psychiatry, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan.
| | - Sho Moriguchi
- Department of Functional Brain Imaging, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan; Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario M5T1R8, Canada
| | - Keisuke Takahata
- Department of Functional Brain Imaging, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan; Department of Neuropsychiatry, Keio University Graduate School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Shinichiro Nakajima
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario M5T1R8, Canada; Department of Neuropsychiatry, Keio University Graduate School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Nobuyuki Horita
- Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| |
Collapse
|
40
|
Yamazaki M, Honda S, Tamaki K, Irie M, Mihara T. Effects of (+)-bicuculline, a GABAa receptor antagonist, on auditory steady state response in free-moving rats. PLoS One 2020; 15:e0236363. [PMID: 32706815 PMCID: PMC7380603 DOI: 10.1371/journal.pone.0236363] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 07/03/2020] [Indexed: 12/11/2022] Open
Abstract
Auditory steady-state responses (ASSRs) are states in which the electrical activity of the brain reacts steadily to repeated auditory stimuli. They are known to be useful for testing the functional integrity of neural circuits in the cortex, as well as for their capacity to generate synchronous activity in both human and animal models. Furthermore, abnormal gamma oscillations on ASSR are typically observed in patients with schizophrenia (SZ). Changes in neural synchrony may reflect aberrations in cortical gamma-aminobutyric acid (GABA) neurotransmission. However, GABA’s impact and effects related to ASSR are still unclear. Here, we examined the effect of a GABAa receptor antagonist, (+)-bicuculline, on ASSR in free-moving rats. (+)-Bicuculline (1, 2 and 4 mg/kg, sc) markedly and dose-dependently reduced ASSR signals, consistent with current hypotheses. In particular, (+)-bicuculline significantly reduced event-related spectral perturbations (ERSPs) at 2 and 4 mg/kg between 10 and 30 minutes post-dose. Further, bicuculline (2 and 4 mg/kg) significantly and dose-dependently increased baseline gamma power. Furthermore, the occurrence of convulsions was consistent with the drug’s pharmacokinetics. For example, high doses of (+)-bicuculline such as those greater than 880 ng/g in the brain induced convulsion. Additionally, time-dependent changes in ERSP with (+)-bicuculline were observed in accordance with drug concentration. This study partially unraveled the contribution of GABAa receptor signals to the generation of ASSR.
Collapse
Affiliation(s)
- Mayako Yamazaki
- Department of Neuroscience, Drug Discovery Research, Astellas Pharma Inc., Tsukuba-shi, Ibaraki, Japan
- * E-mail:
| | - Sokichi Honda
- Department of Neuroscience, Drug Discovery Research, Astellas Pharma Inc., Tsukuba-shi, Ibaraki, Japan
| | - Keisuke Tamaki
- Department of Neuroscience, Drug Discovery Research, Astellas Pharma Inc., Tsukuba-shi, Ibaraki, Japan
| | - Megumi Irie
- Analysis & Pharmacokinetics Research Labs., Drug Discovery Research, Astellas Pharma Inc., Tsukuba-shi, Ibaraki, Japan
| | - Takuma Mihara
- Department of Neuroscience, Drug Discovery Research, Astellas Pharma Inc., Tsukuba-shi, Ibaraki, Japan
| |
Collapse
|
41
|
Kirk IJ, Spriggs MJ, Sumner RL. Human EEG and the mechanisms of memory: investigating long-term potentiation (LTP) in sensory-evoked potentials. J R Soc N Z 2020. [DOI: 10.1080/03036758.2020.1780274] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Ian J. Kirk
- Cognitive Neuroscience Research Group, School of Psychology, University of Auckland, Auckland, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- Brain Research New Zealand, New Zealand
| | - Meg J. Spriggs
- Centre for Psychedelic Research, Division of Brain Sciences, Centre for Psychiatry, Imperial College London, London, UK
| | | |
Collapse
|
42
|
Watanabe M, Marcy B, Kinoshita K, Fukasawa M, Hikichi H, Chaki S, Okuyama S, Gevorkyan H, Yoshida S. Safety and pharmacokinetic profiles of MGS0274 besylate (TS-134), a novel metabotropic glutamate 2/3 receptor agonist prodrug, in healthy subjects. Br J Clin Pharmacol 2020; 86:2286-2301. [PMID: 32353162 PMCID: PMC7576618 DOI: 10.1111/bcp.14331] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/30/2020] [Accepted: 04/20/2020] [Indexed: 01/07/2023] Open
Abstract
Aims The safety and pharmacokinetics of single and multiple doses of a novel mGlu2/3 receptor agonist prodrug, MGS0274 besylate (TS‐134), were investigated in healthy subjects. Methods Phase 1 single‐ascending dose (5–20 mg) and multiple‐ascending dose titration (5–80 mg) studies were conducted in healthy male and female subjects. Both studies were randomized, double‐blinded and placebo‐controlled. In one cohort of single‐ascending dose study (10 mg), concentrations of MGS0008, the active compound, in the cerebrospinal fluid (CSF) were measured for up to 24 hours postdose. Results Following single and multiple oral administrations, MGS0274 was rapidly absorbed and extensively converted into MGS0008, which reached a maximum concentration (Cmax) in plasma within 4 hours postdose and declined with a terminal half‐life (t1/2) of around 10 hours. Plasma exposure to MGS0274 was minimal, accounting for approximately 3% of the area under the concentration–time curve (AUC) of MGS0008. Plasma Cmax and AUC of MGS0008 at steady state increased dose proportionally (5–80 mg). MGS0008 penetrated into CSF, with a CSF‐to‐plasma Cmax ratio of 3.66%, and was eliminated with a t1/2 of approximately 16 hours. The most frequent treatment‐emergent adverse events observed following single and multiple oral administration included headache, nausea, somnolence, dizziness and vomiting. Conclusion TS‐134 is orally bioavailable in humans and converts rapidly and extensively to MGS0008, which exhibits good CSF penetration. Orally administered TS‐134 was safe and generally well‐tolerated; hence, TS‐134 is a promising candidate for further clinical development for the treatment of disorders in which glutamatergic abnormalities are involved, such as schizophrenia.
Collapse
|
43
|
Herman AB, Brown EG, Dale CL, Hinkley LB, Subramaniam K, Houde JF, Fisher M, Vinogradov S, Nagarajan SS. The Visual Word Form Area compensates for auditory working memory dysfunction in schizophrenia. Sci Rep 2020; 10:8881. [PMID: 32483253 PMCID: PMC7264140 DOI: 10.1038/s41598-020-63962-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 03/28/2020] [Indexed: 11/23/2022] Open
Abstract
Auditory working memory impairments feature prominently in schizophrenia. However, the existence of altered and perhaps compensatory neural dynamics, sub-serving auditory working memory, remains largely unexplored. We compared the dynamics of induced high gamma power (iHGP) across cortex in humans during speech-sound working memory in individuals with schizophrenia (SZ) and healthy comparison subjects (HC) using magnetoencephalography (MEG). SZ showed similar task performance to HC while utilizing different brain regions. During encoding of speech sounds, SZ lacked the correlation of iHGP with task performance in posterior superior temporal gyrus (STGp) that was observed in healthy subjects. Instead, SZ recruited the visual word form area (VWFA) during both stimulus encoding and response preparation. Importantly, VWFA activity during encoding correlated with the magnitude of SZ hallucinations, task performance and an independent measure of verbal working memory. These findings suggest that VWFA plasticity is harnessed to compensate for STGp dysfunction in schizophrenia patients with hallucinations.
Collapse
Affiliation(s)
- Alexander B Herman
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
- UCB-UCSF Graduate Program in Bioengineering, University of California, Berkeley, Berkeley, CA, United States
- Department of Psychiatry, University of Minnesota, Minneapolis, MN, United States
| | - Ethan G Brown
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
- Department of Neurology, University of California, San Francisco, San Francisco, CA, United States
| | - Corby L Dale
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
| | - Leighton B Hinkley
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
| | - Karuna Subramaniam
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
| | - John F Houde
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
| | - Melissa Fisher
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
- Department of Psychiatry, University of Minnesota, Minneapolis, MN, United States
- San Francisco Veterans' Affairs Medical Center, San Francisco, CA, United States
| | - Sophia Vinogradov
- Department of Psychiatry, University of Minnesota, Minneapolis, MN, United States
- San Francisco Veterans' Affairs Medical Center, San Francisco, CA, United States
| | - Srikantan S Nagarajan
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States.
- Department of Neurology, University of California, San Francisco, San Francisco, CA, United States.
| |
Collapse
|
44
|
Snyder MA, Gao WJ. NMDA receptor hypofunction for schizophrenia revisited: Perspectives from epigenetic mechanisms. Schizophr Res 2020; 217:60-70. [PMID: 30979669 PMCID: PMC7258307 DOI: 10.1016/j.schres.2019.03.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 03/08/2019] [Accepted: 03/11/2019] [Indexed: 02/06/2023]
Abstract
Schizophrenia (SZ) is a neurodevelopmental disorder with cognitive deficits manifesting during early stages of the disease. Evidence suggests that genetic factors in combination with environmental insults lead to complex changes to glutamatergic, GABAergic, and dopaminergic systems. In particular, the N-methyl-d-aspartate receptor (NMDAR), a major glutamate receptor subtype, is implicated in both the disease progression and symptoms of SZ. NMDARs are critical for synaptic plasticity and cortical maturation, as well as learning and memory processes. In fact, any deviation from normal NMDAR expression and function can have devastating consequences. Surprisingly, there is little evidence from human patients that direct mutations of NMDAR genes contribute to SZ. One intriguing hypothesis is that epigenetic changes, which could result from early insults, alter protein expression and contribute to the NMDAR hypofunction found in SZ. Epigenetics is referred to as modifications that alter gene transcription without changing the DNA sequence itself. In this review, we first discuss how epigenetic changes to NMDAR genes could contribute to NMDAR hypofunction. We then explore how NMDAR hypofunction may contribute to epigenetic changes in other proteins or genes that lead to synaptic dysfunction and symptoms in SZ. We argue that NMDAR hypofunction occurs in early stage of the disease, and it may consequentially initiate GABA and dopamine deficits. Therefore, targeting NMDAR dysfunction during the early stages would be a promising avenue for prevention and therapeutic intervention of cognitive and social deficits that remain untreatable. Finally, we discuss potential questions regarding the epigenetic of SZ and future directions for research.
Collapse
Affiliation(s)
- Melissa A. Snyder
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada, K1H 8M5,Correspondence: Wen-Jun Gao, M.D., Ph.D., Department of Neurobiology and Anatomy, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, Phone: (215) 991-8907, Fax: (215) 843-9802, ; Melissa A. Snyder, Ph.D.,
| | - Wen-Jun Gao
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, United States of America.
| |
Collapse
|
45
|
Kaminski J, Gleich T, Fukuda Y, Katthagen T, Gallinat J, Heinz A, Schlagenhauf F. Association of Cortical Glutamate and Working Memory Activation in Patients With Schizophrenia: A Multimodal Proton Magnetic Resonance Spectroscopy and Functional Magnetic Resonance Imaging Study. Biol Psychiatry 2020; 87:225-233. [PMID: 31521336 DOI: 10.1016/j.biopsych.2019.07.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 07/01/2019] [Accepted: 07/04/2019] [Indexed: 01/06/2023]
Abstract
BACKGROUND Cognitive deficits such as working memory (WM) impairment are core features of schizophrenia. One candidate marker for the integrity of synaptic neurotransmission necessary for cognitive processes is glutamate. It is frequently postulated that antipsychotic medication possibly alters functional mechanisms in the living brain. We tested in vivo for group differences in activation of the dorsolateral prefrontal cortex (DLPFC) during WM performance and the association with glutamate concentration in DLPFC depending on medication status. METHODS A total of 90 subjects (35 control subjects, 36 medicated patients, and 19 unmedicated patients) contributed magnetic resonance spectroscopy data. We estimated glutamate in left DLPFC. Subjects performed an n-back WM task (2-back vs. 0-back) during functional magnetic resonance imaging, and local activation in left DLPFC was measured. For analysis of association with medication status, we calculated linear regression models including an interaction effect with group. RESULTS Medicated and unmedicated patients with schizophrenia showed impaired performance. We found significantly reduced WM activation in left DLPFC in medicated patients and a trendwise reduction in unmedicated patients as compared with control subjects. We found no group difference in local glutamate concentration. However, we found differential effects of medication status on the association between local glutamate concentration and WM activation in left DLPFC, with a positive association in unmedicated patients but not in medicated patients. CONCLUSIONS We provide evidence that WM-dependent activation is associated with glutamate concentration in unmedicated patients with schizophrenia. Our finding points to putative allostatic changes that affect the functioning of the brain and might be altered through medication.
Collapse
Affiliation(s)
- Jakob Kaminski
- Department of Psychiatry and Psychotherapy Charité Campus Mitte, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany; Berlin Institute of Health, Berlin, Germany.
| | - Tobias Gleich
- Department of Psychiatry and Psychotherapy Charité Campus Mitte, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Yu Fukuda
- Department of Psychiatry and Psychotherapy Charité Campus Mitte, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Teresa Katthagen
- Department of Psychiatry and Psychotherapy Charité Campus Mitte, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jürgen Gallinat
- University Clinic Hamburg-Eppendorf, Clinic and Policlinic for Psychiatry and Psychotherapy, Hamburg, Germany
| | - Andreas Heinz
- Department of Psychiatry and Psychotherapy Charité Campus Mitte, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Florian Schlagenhauf
- Department of Psychiatry and Psychotherapy Charité Campus Mitte, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany; Berlin Institute of Health, Berlin, Germany; Bernstein Center for Computational Neuroscience, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
46
|
Pinner AL, Mueller TM, Alganem K, McCullumsmith R, Meador-Woodruff JH. Protein expression of prenyltransferase subunits in postmortem schizophrenia dorsolateral prefrontal cortex. Transl Psychiatry 2020; 10:3. [PMID: 32066669 PMCID: PMC7026430 DOI: 10.1038/s41398-019-0610-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 09/09/2019] [Accepted: 10/03/2019] [Indexed: 12/22/2022] Open
Abstract
The pathophysiology of schizophrenia includes altered neurotransmission, dysregulated intracellular signaling pathway activity, and abnormal dendritic morphology that contribute to deficits of synaptic plasticity in the disorder. These processes all require dynamic protein-protein interactions at cell membranes. Lipid modifications target proteins to membranes by increasing substrate hydrophobicity by the addition of a fatty acid or isoprenyl moiety, and recent evidence suggests that dysregulated posttranslational lipid modifications may play a role in multiple neuropsychiatric disorders, including schizophrenia. Consistent with these emerging findings, we have recently reported decreased protein S-palmitoylation in schizophrenia. Protein prenylation is a lipid modification that occurs upstream of S-palmitoylation on many protein substrates, facilitating membrane localization and activity of key intracellular signaling proteins. Accordingly, we hypothesized that, in addition to palmitoylation, protein prenylation may be abnormal in schizophrenia. To test this, we assayed protein expression of the five prenyltransferase subunits (FNTA, FNTB, PGGT1B, RABGGTA, and RABGGTB) in postmortem dorsolateral prefrontal cortex from patients with schizophrenia and paired comparison subjects (n = 13 pairs). We found decreased levels of FNTA (14%), PGGT1B (13%), and RABGGTB (8%) in schizophrenia. To determine whether upstream or downstream factors may be driving these changes, we also assayed protein expression of the isoprenoid synthases FDPS and GGPS1 and prenylation-dependent processing enzymes RCE and ICMT. We found these upstream and downstream enzymes to have normal protein expression. To rule out effects from chronic antipsychotic treatment, we assayed FNTA, PGGT1B, and RABGGTB in the cortex from rats treated long-term with haloperidol decanoate and found no change in the expression of these proteins. Given the role prenylation plays in localization of key signaling proteins found at the synapse, these data offer a potential mechanism underlying abnormal protein-protein interactions and protein localization in schizophrenia.
Collapse
Affiliation(s)
- Anita L Pinner
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294-0021, USA.
| | - Toni M Mueller
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294-0021, USA
| | - Khaled Alganem
- Department of Neurosciences, University of Toledo, Toledo, OH, 43614-2598, USA
| | | | - James H Meador-Woodruff
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294-0021, USA
| |
Collapse
|
47
|
Deutsch SI, Burket JA. An Evolving Therapeutic Rationale for Targeting the α 7 Nicotinic Acetylcholine Receptor in Autism Spectrum Disorder. Curr Top Behav Neurosci 2020; 45:167-208. [PMID: 32468495 DOI: 10.1007/7854_2020_136] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Abnormalities of cholinergic nuclei, cholinergic projections, and cholinergic receptors, as well as abnormalities of growth factors involved in the maturation and maintenance of cholinergic neurons, have been described in postmortem brains of persons with autism spectrum disorder (ASD). Further, microdeletions of the 15q13.3 locus that encompasses CHRNA7, the gene coding the α7 nicotinic acetylcholine receptor (α7 nAChR), are associated with a spectrum of neurodevelopmental disorders, including ASD. The heterozygous 15q13.3 microdeletion syndrome suggests that diminished or impaired transduction of the acetylcholine (ACh) signal by the α7 nAChR can be a pathogenic mechanism of ASD. The α7 nAChR has a role in regulating the firing and function of parvalbumin (PV)-expressing GABAergic projections, which synchronize the oscillatory output of assemblies of pyramidal neurons onto which they project. Synchronous oscillatory output is an electrophysiological substrate for higher executive functions, such as working memory, and functional connectivity between discrete anatomic areas of the brain. The α7 nAChR regulates PV expression and works cooperatively with the co-expressed NMDA receptor in subpopulations of GABAergic interneurons in mouse models of ASD. An evolving literature supports therapeutic exploration of selectively targeted cholinergic interventions for the treatment of ASD, especially compounds that target the α7 nAChR subtype. Importantly, development and availability of high-affinity, brain-penetrable, α7 nAChR-selective agonists, partial agonists, allosteric agonists, and positive allosteric modulators (PAMs) should facilitate "proof-of-principle/concept" clinical trials. nAChRs are pentameric allosteric proteins that function as ligand-gated ion channel receptors constructed from five constituent polypeptide subunits, all of which share a common structural motif. Importantly, in addition to α7 nAChR-gated Ca2+ conductance causing membrane depolarization, there are emerging data consistent with possible metabotropic functions of this ionotropic receptor. The ability of α7-selective type II PAMs to "destabilize" the desensitized state and promote ion channel opening may afford them therapeutic advantages over orthosteric agonists. The current chapter reviews historic and recent literature supporting selective therapeutic targeting of the α7 nAChR in persons affected with ASD.
Collapse
Affiliation(s)
- Stephen I Deutsch
- Department of Psychiatry and Behavioral Sciences, Eastern Virginia Medical School, Norfolk, VA, USA.
| | - Jessica A Burket
- Department of Molecular Biology and Chemistry, Christopher Newport University, Newport News, VA, USA
| |
Collapse
|
48
|
Driver DI, Thomas S, Gogtay N, Rapoport JL. Childhood-Onset Schizophrenia and Early-onset Schizophrenia Spectrum Disorders: An Update. Child Adolesc Psychiatr Clin N Am 2020; 29:71-90. [PMID: 31708054 DOI: 10.1016/j.chc.2019.08.017] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The clinical severity, impact on development, and poor prognosis of childhood-onset schizophrenia may represent a more homogeneous group. Positive symptoms in children are necessary for the diagnosis, and hallucinations are more often multimodal. In healthy children and children with a variety of other psychiatric illnesses, hallucinations are not uncommon and diagnosis should not be based on these alone. Childhood-onset schizophrenia is an extraordinarily rare illness that is poorly understood but seems continuous with the adult-onset disorder. Once a diagnosis is confirmed, aggressive medication treatment combined with family education and individual counseling may prevent further deterioration.
Collapse
Affiliation(s)
- David I Driver
- Child Psychiatry Branch, National Institutes of Mental Health (NIMH), National Institutes Health (NIH), Building 10, Room 4N313C, 10 Center Drive, Bethesda, MD 20814, USA.
| | - Shari Thomas
- Healthy Foundations Group, 4350 East West Highway, Suite 200, Bethesda, Maryland 20814, USA
| | - Nitin Gogtay
- National Institutes Health (NIH), NSC Building, Room 6104, 6001 Executive Boulevard, Rockville, MD 20852, USA
| | - Judith L Rapoport
- National Institutes Health (NIH), Building 10-CRC, Room 6-5332, 10 Center Drive, Bethesda, MD 20814, USA
| |
Collapse
|
49
|
García-Bea A, Miranda-Azpiazu P, Muguruza C, Marmolejo-Martinez-Artesero S, Diez-Alarcia R, Gabilondo AM, Callado LF, Morentin B, González-Maeso J, Meana JJ. Serotonin 5-HT 2A receptor expression and functionality in postmortem frontal cortex of subjects with schizophrenia: Selective biased agonism via G αi1-proteins. Eur Neuropsychopharmacol 2019; 29:1453-1463. [PMID: 31734018 DOI: 10.1016/j.euroneuro.2019.10.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 10/17/2019] [Accepted: 10/29/2019] [Indexed: 12/20/2022]
Abstract
Serotonin 5-HT2A receptors (5-HT2ARs) have been implicated in schizophrenia. However, postmortem studies on 5-HT2ARs expression and functionality in schizophrenia are scarce. The 5-HT2AR mRNA and immunoreactive protein expression were evaluated in postmortem tissue from dorsolateral prefrontal cortex (DLPFC) of antipsychotic-free (n = 18) and antipsychotic-treated (n = 9) subjects with schizophrenia, and matched controls (n = 27). Functional coupling of 5-HT2AR to G-proteins was tested by measuring the activation induced by the agonist (±)-2,5-dimethoxy-4-iodoamphetamine hydrochloride ((±)DOI) in antibody-capture [35S]GTPγS scintillation proximity assays (SPA). In antipsychotic-free schizophrenia subjects, 5-HT2AR mRNA expression and protein immunoreactivity in total homogenates was similar to controls. In contrast, in antipsychotic-treated schizophrenia subjects, lower mRNA expression (60±9% vs controls) and a trend to reduced protein immunoreactivity (86±5% vs antipsychotic-free subjects) just in membrane-enriched fractions was observed. [35S]GTPγS SPA revealed a significant ~6% higher stimulation of Gαi1-protein by (±)DOI in schizophrenia, whereas activation of the canonical Gαq/11-protein pathway by (±)DOI remained unchanged. Expression of Gαi1- and Gαq/11-proteins did not differ between groups. Accordingly, in rats chronically treated with clozapine, but not with haloperidol, a 30-40% reduction was observed in 5-HT2AR mRNA expression, 5-HT2AR protein immunoreactivity and [3H]ketanserin binding in brain cortical membranes. Overall, the data suggest a supersensitive 5-HT2AR signaling through inhibitory Gαi1-proteins in schizophrenia. Together with previous results, a dysfunctional pro-hallucinogenic agonist-sensitive 5-HT2AR conformation in postmortem DLPFC of subjects with schizophrenia is proposed. Atypical antipsychotic treatment would contribute to counterbalance this 5-HT2AR supersensitivity by reducing receptor expression.
Collapse
Affiliation(s)
- Aintzane García-Bea
- Department of Pharmacology, University of the Basque Country UPV/EHU, 48940 Leioa, Bizkaia, Spain; Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain
| | - Patricia Miranda-Azpiazu
- Department of Pharmacology, University of the Basque Country UPV/EHU, 48940 Leioa, Bizkaia, Spain; Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain
| | - Carolina Muguruza
- Department of Pharmacology, University of the Basque Country UPV/EHU, 48940 Leioa, Bizkaia, Spain; Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain
| | | | - Rebeca Diez-Alarcia
- Department of Pharmacology, University of the Basque Country UPV/EHU, 48940 Leioa, Bizkaia, Spain; Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain; Biocruces Bizkaia Health Research Institute, Spain
| | - Ane M Gabilondo
- Department of Pharmacology, University of the Basque Country UPV/EHU, 48940 Leioa, Bizkaia, Spain; Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain; Biocruces Bizkaia Health Research Institute, Spain
| | - Luis F Callado
- Department of Pharmacology, University of the Basque Country UPV/EHU, 48940 Leioa, Bizkaia, Spain; Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain; Biocruces Bizkaia Health Research Institute, Spain
| | - Benito Morentin
- Biocruces Bizkaia Health Research Institute, Spain; Basque Institute of Legal Medicine, Spain
| | - Javier González-Maeso
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, VA, USA
| | - J Javier Meana
- Department of Pharmacology, University of the Basque Country UPV/EHU, 48940 Leioa, Bizkaia, Spain; Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain; Biocruces Bizkaia Health Research Institute, Spain.
| |
Collapse
|
50
|
Zhang XQ, Yu ZP, Ling Y, Zhao QQ, Zhang ZY, Wang ZC, Shen HW. Enduring effects of juvenile social isolation on physiological properties of medium spiny neurons in nucleus accumbens. Psychopharmacology (Berl) 2019; 236:3281-3289. [PMID: 31197434 DOI: 10.1007/s00213-019-05284-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 05/24/2019] [Indexed: 12/13/2022]
Abstract
RATIONALE Juvenile social isolation (SI) and neglect is associated with a wide range of psychiatric disorders. While dysfunction of the corticolimbic pathway is considered to link various abnormal behaviors in SI models of schizophrenia, the enduring effects of early social deprivation on physiological properties of medium spiny neurons (MSNs) in nucleus accumbens (NAc) are not well understood. OBJECTIVES This study investigated the impacts of juvenile SI on locomotor activity to methamphetamine (METH) and neurophysiological characteristics of MSNs in the core of NAc. METHODS Socially isolated C57BL/6 mice experienced single housing for 4 weeks on postnatal day (PND) 21. The locomotor response to METH (1.0 mg/kg) was observed in both socially isolated and group-housed mice at PND 56. The effects of juvenile SI on the excitatory synaptic events in MSNs and the intrinsic excitability of MSNs in NAc core were investigated in other batches during PND 63-70. RESULTS Socially isolated mice showed locomotor hypersensitivity to METH, although the expression of locomotor sensitization to METH in socially isolated mice was not different from group-housed mice. The recordings from MSNs of SI-reared mice exhibited higher frequency and smaller amplitude of miniature/spontaneous excitatory postsynaptic current than those from group-reared mice. Moreover, SI resulted in increased intrinsic excitability of MSNs in adult mice. CONCLUSIONS These results demonstrate neuronal hyperactivity in the NAc of socially isolated mice, which could contribute to locomotor hypersensitivity to METH. Furthermore, the findings indicate a biological link between early negative life events and the vulnerability to psychostimulant-induced psychosis in adulthood.
Collapse
Affiliation(s)
- Xiao-Qin Zhang
- Department of Pharmacology, Medical School of Ningbo University, 818 Fenghua Rd, Ningbo, Zhejiang, 315211, China
| | - Zhi-Peng Yu
- Department of Pharmacology, Medical School of Ningbo University, 818 Fenghua Rd, Ningbo, Zhejiang, 315211, China
| | - Yu Ling
- Department of Pharmacology, Medical School of Ningbo University, 818 Fenghua Rd, Ningbo, Zhejiang, 315211, China
| | - Qi-Qi Zhao
- Department of Pharmacology, Medical School of Ningbo University, 818 Fenghua Rd, Ningbo, Zhejiang, 315211, China
| | - Zhong-Yu Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Rd, Beijing, 100191, China
| | - Zheng-Chun Wang
- Department of Pharmacology, Medical School of Ningbo University, 818 Fenghua Rd, Ningbo, Zhejiang, 315211, China
| | - Hao-Wei Shen
- Department of Pharmacology, Medical School of Ningbo University, 818 Fenghua Rd, Ningbo, Zhejiang, 315211, China.
| |
Collapse
|