1
|
Hojjati SH, Chen K, Chiang GC, Kuceyeski A, Wang XH, Razlighi QR, Pahlajani S, Glodzik L, Tanzi EB, Reinhardt M, Butler TA. Utilizing structural MRI and unsupervised clustering to differentiate schizophrenia and Alzheimer's disease in late-onset psychosis. Behav Brain Res 2024; 480:115386. [PMID: 39644998 DOI: 10.1016/j.bbr.2024.115386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/14/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
Late-onset psychosis (LOP) represents a highly heterogeneous and understudied condition, with potential origins ranging from atypically late onset of schizophrenia (SCZ) to Alzheimer's Disease (AD). Despite the clinical necessity of differentiating these conditions to guide effective treatment, achieving an accurate diagnosis remains challenging. This study aimed to utilize data-driven analyses of structural magnetic resonance imaging (MRI) to distinguish between these diagnostic possibilities. Utilizing publicly available datasets of MRI scans from 699 healthy control (HC) participants and 469 patients diagnosed with SCZ or AD, our analysis focused on bilateral subcortical volumetric measures in the caudate, hippocampus, putamen, and amygdala. We first trained an unsupervised K-means clustering algorithm based on SCZ and AD patients and achieved a clustering accuracy of 81 % and an area under curvature (AUC) of 0.79 in distinguishing between these two groups. Subsequently, we calculated the Euclidean distance between the AD and SCZ cluster centroids for each of ten patients with unexplained onset of psychosis after age 45 from a clinical MRI registry. Six patients were classified as AD and four as SCZ. Our findings revealed that among LOP participants, those classified in the SCZ cluster exhibited significantly greater right putamen volumes compared to those in the AD cluster (p < 0.0025). There were also intriguing clinical differences. While we do not have diagnostic biomarker information to confirm these classifications, this study sheds light on the heterogeneity of psychoses in late life and illustrates the potential use of widely available structural MRI and data-driven methods to enhance diagnostic accuracy and treatment outcomes for LOP patients.
Collapse
Affiliation(s)
- Seyed Hani Hojjati
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, New York, NY, USA.
| | - Kewei Chen
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA; School of Mathematics and Statistics, Arizona State University, Phoenix, AZ, USA; Department of Neurology, University of Arizona College of Medicine, Phoenix, AZ, USA; Banner Alzheimer's Institute, Phoenix, AZ, USA
| | - Gloria C Chiang
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, New York, NY, USA
| | - Amy Kuceyeski
- Department of Computational Biology, Cornell University, Ithaca, NY, USA; Department of Psychiatry, Brain Health Institute, Rutgers University, Piscataway, NJ, USA
| | - Xiuyuan H Wang
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, New York, NY, USA
| | - Qolamreza R Razlighi
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, New York, NY, USA
| | - Silky Pahlajani
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, New York, NY, USA
| | - Lidia Glodzik
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, New York, NY, USA
| | - Emily B Tanzi
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, New York, NY, USA
| | - Michael Reinhardt
- Department of Psychiatry, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Tracy A Butler
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
2
|
Soon CW, Gaurav A, Gautam V, Al-Nema M. Structural insight into the lead identification of a dual inhibitor of PDE1B and PDE10A: Integrating pharmacophore-based virtual screening, molecular docking, and structure-activity-relationship approaches. Heliyon 2024; 10:e38305. [PMID: 39391487 PMCID: PMC11466560 DOI: 10.1016/j.heliyon.2024.e38305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 09/05/2024] [Accepted: 09/22/2024] [Indexed: 10/12/2024] Open
Abstract
Schizophrenia is a chronic neuropsychiatric disorder affecting more than 1% of the world's population. Current antipsychotic treatments show inadequacy in mitigating the negative and cognitive symptoms of schizophrenia. In addition, these medications cause undesirable extrapyramidal side effects. According to the studies, inhibition of phosphodiesterase (PDE) 1B and PDE10A simultaneously can alleviate positive, negative, and cognitive symptoms of schizophrenia. Thus, this study aims to identify new dual inhibitors of PDE1B and PDE10A using ligand-based pharmacophore modelling, virtual screening, and molecular docking studies. Accordingly, the generated pharmacophore models of PDE1B and PDE10A comprised hydrogen bond acceptor, aromatic ring, and hydrophobic features. These features were essential for retrieving the active hits from the Universal Natural Product Database in the virtual screening. Additional filters were subsequently employed to identify potential hits that could be developed into central nervous system-active compounds. Hits meeting all the screening criteria were subjected to docking studies with PDE1B and PDE10A. Among these hits, UNPD167314 exhibited significant binding affinities for the target receptors. It occupied the P-clamp and displayed hydrophobic, aromatic, and hydrogen bond interactions with the active site residues of both receptors, thus selected as a lead compound for the design of potent and selective dual inhibitors. The structural modifications of UNPD167314 resulted in the design of 35 novel inhibitors. Out of 35, four compounds exhibited high and comparable binding affinities for both PDE1B and PDE10A, making them promising candidates for further evaluation and optimisation.
Collapse
Affiliation(s)
- Ching Wen Soon
- Faculty of Pharmaceutical Sciences, UCSI University, Taman Connaught, Cheras, Kuala Lumpur, 56000, Malaysia
| | - Anand Gaurav
- Department of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun, 248007, Uttarakhand, India
- Faculty of Health Sciences, Villa College, Male', 20373, Maldives
| | - Vertika Gautam
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, Uttar Pradesh, India
| | - Mayasah Al-Nema
- Faculty of Pharmaceutical Sciences, UCSI University, Taman Connaught, Cheras, Kuala Lumpur, 56000, Malaysia
- Klarity, Westbourne, Bournemouth, BH4 8DT, United Kingdom
| |
Collapse
|
3
|
Andreou D, Jørgensen KN, Nerland S, Calkova T, Mørch-Johnsen L, Smelror RE, Wortinger LA, Lundberg M, Bohman H, Myhre AM, Jönsson EG, Andreassen OA, Agartz I. Caudate nucleus volume in medicated and unmedicated patients with early- and adult-onset schizophrenia. Sci Rep 2024; 14:22755. [PMID: 39353988 PMCID: PMC11445249 DOI: 10.1038/s41598-024-73322-x] [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: 03/20/2024] [Accepted: 09/16/2024] [Indexed: 10/03/2024] Open
Abstract
The caudate nucleus is a part of the striatum, and striatal hyperdopaminergia is considered central to the pathophysiology of schizophrenia. How caudate volume is affected in schizophrenia and what role antipsychotics play remains unclear. In early-onset schizophrenia (EOS), where psychosis emerges during a neurodevelopmentally critical phase, the caudate may exhibit a heightened vulnerability to the effects of antipsychotic medications. We hypothesized effects of both antipsychotic medication use and age of onset on caudate in schizophrenia. We included adult patients with EOS (n = 83) and adult-onset schizophrenia (AOS) (n = 246), adult healthy controls (HC, n = 774), adolescent patients with non-affective psychosis (n = 56) and adolescent HC (n = 97). We obtained T1-weighted MRI scans using a 1.5T Siemens scanner and General Electric 3T scanners. In our main analysis, we tested for main and interaction effects of diagnosis and current antipsychotic medication use on caudate volume. Adult patients with EOS (p < 0.001) and AOS (p = 0.002) had both larger caudate than HC. Age of onset (EOS/AOS) interacted with antipsychotic use (p = 0.004) which was associated with larger caudate in EOS (p < 0.001) but not in AOS (p = 0.654). Conversely, among medicated patients only, EOS had larger caudate than AOS (p < 0.001). No other subcortical structures showed differences between medicated EOS and AOS. Medicated adolescent patients with non-affective psychosis and medicated adult patients with EOS showed similar caudate volumes. The results may indicate a schizophrenia-related and a medication-induced caudate increase, the latter restricted to patients with EOS and possibly occurring already in adolescence shortly after disease onset.
Collapse
Affiliation(s)
- Dimitrios Andreou
- Department of Psychiatric Research, Diakonhjemmet Hospital, Forskningsveien 7, 0373, Oslo, Norway.
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Stockholm Region, Stockholm, Sweden.
| | - Kjetil Nordbø Jørgensen
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Vestre Viken Hospital Trust, Drammen, Norway
| | - Stener Nerland
- Department of Psychiatric Research, Diakonhjemmet Hospital, Forskningsveien 7, 0373, Oslo, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Tereza Calkova
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Stockholm Region, Stockholm, Sweden
- Centre for Clinical Research, Vastmanland Hospital Vasteras, Region Vastmanland - Uppsala University, Västerås, Sweden
| | - Lynn Mørch-Johnsen
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychiatry, Department of Clinical Research, Østfold Hospital, Grålum, Norway
| | - Runar Elle Smelror
- Department of Psychiatric Research, Diakonhjemmet Hospital, Forskningsveien 7, 0373, Oslo, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Laura A Wortinger
- Department of Psychiatric Research, Diakonhjemmet Hospital, Forskningsveien 7, 0373, Oslo, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Mathias Lundberg
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Hannes Bohman
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
- Department of Neuroscience, Child and Adolescent Psychiatry and Psychiatry Unit, Uppsala University, Uppsala, Sweden
| | - Anne Margrethe Myhre
- Division of Mental Health and Addiction, Departement of Research and innovation, Oslo University Hospital, Oslo, Norway
- Child and Adolescent Psychiatry Unit, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Erik G Jönsson
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Stockholm Region, Stockholm, Sweden
| | - Ole A Andreassen
- Division of Mental Health and Addiction, Norwegian Centre for Mental Disorders Research (NORMENT), Oslo University Hospital, Oslo, Norway
| | - Ingrid Agartz
- Department of Psychiatric Research, Diakonhjemmet Hospital, Forskningsveien 7, 0373, Oslo, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Stockholm Region, Stockholm, Sweden
| |
Collapse
|
4
|
Basso V, Döbrössy MD, Thompson LH, Kirik D, Fuller HR, Gates MA. State of the Art in Sub-Phenotyping Midbrain Dopamine Neurons. BIOLOGY 2024; 13:690. [PMID: 39336117 PMCID: PMC11428604 DOI: 10.3390/biology13090690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 09/30/2024]
Abstract
Dopaminergic neurons in the ventral tegmental area (VTA) and the substantia nigra pars compacta (SNpc) comprise around 75% of all dopaminergic neurons in the human brain. While both groups of dopaminergic neurons are in close proximity in the midbrain and partially overlap, development, function, and impairments in these two classes of neurons are highly diverse. The molecular and cellular mechanisms underlying these differences are not yet fully understood, but research over the past decade has highlighted the need to differentiate between these two classes of dopaminergic neurons during their development and in the mature brain. This differentiation is crucial not only for understanding fundamental circuitry formation in the brain but also for developing therapies targeted to specific dopaminergic neuron classes without affecting others. In this review, we summarize the state of the art in our understanding of the differences between the dopaminergic neurons of the VTA and the SNpc, such as anatomy, structure, morphology, output and input, electrophysiology, development, and disorders, and discuss the current technologies and methods available for studying these two classes of dopaminergic neurons, highlighting their advantages, limitations, and the necessary improvements required to achieve more-precise therapeutic interventions.
Collapse
Affiliation(s)
- Valentina Basso
- School of Medicine, Keele University, Staffordshire ST5 5BG, UK
| | - Máté D Döbrössy
- Laboratory of Stereotaxy and Interventional Neurosciences, Department of Stereotactic and Functional, Neurosurgery, Medical Center, University of Freiburg, 79106 Freiburg im Breisgau, Germany
- Department of Stereotactic and Functional Neurosurgery, Medical Center, University of Freiburg, 79106 Freiburg im Breisgau, Germany
- Faculty of Biology, University of Freiburg, 79104 Freiburg im Breisgau, Germany
| | - Lachlan H Thompson
- Charles Perkins Centre, Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, NSW 2006, Australia
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Deniz Kirik
- Brain Repair and Imaging in Neural Systems (B.R.A.I.N.S) Unit, Department of Experimental Medical Science, Lund University, BMC D11, 22184 Lund, Sweden
| | - Heidi R Fuller
- School of Pharmacy and Bioengineering, Keele University, Staffordshire ST5 5BG, UK
- Wolfson Centre for Inherited Neuromuscular Disease, TORCH Building, RJAH Orthopaedic Hospital, Oswestry SY10 7AG, UK
| | - Monte A Gates
- School of Medicine, Keele University, Staffordshire ST5 5BG, UK
| |
Collapse
|
5
|
Redinbaugh MJ, Saalmann YB. Contributions of Basal Ganglia Circuits to Perception, Attention, and Consciousness. J Cogn Neurosci 2024; 36:1620-1642. [PMID: 38695762 PMCID: PMC11223727 DOI: 10.1162/jocn_a_02177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Research into ascending sensory pathways and cortical networks has generated detailed models of perception. These same cortical regions are strongly connected to subcortical structures, such as the basal ganglia (BG), which have been conceptualized as playing key roles in reinforcement learning and action selection. However, because the BG amasses experiential evidence from higher and lower levels of cortical hierarchies, as well as higher-order thalamus, it is well positioned to dynamically influence perception. Here, we review anatomical, functional, and clinical evidence to demonstrate how the BG can influence perceptual processing and conscious states. This depends on the integrative relationship between cortex, BG, and thalamus, which allows contributions to sensory gating, predictive processing, selective attention, and representation of the temporal structure of events.
Collapse
Affiliation(s)
| | - Yuri B Saalmann
- University of Wisconsin-Madison
- Wisconsin National Primate Research Center
| |
Collapse
|
6
|
Hirsch F, Wohlschlaeger A. Subcortical influences on the topology of cortical networks align with functional processing hierarchies. Neuroimage 2023; 283:120417. [PMID: 37866758 DOI: 10.1016/j.neuroimage.2023.120417] [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: 04/17/2023] [Revised: 10/18/2023] [Accepted: 10/19/2023] [Indexed: 10/24/2023] Open
Abstract
fMRI of the human brain reveals spatiotemporal patterns of functional connectivity (FC), forming distinct cortical networks. Lately, subcortical contributions to these configurations are receiving renewed interest, but investigations rarely focus explicitly on their effects on cortico-cortical FC. Here, we employ a straightforward multivariable approach and graph-theoretic tools to assess subcortical impact on topological features of cortical networks. Given recent evidence showing that structures like the thalamus and basal ganglia integrate input from multiple networks, we expect increased segregation between cortical networks after removal of subcortical effects on their FC patterns. We analyze resting state data of young and healthy participants (male and female; N = 100) from the human connectome project. We find that overall, the cortical network architecture becomes less segregated, and more integrated, when subcortical influences are accounted for. Underlying these global effects are the following trends: 'Transmodal' systems become more integrated with the rest of the network, while 'unimodal' networks show the opposite effect. For single nodes this hierarchical organization is reflected by a close correspondence with the spatial layout of the principal gradient of FC (Margulies et al., 2016). Lastly, we show that the limbic system is significantly less coherent with subcortical influences removed. The findings are validated in a (split-sample) replication dataset. Our results provide new insight regarding the interplay between subcortex and cortical networks, by putting the integrative impact of subcortex in the context of macroscale patterns of cortical organization.
Collapse
Affiliation(s)
- Fabian Hirsch
- Department of Diagnostic and Interventional Neuroradiology, Klinikum R.d.Isar, Technical University Munich, Ismaninger Str. 22, Munich 81675, Germany.
| | - Afra Wohlschlaeger
- Department of Diagnostic and Interventional Neuroradiology, Klinikum R.d.Isar, Technical University Munich, Ismaninger Str. 22, Munich 81675, Germany
| |
Collapse
|
7
|
Hinkley LBN, Haas SS, Cheung SW, Nagarajan SS, Subramaniam K. Reduced neural connectivity in the caudate anterior head predicts hallucination severity in schizophrenia. Schizophr Res 2023; 261:1-5. [PMID: 37678144 PMCID: PMC10878029 DOI: 10.1016/j.schres.2023.08.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 06/13/2023] [Accepted: 08/31/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND Caudate functional abnormalities have been identified as one critical neural substrate underlying sensory gating impairments that lead to auditory phantom hallucinations in both patients with schizophrenia (SZ) and tinnitus, characterized by the perception of internally generated sounds in the absence of external environmental auditory stimuli. In this study, we tested the hypothesis as to whether functional connectivity abnormalities in distinct caudate subdivisions implicated in sensory gating and auditory phantom percepts in tinnitus, which are currently being localized for neuromodulation targeting using deep brain stimulation techniques, would be associated with auditory phantom hallucination severity in SZ. METHODS Twenty five SZ and twenty eight demographically-matched healthy control (HC) participants, completed this fMRI resting-state study and clinical assessments. RESULTS Between-group seed-to-voxel analyses revealed only one region, the caudate anterior head, which showed reduced functional connectivity with the thalamus that survived whole-brain multiple comparison corrections. Importantly, connectivity between the caudate anterior head with thalamus negatively correlated with hallucination severity. CONCLUSIONS In the present study, we deliver the first evidence of caudate subdivision specificity for the neural pathophysiology underlying hallucinations in schizophrenia within a sensory gating framework that has been developed for auditory phantoms in patients with tinnitus. Our findings provide transdiagnostic convergent evidence for the role of the caudate in the gating of auditory phantom hallucinations, observed across patients with SZ and tinnitus by specifying the anterior caudate division is key to mediation of hallucinations, and creating a path towards personalized treatment approaches to arrest auditory phantom hallucinations from reaching perceptual awareness.
Collapse
Affiliation(s)
- Leighton B N Hinkley
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94143, USA
| | - Shalaila S Haas
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY 10029, USA
| | - Steven W Cheung
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, CA 94143, USA; Surgical Services, San Francisco Veterans Health Care System, San Francisco, CA 94121, USA
| | - Srikantan S Nagarajan
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94143, USA
| | - Karuna Subramaniam
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA 94143, USA.
| |
Collapse
|
8
|
García Saborit M, Jara A, Muñoz N, Milovic C, Tepper A, Alliende LM, Mena C, Iruretagoyena B, Ramirez-Mahaluf JP, Diaz C, Nachar R, Castañeda CP, González A, Undurraga J, Crossley N, Tejos C. Quantitative Susceptibility Mapping MRI in Deep-Brain Nuclei in First-Episode Psychosis. Schizophr Bull 2023; 49:1355-1363. [PMID: 37030007 PMCID: PMC10483330 DOI: 10.1093/schbul/sbad041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/10/2023]
Abstract
BACKGROUND Psychosis is related to neurochemical changes in deep-brain nuclei, particularly suggesting dopamine dysfunctions. We used an magnetic resonance imaging-based technique called quantitative susceptibility mapping (QSM) to study these regions in psychosis. QSM quantifies magnetic susceptibility in the brain, which is associated with iron concentrations. Since iron is a cofactor in dopamine pathways and co-localizes with inhibitory neurons, differences in QSM could reflect changes in these processes. METHODS We scanned 83 patients with first-episode psychosis and 64 healthy subjects. We reassessed 22 patients and 21 control subjects after 3 months. Mean susceptibility was measured in 6 deep-brain nuclei. Using linear mixed models, we analyzed the effect of case-control differences, region, age, gender, volume, framewise displacement (FD), treatment duration, dose, laterality, session, and psychotic symptoms on QSM. RESULTS Patients showed a significant susceptibility reduction in the putamen and globus pallidus externa (GPe). Patients also showed a significant R2* reduction in GPe. Age, gender, FD, session, group, and region are significant predictor variables for QSM. Dose, treatment duration, and volume were not predictor variables of QSM. CONCLUSIONS Reduction in QSM and R2* suggests a decreased iron concentration in the GPe of patients. Susceptibility reduction in putamen cannot be associated with iron changes. Since changes observed in putamen and GPe were not associated with symptoms, dose, and treatment duration, we hypothesize that susceptibility may be a trait marker rather than a state marker, but this must be verified with long-term studies.
Collapse
Affiliation(s)
- Marisleydis García Saborit
- Department of Electrical Engineering, Pontificia Universidad Catolica de Chile, Santiago, Chile
- Biomedical Imaging Center, Pontificia Universidad Catolica de Chile, Santiago, Chile
- Millennium Institute for Intelligent Healthcare Engineering, Santiago, Chile
| | - Alejandro Jara
- Department of Statistics, Mathematics Faculty, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Néstor Muñoz
- Department of Electrical Engineering, Pontificia Universidad Catolica de Chile, Santiago, Chile
- Biomedical Imaging Center, Pontificia Universidad Catolica de Chile, Santiago, Chile
- Millennium Institute for Intelligent Healthcare Engineering, Santiago, Chile
| | - Carlos Milovic
- School of Electrical Engineering, Pontificia Universidad Catolica de Valparaiso, Valparaiso, Chile
| | - Angeles Tepper
- Millennium Institute for Intelligent Healthcare Engineering, Santiago, Chile
- Department of Psychiatry, School of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Luz María Alliende
- Department of Psychiatry, School of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Carlos Mena
- Department of Psychiatry, School of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Bárbara Iruretagoyena
- Department of Psychiatry, School of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | | | - Camila Diaz
- Pharmacovigilance, Instituto Psiquiátrico Dr. J. Horwitz Barak, Santiago, Chile
| | - Ruben Nachar
- Pharmacovigilance, Instituto Psiquiátrico Dr. J. Horwitz Barak, Santiago, Chile
| | | | - Alfonso González
- Early Intervention Program, Instituto Psiquiátrico Dr J. Horwitz Barak, Santiago, Chile
- School of Medicine, Universidad Finis Terrae, Santiago, Chile
| | - Juan Undurraga
- Early Intervention Program, Instituto Psiquiátrico Dr J. Horwitz Barak, Santiago, Chile
- Department of Neurology and Psychiatry, Faculty of Medicine, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - Nicolas Crossley
- Biomedical Imaging Center, Pontificia Universidad Catolica de Chile, Santiago, Chile
- Millennium Institute for Intelligent Healthcare Engineering, Santiago, Chile
- Department of Psychiatry, School of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Cristian Tejos
- Department of Electrical Engineering, Pontificia Universidad Catolica de Chile, Santiago, Chile
- Biomedical Imaging Center, Pontificia Universidad Catolica de Chile, Santiago, Chile
- Millennium Institute for Intelligent Healthcare Engineering, Santiago, Chile
| |
Collapse
|
9
|
Maury EA, Sherman MA, Genovese G, Gilgenast TG, Kamath T, Burris S, Rajarajan P, Flaherty E, Akbarian S, Chess A, McCarroll SA, Loh PR, Phillips-Cremins JE, Brennand KJ, Macosko EZ, Walters JT, O’Donovan M, Sullivan P, Sebat J, Lee EA, Walsh CA. Schizophrenia-associated somatic copy-number variants from 12,834 cases reveal recurrent NRXN1 and ABCB11 disruptions. CELL GENOMICS 2023; 3:100356. [PMID: 37601975 PMCID: PMC10435376 DOI: 10.1016/j.xgen.2023.100356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/21/2022] [Accepted: 06/09/2023] [Indexed: 08/22/2023]
Abstract
While germline copy-number variants (CNVs) contribute to schizophrenia (SCZ) risk, the contribution of somatic CNVs (sCNVs)-present in some but not all cells-remains unknown. We identified sCNVs using blood-derived genotype arrays from 12,834 SCZ cases and 11,648 controls, filtering sCNVs at loci recurrently mutated in clonal blood disorders. Likely early-developmental sCNVs were more common in cases (0.91%) than controls (0.51%, p = 2.68e-4), with recurrent somatic deletions of exons 1-5 of the NRXN1 gene in five SCZ cases. Hi-C maps revealed ectopic, allele-specific loops forming between a potential cryptic promoter and non-coding cis-regulatory elements upon 5' deletions in NRXN1. We also observed recurrent intragenic deletions of ABCB11, encoding a transporter implicated in anti-psychotic response, in five treatment-resistant SCZ cases and showed that ABCB11 is specifically enriched in neurons forming mesocortical and mesolimbic dopaminergic projections. Our results indicate potential roles of sCNVs in SCZ risk.
Collapse
Affiliation(s)
- Eduardo A. Maury
- Division of Genetics and Genomics, Manton Center for Orphan Disease, Boston Children’s Hospital, Boston, MA, USA
- Bioinformatics & Integrative Genomics Program and Harvard/MIT MD-PHD Program, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Maxwell A. Sherman
- Brigham and Women’s Hospital, Division of Genetics & Center for Data Sciences, Boston, MA, USA
| | - Giulio Genovese
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Thomas G. Gilgenast
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Tushar Kamath
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Graduate Program in Biophysics, Harvard University, Cambridge, MA, USA
| | - S.J. Burris
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Prashanth Rajarajan
- Nash Family Department of Neuroscience, Friedman Brain Institute, Department of Genetics & Genomics, Icahn Institute of Genomics and Multiscale Biology, Department of Psychiatry, Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine of Mount Sinai, New York, NY, USA
| | - Erin Flaherty
- Nash Family Department of Neuroscience, Friedman Brain Institute, Department of Genetics & Genomics, Icahn Institute of Genomics and Multiscale Biology, Department of Psychiatry, Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine of Mount Sinai, New York, NY, USA
| | - Schahram Akbarian
- Nash Family Department of Neuroscience, Friedman Brain Institute, Department of Genetics & Genomics, Icahn Institute of Genomics and Multiscale Biology, Department of Psychiatry, Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine of Mount Sinai, New York, NY, USA
| | - Andrew Chess
- Nash Family Department of Neuroscience, Friedman Brain Institute, Department of Genetics & Genomics, Icahn Institute of Genomics and Multiscale Biology, Department of Psychiatry, Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine of Mount Sinai, New York, NY, USA
| | - Steven A. McCarroll
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Po-Ru Loh
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Brigham and Women’s Hospital, Division of Genetics & Center for Data Sciences, Boston, MA, USA
| | | | - Kristen J. Brennand
- Nash Family Department of Neuroscience, Friedman Brain Institute, Department of Genetics & Genomics, Icahn Institute of Genomics and Multiscale Biology, Department of Psychiatry, Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine of Mount Sinai, New York, NY, USA
- Departments of Psychiatry and Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Evan Z. Macosko
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Massachusetts General Hospital, Department of Psychiatry, Boston, MA, USA
| | - James T.R. Walters
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychiatry and Clinical Neurosciences, Cardiff University, Cardiff, Wales
| | - Michael O’Donovan
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychiatry and Clinical Neurosciences, Cardiff University, Cardiff, Wales
| | - Patrick Sullivan
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jonathan Sebat
- University of California San Diego, Department of Psychiatry, Department of Cellular & Molecular Medicine, Beyster Center of Psychiatric Genomics, San Diego, CA, USA
| | - Eunjung A. Lee
- Division of Genetics and Genomics, Manton Center for Orphan Disease, Boston Children’s Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Christopher A. Walsh
- Division of Genetics and Genomics, Manton Center for Orphan Disease, Boston Children’s Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, MA, USA
| |
Collapse
|
10
|
Roberts D, Rösler L, Wijnen JP, Thakkar KN. Associations between N-Acetylaspartate and white matter integrity in individuals with schizophrenia and unaffected relatives. Psychiatry Res Neuroimaging 2023; 330:111612. [PMID: 36805928 PMCID: PMC10023491 DOI: 10.1016/j.pscychresns.2023.111612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 01/27/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023]
Abstract
Compromised white matter has been reported in schizophrenia; however, few studies have investigated neurochemical abnormalities underlying microstructural differences. N-acetylaspartate (NAA) is used to synthesize myelin and is often reduced in persons with schizophrenia (PSZ) and their unaffected first-degree relatives (REL). Low levels of NAA could affect white matter by preventing the synthesis or repair of myelin. We used magnetic resonance spectroscopy and diffusion tensor imaging to investigate the relationship between NAA and white matter integrity in PSZ. REL were included to examine whether putative relationships are associated with symptom expression or illness liability. 52 controls, 23 REL and 25 PSZ underwent 7T proton magnetic resonance spectroscopy and/or 3T diffusion tensor imaging. NAA in the visual cortex and basal ganglia were measured and compared across groups. Diffusivity measures were compared across groups using tract-based spatial statistics and related to NAA concentrations. Visual cortex NAA was significantly reduced in PSZ compared to controls. White matter integrity did not differ between groups. Reduced cortical and subcortical NAA were associated with diffusivity measures of poor white matter microstructure. These data suggest that levels of neural NAA may be related to white matter integrity similarly across individuals with schizophrenia, those at genetic risk, and controls.
Collapse
Affiliation(s)
- Dominic Roberts
- Department of Psychology, Michigan State University, East Lansing, MI, United States
| | - Lara Rösler
- Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Jannie P Wijnen
- Department of Radiology, High Field MR Research, Centre for Image Sciences, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Katharine N Thakkar
- Department of Psychology, Michigan State University, East Lansing, MI, United States; Department of Psychiatry and Behavioral Medicine, Michigan State University, East Lansing, Michigan, United States.
| |
Collapse
|
11
|
van Hooijdonk CFM, van der Pluijm M, Bosch I, van Amelsvoort TAMJ, Booij J, de Haan L, Selten JP, Giessen EVD. The substantia nigra in the pathology of schizophrenia: A review on post-mortem and molecular imaging findings. Eur Neuropsychopharmacol 2023; 68:57-77. [PMID: 36640734 DOI: 10.1016/j.euroneuro.2022.12.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 12/19/2022] [Accepted: 12/21/2022] [Indexed: 01/14/2023]
Abstract
Dysregulation of striatal dopamine is considered to be an important driver of pathophysiological processes in schizophrenia. Despite being one of the main origins of dopaminergic input to the striatum, the (dys)functioning of the substantia nigra (SN) has been relatively understudied in schizophrenia. Hence, this paper aims to review different molecular aspects of nigral functioning in patients with schizophrenia compared to healthy controls by integrating post-mortem and molecular imaging studies. We found evidence for hyperdopaminergic functioning in the SN of patients with schizophrenia (i.e. increased AADC activity in antipsychotic-free/-naïve patients and elevated neuromelanin accumulation). Reduced GABAergic inhibition (i.e. decreased density of GABAergic synapses, lower VGAT mRNA levels and lower mRNA levels for GABAA receptor subunits), excessive glutamatergic excitation (i.e. increased NR1 and Glur5 mRNA levels and a reduced number of astrocytes), and several other disturbances implicating the SN (i.e. immune functioning and copper concentrations) could potentially underlie this nigral hyperactivity and associated striatal hyperdopaminergic functioning in schizophrenia. These results highlight the importance of the SN in schizophrenia pathology and suggest that some aspects of molecular functioning in the SN could potentially be used as treatment targets or biomarkers.
Collapse
Affiliation(s)
- Carmen F M van Hooijdonk
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), University of Maastricht, Maastricht, the Netherlands; Rivierduinen, Institute for Mental Health Care, Leiden, the Netherlands.
| | - Marieke van der Pluijm
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Iris Bosch
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Therese A M J van Amelsvoort
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), University of Maastricht, Maastricht, the Netherlands
| | - Jan Booij
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Lieuwe de Haan
- Department of Psychiatry, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Jean-Paul Selten
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), University of Maastricht, Maastricht, the Netherlands; Rivierduinen, Institute for Mental Health Care, Leiden, the Netherlands
| | - Elsmarieke van de Giessen
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, the Netherlands
| |
Collapse
|
12
|
Giordano GM, Pezzella P, Giuliani L, Fazio L, Mucci A, Perrottelli A, Blasi G, Amore M, Rocca P, Rossi A, Bertolino A, Galderisi S. Resting-State Brain Activity Dysfunctions in Schizophrenia and Their Associations with Negative Symptom Domains: An fMRI Study. Brain Sci 2023; 13:brainsci13010083. [PMID: 36672064 PMCID: PMC9856573 DOI: 10.3390/brainsci13010083] [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: 11/21/2022] [Revised: 12/07/2022] [Accepted: 12/22/2022] [Indexed: 01/03/2023] Open
Abstract
The aim of the present study was to examine the neurobiological correlates of the two negative symptom domains of schizophrenia, the Motivational Deficit domain (including avolition, anhedonia, and asociality) and the Expressive Deficit domain (including blunted affect and alogia), focusing on brain areas that are most commonly found to be associated with negative symptoms in previous literature. Resting-state (rs) fMRI data were analyzed in 62 subjects affected by schizophrenia (SZs) and 46 healthy controls (HCs). The SZs, compared to the HCs, showed higher rs brain activity in the right inferior parietal lobule and the right temporoparietal junction, and lower rs brain activity in the right dorsolateral prefrontal cortex, the bilateral anterior dorsal cingulate cortex, and the ventral and dorsal caudate. Furthermore, in the SZs, the rs brain activity in the left orbitofrontal cortex correlated with negative symptoms (r = -0.436, p = 0.006), in particular with the Motivational Deficit domain (r = -0.424, p = 0.002), even after controlling for confounding factors. The left ventral caudate correlated with negative symptoms (r = -0.407, p = 0.003), especially with the Expressive Deficit domain (r = -0.401, p = 0.003); however, these results seemed to be affected by confounding factors. In line with the literature, our results demonstrated that the two negative symptom domains might be underpinned by different neurobiological mechanisms.
Collapse
Affiliation(s)
- Giulia Maria Giordano
- Department of Psychiatry, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Pasquale Pezzella
- Department of Psychiatry, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Luigi Giuliani
- Department of Psychiatry, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
- Correspondence: ; Tel.: +39-0815666512
| | - Leonardo Fazio
- Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro’, 70124 Bari, Italy
- Department of Medicine and Surgery, LUM University, 70010 Casamassima, Italy
| | - Armida Mucci
- Department of Psychiatry, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Andrea Perrottelli
- Department of Psychiatry, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Giuseppe Blasi
- Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro’, 70124 Bari, Italy
| | - Mario Amore
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics and Maternal and Child Health, Section of Psychiatry, University of Genoa, 16132 Genoa, Italy
| | - Paola Rocca
- Department of Neuroscience, Section of Psychiatry, University of Turin, 10126 Turin, Italy
| | - Alessandro Rossi
- Department of Biotechnological and Applied Clinical Sciences, Section of Psychiatry, University of L’Aquila, 67100 L’Aquila, Italy
| | - Alessandro Bertolino
- Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro’, 70124 Bari, Italy
| | - Silvana Galderisi
- Department of Psychiatry, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | | |
Collapse
|
13
|
Alhassen W, Alhassen S, Chen J, Monfared RV, Alachkar A. Cilia in the Striatum Mediate Timing-Dependent Functions. Mol Neurobiol 2023; 60:545-565. [PMID: 36322337 PMCID: PMC9849326 DOI: 10.1007/s12035-022-03095-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 10/16/2022] [Indexed: 11/07/2022]
Abstract
Almost all brain cells contain cilia, antennae-like microtubule-based organelles. Yet, the significance of cilia, once considered vestigial organelles, in the higher-order brain functions is unknown. Cilia act as a hub that senses and transduces environmental sensory stimuli to generate an appropriate cellular response. Similarly, the striatum, a brain structure enriched in cilia, functions as a hub that receives and integrates various types of environmental information to drive appropriate motor response. To understand cilia's role in the striatum functions, we used loxP/Cre technology to ablate cilia from the dorsal striatum of male mice and monitored the behavioral consequences. Our results revealed an essential role for striatal cilia in the acquisition and brief storage of information, including learning new motor skills, but not in long-term consolidation of information or maintaining habitual/learned motor skills. A fundamental aspect of all disrupted functions was the "time perception/judgment deficit." Furthermore, the observed behavioral deficits form a cluster pertaining to clinical manifestations overlapping across psychiatric disorders that involve the striatum functions and are known to exhibit timing deficits. Thus, striatal cilia may act as a calibrator of the timing functions of the basal ganglia-cortical circuit by maintaining proper timing perception. Our findings suggest that dysfunctional cilia may contribute to the pathophysiology of neuro-psychiatric disorders, as related to deficits in timing perception.
Collapse
Affiliation(s)
- Wedad Alhassen
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California-Irvine, 356A Med Surge II, Irvine, CA 92697-4625 USA
| | - Sammy Alhassen
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California-Irvine, 356A Med Surge II, Irvine, CA 92697-4625 USA
| | - Jiaqi Chen
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California-Irvine, 356A Med Surge II, Irvine, CA 92697-4625 USA
| | - Roudabeh Vakil Monfared
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California-Irvine, 356A Med Surge II, Irvine, CA 92697-4625 USA
| | - Amal Alachkar
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California-Irvine, 356A Med Surge II, Irvine, CA 92697-4625 USA ,UC Irvine Center for the Neurobiology of Learning and Memory, University of California-Irvine, Irvine, CA 92697 USA ,Institute for Genomics and Bioinformatics, School of Information and Computer Sciences, University of California-Irvine, Irvine, CA 92697 USA
| |
Collapse
|
14
|
Al-Nema M, Gaurav A, Lee MT, Okechukwu P, Nimmanpipug P, Lee VS. Evaluation of the acute oral toxicity and antipsychotic activity of a dual inhibitor of PDE1B and PDE10A in rat model of schizophrenia. PLoS One 2022; 17:e0278216. [PMID: 36454774 PMCID: PMC9714703 DOI: 10.1371/journal.pone.0278216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 11/11/2022] [Indexed: 12/03/2022] Open
Abstract
Phosphodiesterase 1B (PDE1B) and PDE10A are dual-specificity PDEs that hydrolyse both cyclic adenosine monophosphate and cyclic guanosine monophosphate, and are highly expressed in the striatum. Several reports have suggested that PDE10A inhibitors may present a promising approach for the treatment of positive symptoms of schizophrenia, whereas PDE1B inhibitors may present a novel mechanism to modulate cognitive deficits. Previously, we have reported a novel dual inhibitor of PDE1B and PDE10A, compound 2 [(3-fluorophenyl)(2-methyl-2,3-dihydro-4H-benzo[b][1,4]oxazin-4-yl)methanone] which has shown inhibitory activity for human recombinant PDE1B and PDE10A in vitro. In the present study, the safety profile of compound 2 has been evaluated in rats in the acute oral toxicity study, as well as; the antipsychotic-like effects in the rat model of schizophrenia. Compound 2 was tolerated up to 1 g/kg when administered at a single oral dose. Additionally, compound 2 has strongly suppressed ketamine-induced hyperlocomotion, which presented a model for the positive symptoms of schizophrenia. It has also shown an ability to attenuate social isolation induced by chronic administration of ketamine and enhanced recognition memory of rats in the novel object recognition test. Altogether, our results suggest that compound 2 represents a promising therapy for the treatment of the three symptomatic domains of schizophrenia.
Collapse
Affiliation(s)
- Mayasah Al-Nema
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia
| | - Anand Gaurav
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia
- * E-mail: (AG); (VSL)
| | - Ming Tatt Lee
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia
- Office of Postgraduate Studies, UCSI University, Kuala Lumpur, Malaysia
- Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Patrick Okechukwu
- Faculty of Applied Sciences, UCSI University, Kuala Lumpur, Malaysia
| | - Piyarat Nimmanpipug
- Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence for Innovation in Analytical Science and Technology for Biodiversity-based Economic and Society (I-ANALY-S-T), Chiang Mai University, Chiang Mai, Thailand
| | - Vannajan Sanghiran Lee
- Department of Chemistry, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
- * E-mail: (AG); (VSL)
| |
Collapse
|
15
|
Regional Cerebral Blood Flow Correlates of Neuropsychiatric Symptom Domains in Early Alzheimer’s Disease. Diagnostics (Basel) 2022; 12:diagnostics12051246. [PMID: 35626401 PMCID: PMC9140211 DOI: 10.3390/diagnostics12051246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/13/2022] [Accepted: 05/16/2022] [Indexed: 12/10/2022] Open
Abstract
Although various neuropsychiatric symptoms are frequently accompanied with Alzheimer’s disease (AD) and pose a substantial burden to both patients and caregivers, their neurobiological underpinnings remain unclear. This study investigated associations between regional cerebral blood flow (rCBF) and neuropsychiatric symptom domains in early AD. A total of 59 patients with early AD underwent brain technetium-99m hexamethylpropylene amine oxime (99mTc-HMPAO) single-photon emission computed tomography (SPECT) scans. Neuropsychiatric symptoms were assessed by the Neuropsychiatric Inventory and clustered into the affective, apathy, hyperactivity, and psychotic domains. A voxel-wise multiple regression analysis was performed with four domain scores as independent variables and age, sex, and Mini-Mental State Examination scores as covariates. The affective domain score was negatively correlated with rCBF in the prefrontal cortex, thalamus, and caudate. The apathy domain score showed inverse correlations with rCBF in the prefrontal and pre/postcentral gyri and midbrain. Patients with higher hyperactivity domain scores had increased rCBF in the prefrontal and temporal lobes. The psychotic symptom domain was positively correlated with rCBF in the cuneus and negatively associated with rCBF in the prefrontal, cingulate, and occipital regions and putamen. The score of each neuropsychiatric symptom domain showed the differential correlates of brain perfusion, while altered rCBF in the prefrontal cortex was found in all domains. Although preliminary, our results may suggest common and distinct patterns of rCBF underlying neuropsychiatric symptoms in early AD. Further studies with larger samples and control participants are warranted to confirm these findings.
Collapse
|
16
|
Phillips RA, Tuscher JJ, Black SL, Andraka E, Fitzgerald ND, Ianov L, Day JJ. An atlas of transcriptionally defined cell populations in the rat ventral tegmental area. Cell Rep 2022; 39:110616. [PMID: 35385745 PMCID: PMC10888206 DOI: 10.1016/j.celrep.2022.110616] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 12/28/2021] [Accepted: 03/11/2022] [Indexed: 01/06/2023] Open
Abstract
The ventral tegmental area (VTA) is a complex brain region that is essential for reward function and frequently implicated in neuropsychiatric disease. While decades of research on VTA function have focused on dopamine neurons, recent evidence has identified critical roles for GABAergic and glutamatergic neurons in reward processes. Additionally, although subsets of VTA neurons express genes involved in the synthesis and transport of multiple neurotransmitters, characterization of these combinatorial populations has largely relied on low-throughput methods. To comprehensively define the molecular architecture of the VTA, we performed single-nucleus RNA sequencing on 21,600 cells from the rat VTA. Analysis of neuronal subclusters identifies selective markers for dopamine and combinatorial neurons, reveals expression profiles for receptors targeted by drugs of abuse, and demonstrates population-specific enrichment of gene sets linked to brain disorders. These results highlight the heterogeneity of the VTA and provide a resource for further exploration of VTA gene expression.
Collapse
Affiliation(s)
- Robert A Phillips
- Department of Neurobiology & Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jennifer J Tuscher
- Department of Neurobiology & Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Samantha L Black
- Department of Neurobiology & Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Emma Andraka
- Department of Neurobiology & Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - N Dalton Fitzgerald
- Department of Neurobiology & Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Lara Ianov
- Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jeremy J Day
- Department of Neurobiology & Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
17
|
Nikvarz N, Sabouri S. Drug-induced stuttering: A comprehensive literature review. World J Psychiatry 2022; 12:236-263. [PMID: 35317340 PMCID: PMC8900588 DOI: 10.5498/wjp.v12.i2.236] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 07/29/2021] [Accepted: 12/25/2021] [Indexed: 02/06/2023] Open
Abstract
Drug-induced stuttering (DIS) is a type of neurogenic stuttering (NS). Although DIS has not been reported as frequently as other cases of NS in the literature, it is not a negligible adverse drug reaction (ADR) which can significantly affect the quality of life if not treated. This literature review aims to evaluate the epidemiological and clinical characteristics of DIS and suggests some pathophysiological mechanisms for this ADR. Relevant English-language reports in Google Scholar, PubMed, Web of Science, and Scopus were identified and assessed without time restriction. Finally, a total of 62 reports were included. Twenty-seven drugs caused 86 episodes of stuttering in 82 cases. The most episodes of DIS were related to antipsychotic drugs (57%), mostly including clozapine, followed by central nervous system agents (11.6%) and anticonvulsant drugs (9.3%). The majority of the cases were male and between the ages of 31 and 40 years. Repetitions were the most frequent core manifestations of DIS. In 55.8% of the episodes of DIS, the offending drug was withdrawn to manage stuttering, which resulted in significant improvement or complete relief of stuttering in all cases. Based on the suggested pathophysiological mechanisms for developmental stuttering and neurotransmitters dysfunctions involved in speech dysfluency, it seems that the abnormalities of several neurotransmitters, especially dopamine and glutamate, in different circuits and areas of the brain, including cortico-basal ganglia-thalamocortical loop and white matter fiber tracts, may be engaged in the pathogenesis of DIS.
Collapse
Affiliation(s)
- Naemeh Nikvarz
- Department of Clinical Pharmacy, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman 7616911319, Iran
| | - Salehe Sabouri
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman 7616911319, Iran
- Department of Pharmaceutical Biotechnology, Kerman University of Medical Sciences, Kerman 7616911319, Iran
| |
Collapse
|
18
|
Chen CH, Leu SJJ, Hsu CP, Pan CC, Shyue SK, Lee TS. Atypical antipsychotic drugs deregulate the cholesterol metabolism of macrophage-foam cells by activating NOX-ROS-PPARγ-CD36 signaling pathway. Metabolism 2021; 123:154847. [PMID: 34364926 DOI: 10.1016/j.metabol.2021.154847] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 08/02/2021] [Accepted: 08/03/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND Clinical reports indicate that schizophrenia patients taking atypical antipsychotic drugs suffer from metabolism diseases including atherosclerosis. However, the mechanisms underlying the detrimental effect of atypical antipsychotic drugs on atherosclerosis remain to be explored. METHODS In this study, we used apolipoprotein E-deficient (apoe-/-) hyperlipidemic mice and apoe-/-cd36-/- mice to investigate the underlying mechanism of atypical antipsychotic drugs on atherosclerosis and macrophage-foam cells. RESULTS In vivo studies showed that genetic deletion of cd36 gene ablated the pro-atherogenic effect of olanzapine in apoe-/- mice. Moreover, in vitro studies revealed that genetic deletion or siRNA-mediated knockdown of cd36 or pharmacological inhibition of CD36 prevented atypical antipsychotic drugs-induced oxLDL accumulation in macrophages. Additionally, olanzapine and clozapine activated NADPH oxidase (NOX) to generate reactive oxygen species (ROS) which upregulated the activity of peroxisome proliferator-activated receptor γ (PPARγ) and subsequently elevated CD36 expression. Inhibition of NOX activity, ROS production or PPARγ activity suppressed CD36 expression and abolished the detrimental effects of olanzapine and clozapine on oxLDL accumulation in macrophages. CONCLUSION Collectively, our results suggest that atypical antipsychotic drugs exacerbate atherosclerosis and macrophage-foam cell formation by activating the NOX-ROS-PPARγ-CD36 pathway.
Collapse
Affiliation(s)
- Chia-Hui Chen
- Graduate Institute and Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shr-Jeng Jim Leu
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chiao-Po Hsu
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan; Division of Cardiovascular Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ching-Chian Pan
- Cardiovascular Division, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Song-Kun Shyue
- Cardiovascular Division, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
| | - Tzong-Shyuan Lee
- Graduate Institute and Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
19
|
Al-Nema MY, Gaurav A. Phosphodiesterase as a Target for Cognition Enhancement in Schizophrenia. Curr Top Med Chem 2021; 20:2404-2421. [PMID: 32533817 DOI: 10.2174/1568026620666200613202641] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 05/04/2020] [Accepted: 05/08/2020] [Indexed: 12/20/2022]
Abstract
Schizophrenia is a severe mental disorder that affects more than 1% of the population worldwide. Dopamine system dysfunction and alterations in glutamatergic neurotransmission are strongly implicated in the aetiology of schizophrenia. To date, antipsychotic drugs are the only available treatment for the symptoms of schizophrenia. These medications, which act as D2-receptor antagonist, adequately address the positive symptoms of the disease, but they fail to improve the negative symptoms and cognitive impairment. In schizophrenia, cognitive impairment is a core feature of the disorder. Therefore, the treatment of cognitive impairment and the other symptoms related to schizophrenia remains a significant unmet medical need. Currently, phosphodiesterases (PDEs) are considered the best drug target for the treatment of schizophrenia since many PDE subfamilies are abundant in the brain regions that are relevant to cognition. Thus, this review aims to illustrate the mechanism of PDEs in treating the symptoms of schizophrenia and summarises the encouraging results of PDE inhibitors as anti-schizophrenic drugs in preclinical and clinical studies.
Collapse
Affiliation(s)
- Mayasah Y Al-Nema
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia
| | - Anand Gaurav
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia
| |
Collapse
|
20
|
Casquero-Veiga M, Romero-Miguel D, MacDowell KS, Torres-Sanchez S, Garcia-Partida JA, Lamanna-Rama N, Gómez-Rangel V, Romero-Miranda A, Berrocoso E, Leza JC, Arango C, Desco M, Soto-Montenegro ML. Omega-3 fatty acids during adolescence prevent schizophrenia-related behavioural deficits: Neurophysiological evidences from the prenatal viral infection with PolyI:C. Eur Neuropsychopharmacol 2021; 46:14-27. [PMID: 33735708 DOI: 10.1016/j.euroneuro.2021.02.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 01/11/2021] [Accepted: 02/09/2021] [Indexed: 12/20/2022]
Abstract
The likely involvement of inflammation and oxidative stress (IOS) in mental disease has led to advocate anti-oxidant and anti-inflammatory drugs as therapeutic strategies in the treatment of schizophrenia. Since omega-3 fatty acids (ω-3) show anti-inflammatory/neuroprotective properties, we aim to evaluate whether ω-3 treatment during adolescence in the maternal immune stimulation (MIS) animal model of schizophrenia could prevent the brain and behavioural deficits described in adulthood. At gestational day 15, PolyI:C (4 mg/kg) or saline (VH) were injected to pregnant Wistar rats. Male offspring received ω-3 (800 mg/kg) or saline (Sal) daily from postnatal day (PND) 35-49, defining 4 groups: MIS-ω-3; MIS-Sal; VH-ω-3 and VH-Sal. At PND70, rats were submitted to prepulse inhibition test (PPI). FDG-PET and T2-weighted MRI brain studies were performed in adulthood and analyzed by means of SPM12. IOS markers were measured in selected brain areas. MIS-offspring showed a PPI deficit compared with VH-offspring and ω-3 treatment prevented this deficit. Also, ω-3 reduced the brain metabolism in the deep mesencephalic area and prevented the volumetric abnormalities in the hippocampus but not in the ventricles in MIS-offspring. Besides, ω-3 reduced the expression of iNOS and Keap1 and increased the activity/concentration of HO1, NQO1 and GPX. Our study demonstrates that administration of ω-3 during adolescence prevents PPI behavioural deficits and hippocampal volumetric abnormalities, and partially counteracts IOS deficits via iNOS and Nrf2-ARE pathways in the MIS model. This study highlights the need for novel strategies based on anti-inflammatory/anti-oxidant compounds to alter the disease course in high-risk populations at early stages.
Collapse
Affiliation(s)
- Marta Casquero-Veiga
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; CIBER de Salud Mental (CIBERSAM), Madrid, Spain
| | | | - Karina S MacDowell
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Department of Pharmacology & Toxicology, School of Medicine, University Complutense (UCM), IIS Imas12, IUIN, Madrid, Spain
| | - Sonia Torres-Sanchez
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Neuropsychopharmacology & Psychobiology Research Group, Psychobiology Area, Department of Psychology, University of Cádiz, Puerto Real (Cádiz), Spain; Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Cádiz, Spain
| | - Jose Antonio Garcia-Partida
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Cádiz, Spain; Neuropsychopharmacology & Psychobiology Research Group, Department of Neuroscience, University of Cádiz, Cádiz, Spain
| | | | | | | | - Esther Berrocoso
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Neuropsychopharmacology & Psychobiology Research Group, Psychobiology Area, Department of Psychology, University of Cádiz, Puerto Real (Cádiz), Spain; Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Cádiz, Spain
| | - Juan C Leza
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Department of Pharmacology & Toxicology, School of Medicine, University Complutense (UCM), IIS Imas12, IUIN, Madrid, Spain
| | - Celso Arango
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Department of Child and Adolescent Psychiatry, Institute of Psychiatry and Mental Health, Hospital General Universitario Gregorio Marañón, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Manuel Desco
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Departamento of Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, Leganés, Spain; Centro Nacional de Investigaciones Cardiovasculares, CNIC, Madrid, Spain.
| | - María Luisa Soto-Montenegro
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; CIBER de Salud Mental (CIBERSAM), Madrid, Spain.
| |
Collapse
|
21
|
Hare SM, Du X, Adhikari BM, Chen S, Mo C, Summerfelt A, Kvarta MD, Garcia L, Kochunov P, Elliot Hong L. Mapping local and long-distance resting connectivity markers of TMS-related inhibition reduction in schizophrenia. NEUROIMAGE-CLINICAL 2021; 31:102688. [PMID: 33991855 PMCID: PMC8135038 DOI: 10.1016/j.nicl.2021.102688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 03/07/2021] [Accepted: 04/26/2021] [Indexed: 11/17/2022]
Abstract
Short interval intracortical inhibition (SICI) is a biomarker for altered motor inhibition in schizophrenia, but the manner in which distant sites influence the inhibitory cortical-effector response remains elusive. Our study investigated local and long-distance resting state functional connectivity (rsFC) markers of SICI in a sample of N = 23 patients with schizophrenia and N = 29 controls. Local functional connectivity was quantified using regional homogeneity (ReHo) analysis and long-range connectivity was estimated using seed-based rsFC analysis. Direct and indirect effects of connectivity measures on SICI were modeled using mediation analysis. Higher SICI ratios (indicating reduced inhibition) in patients were associated with lower ReHo in the right insula. Follow-up rsFC analyses showed that higher SICI scores (indicating reduced inhibition) were associated with reduced connectivity between right insula and hubs of the corticospinal pathway: sensorimotor cortex and basal ganglia. Mediation analysis supported a model in which the direct effect of local insular connectivity strength on SICI is mediated by the interhemispheric connectivity between insula and left sensorimotor cortex. The broader clinical implications of these findings are discussed with emphasis on how these preliminary findings might inform novel interventions designed to restore or improve SICI in schizophrenia and deepen our understanding of motor inhibitory control and impact of abnormal signaling in motor-inhibitory pathways in schizophrenia.
Collapse
Affiliation(s)
- Stephanie M Hare
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States.
| | - Xiaoming Du
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Bhim M Adhikari
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Shuo Chen
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Chen Mo
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Ann Summerfelt
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Mark D Kvarta
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Laura Garcia
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Peter Kochunov
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States
| | - L Elliot Hong
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
22
|
Zhang T, Song J, Chen C, Li R, Li Y, Sun Y, Fang T, Xu W, Tian H, Zhuo C. Brain features of nearly drug-naïve female monozygotic twins with first-episode schizophrenia and the classification accuracy of brain feature patterns: A pilot study. Brain Behav 2021; 11:e01992. [PMID: 33295156 PMCID: PMC7882158 DOI: 10.1002/brb3.1992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 11/13/2020] [Accepted: 11/23/2020] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Data on differences in brain features between monozygotic (MZ) twins with and without schizophrenia are scarce. METHODS We compared brain features of female MZ twins with and without first-episode schizophrenia and healthy controls (n = 20 each). Voxel-based morphometry and tract-based spatial statistics were used to analyze differences in brain structure. Whole-brain effective connectivity (EC) and functional connectivity (FC) networks were constructed using resting-state functional magnetic resonance imaging (rs-fMRI) data. RESULTS Female twins with schizophrenia exhibited abnormal gray matter volume (GMV) in the basal ganglia and prefrontal and parietal cortices, impairments in the arcuate fasciculus, and significant disruptions (primarily decreases) in nine EC networks. They exhibited rs-EC alterations involving the limbic areas and subcortex. Combined rs-EC and rs-FC data distinguished twins with first-episode schizophrenia with high accuracy. Combined consideration of structural and functional features enabled the distinction of female MZ twins with schizophrenia from those without schizophrenia and healthy controls with 100% accuracy. CONCLUSIONS Female MZ twins with schizophrenia exhibited increased GMV, white matter impairment, and disruptions in EC and FC networks. The combination of rs-EC + rs-FC data could distinguish female twins with schizophrenia from twins without schizophrenia and healthy controls with 97.4% accuracy, and the addition of structural brain features yielded a 100% accuracy rate. These findings may provide pivotal insight for further study of the mechanisms underlying schizophrenia.
Collapse
Affiliation(s)
- Tao Zhang
- Department of PsychiatryDongying Shengli HospitalDongyingChina
| | - Jie Song
- Department of PsychiatryShanghai Qingpu District Mental Health CenterShanghaiChina
| | - Ce Chen
- Department of PsychiatryWenzhou Seventh HospitalWenzhouChina
| | - Ran Li
- Psychiatric‐Neuroimaging‐Genetics and Comorbidity LaboratoryTianjin Mental Health CentreTianjin Anding HospitalTianjin Medical University Mental Health Teaching HospitalTianjinChina
- Department of PsychiatryTianjin Medical UniversityTianjinChina
| | - Yachen Li
- Psychiatric‐Neuroimaging‐Genetics and Comorbidity LaboratoryTianjin Mental Health CentreTianjin Anding HospitalTianjin Medical University Mental Health Teaching HospitalTianjinChina
- Department of PsychiatryTianjin Medical UniversityTianjinChina
| | - Yun Sun
- Psychiatric‐Neuroimaging‐Genetics and Comorbidity LaboratoryTianjin Mental Health CentreTianjin Anding HospitalTianjin Medical University Mental Health Teaching HospitalTianjinChina
- Department of PsychiatryTianjin Medical UniversityTianjinChina
| | - Tao Fang
- Key Laboratory of Sensory Information Processing Abnormalities in Schizophrenia (SIPP_Lab)Tianjin Fourth Center HospitalTianjin Fourth Center Hospital Affiliated to Nankai UniversityTianjinChina
| | - Weiwei Xu
- Department of PsychiatryDongying Shengli HospitalDongyingChina
| | - Hongjun Tian
- Department of PsychiatryTianjin Medical UniversityTianjinChina
- Key Laboratory of Sensory Information Processing Abnormalities in Schizophrenia (SIPP_Lab)Tianjin Fourth Center HospitalTianjin Fourth Center Hospital Affiliated to Nankai UniversityTianjinChina
| | - Chuanjun Zhuo
- Department of PsychiatryWenzhou Seventh HospitalWenzhouChina
- Psychiatric‐Neuroimaging‐Genetics and Comorbidity LaboratoryTianjin Mental Health CentreTianjin Anding HospitalTianjin Medical University Mental Health Teaching HospitalTianjinChina
- Department of PsychiatryTianjin Medical UniversityTianjinChina
- Key Laboratory of Sensory Information Processing Abnormalities in Schizophrenia (SIPP_Lab)Tianjin Fourth Center HospitalTianjin Fourth Center Hospital Affiliated to Nankai UniversityTianjinChina
| |
Collapse
|
23
|
Szűcs E, Ducza E, Büki A, Kekesi G, Benyhe S, Horvath G. Characterization of dopamine D2 receptor binding, expression and signaling in different brain regions of control and schizophrenia-model Wisket rats. Brain Res 2020; 1748:147074. [DOI: 10.1016/j.brainres.2020.147074] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/21/2020] [Accepted: 08/18/2020] [Indexed: 01/01/2023]
|
24
|
Mujittapha SU, Kauthar M, Azeez IO, Oyem JC. Ascorbic acid improves extrapyramidal syndromes and corpus striatal degeneration induced by dopamine-2 receptor inhibition in Wistar rats. Drug Metab Pers Ther 2020; 0:/j/dmdi.ahead-of-print/dmdi-2020-0137/dmdi-2020-0137.xml. [PMID: 33125337 DOI: 10.1515/dmdi-2020-0137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 08/24/2020] [Indexed: 11/15/2022]
Abstract
Objectives The prolonged uses of fourth-generation antipsychotics have been implicated in inducing extrapyramidal syndromes characterized by the motor deficit. This was attributed to the loss of dopamine-2 receptor (D2R) signaling. However, ascorbic acid (SVCT2R stimulation) in the brain is proposed to modulate D2R activity. We, therefore, investigated the beneficial roles of ascorbic acid in improving the extrapyramidal symptoms seen in D2R loss. Methods Twenty adult male Wistar rats of average weight 200 g were distributed randomly into four groups. The control (NS) received normal saline for 28 days, Untreated D2R inhibition group (-D2R) received normal saline for seven days and then subsequently received chlorpromazine for 21 days, D2R inhibition group treated with ascorbic acid (-D2R+SVCT2R) received chlorpromazine for 21 days and was subsequently treated with ascorbate for seven days while the withdrawal group (WG) received chlorpromazine for 21 days and subsequently received normal saline for seven days. Motor deficits were assessed using a rotarod and cylinder test. The corpus striatum was harvested, processed, and stained using H&E and Nissl stains. Cellular density was analyzed using Image J software 1.8.0. Results Motor deficit was observed in -D2R animals administered chlorpromazine with less improvement in WG compared to control (p<0.05) in both rotarod and cylinder test. Ascorbic acid (SVCT2R stimulation) significantly (p<0.001) improved the latency of fall and climbing attempts observed in -D2R animals. The density of basophilic trigoid bodies was significantly (p<0.001) restored in -D2R+SVCT2R group, suggesting recovery of neural activity in the corpus striatum. Moreover, the hallmarks of neuronal degeneration were less expressed in the ascorbic acid treatment groups. Conclusions Ascorbic acid putatively ameliorates extrapyramidal symptoms observed in D2R blockage by chlorpromazine in Wistar rats.
Collapse
Affiliation(s)
| | - Murtala Kauthar
- Department of Medical Laboratory, Ahmadu Bello University, Kano, Nigeria
| | - Ishola O Azeez
- Department of Human Anatomy, Afe Babalola University, Ado-Ekiti, Nigeria
| | - John C Oyem
- Department of Anatomy, University of Port Harcourt, Port Harcourt, Nigeria
| |
Collapse
|
25
|
Numerical density of oligodendrocytes and oligodendrocyte clusters in the anterior putamen in major psychiatric disorders. Eur Arch Psychiatry Clin Neurosci 2020; 270:841-850. [PMID: 32060609 DOI: 10.1007/s00406-020-01108-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 02/03/2020] [Indexed: 02/07/2023]
Abstract
There is increasing evidence to support the notion that oligodendrocyte and myelin abnormalities may contribute to the functional dysconnectivity found in the major psychiatric disorders. The putamen, which is an important hub in the cortico-striato-thalamo-cortical loop, has been implicated in a broad spectrum of psychiatric illnesses and is a central target of their treatments. Previously we reported a reduction in the numerical density of oligodendrocytes and oligodendrocyte clusters in the prefrontal and parietal cortex in schizophrenia. Oligodendrocyte clusters contain oligodendrocyte progenitors and are involved in functionally dependent myelination. We measured the numerical density (Nv) of oligodendrocytes and oligodendrocyte clusters in the putamen in schizophrenia, bipolar disorder (BPD) and major depressive disorder (MDD) as compared to healthy controls (15 cases per group). Optical disector was used to estimate the Nv of oligodendrocytes and oligodendrocyte clusters. A significant reduction in both the Nv of oligodendrocytes (- 34%; p < 0.01) and the Nv of oligodendrocyte clusters (- 41%; p < 0.05) was found in the schizophrenia group as compared to the control group. Sexual dimorphism for both measurements was found only within the control group. The Nv of oligodendrocytes was significantly lower in male schizophrenia cases as compared to the male control cases. However, the Nv of oligodendrocyte clusters was significantly lower in all male clinical cases as compared to the male control group. The data suggest that lowered density of oligodendrocytes and oligodendrocyte clusters may contribute to the altered functional connectivity in the putamen in subjects with schizophrenia.
Collapse
|
26
|
Świerczek A, Jankowska A, Chłoń-Rzepa G, Pawłowski M, Wyska E. Advances in the Discovery of PDE10A Inhibitors for CNS-Related Disorders. Part 2: Focus on Schizophrenia. Curr Drug Targets 2020; 20:1652-1669. [PMID: 31368871 DOI: 10.2174/1389450120666190801114210] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 07/15/2019] [Accepted: 07/19/2019] [Indexed: 12/31/2022]
Abstract
Schizophrenia is a debilitating mental disorder with relatively high prevalence (~1%), during which positive manifestations (such as psychotic states) and negative symptoms (e.g., a withdrawal from social life) occur. Moreover, some researchers consider cognitive impairment as a distinct domain of schizophrenia symptoms. The imbalance in dopamine activity, namely an excessive release of this neurotransmitter in the striatum and insufficient amounts in the prefrontal cortex is believed to be partially responsible for the occurrence of these groups of manifestations. Second-generation antipsychotics are currently the standard treatment of schizophrenia. Nevertheless, the existent treatment is sometimes ineffective and burdened with severe adverse effects, such as extrapyramidal symptoms. Thus, there is an urgent need to search for alternative treatment options of this disease. This review summarizes the results of recent preclinical and clinical studies on phosphodiesterase 10A (PDE10A), which is highly expressed in the mammalian striatum, as a potential drug target for the treatment of schizophrenia. Based on the literature data, not only selective PDE10A inhibitors but also dual PDE2A/10A, and PDE4B/10A inhibitors, as well as multifunctional ligands with a PDE10A inhibitory potency are compounds that may combine antipsychotic, precognitive, and antidepressant functions. Thus, designing such compounds may constitute a new direction of research for new potential medications for schizophrenia. Despite failures of previous clinical trials of selective PDE10A inhibitors for the treatment of schizophrenia, new compounds with this mechanism of action are currently investigated clinically, thus, the search for new inhibitors of PDE10A, both selective and multitarget, is still warranted.
Collapse
Affiliation(s)
- Artur Świerczek
- Department of Pharmacokinetics and Physical Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland
| | - Agnieszka Jankowska
- Department of Medicinal Chemistry, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland
| | - Grażyna Chłoń-Rzepa
- Department of Medicinal Chemistry, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland
| | - Maciej Pawłowski
- Department of Medicinal Chemistry, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland
| | - Elżbieta Wyska
- Department of Pharmacokinetics and Physical Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland
| |
Collapse
|
27
|
Selemon LD, Begovic A. Reduced Midbrain Dopamine Neuron Number in the Adult Non-human Primate Brain after Fetal Radiation Exposure. Neuroscience 2020; 442:193-201. [PMID: 32659340 DOI: 10.1016/j.neuroscience.2020.07.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 06/17/2020] [Accepted: 07/02/2020] [Indexed: 11/19/2022]
Abstract
Early gestation is a neurodevelopmental period that is especially vulnerable to environmental insult and one in which neurogenesis features prominently. Prenatal perturbation during early gestation has been linked to neuropsychiatric illnesses such as autism and schizophrenia, and severe environmental insult during this period can result in profound mental impairment. Midbrain dopamine neurons are generated during early gestation and play a key role in the motor, cognitive and reward circuitries implicated in neuropsychiatric disease and addiction. This study examined the impact of curtailing neurogenesis in early gestation on neuron number in the midbrain dopamine group, i.e., the substantia nigra and contiguous ventral tegmental area. Rhesus macaque monkeys were exposed in utero on embryonic days 39-41 to x-irradiation (3-4 exposures of 50 cGy over 3-7 days totalling <200 cGy) and allowed to mature to full adulthood. Stereologic cell counts of tyrosine hydroxylase-positive neurons in the midbrain dopamine group were performed in adult monkeys, as were measurements of somal size. Mean total neuron number in the irradiated monkeys was significantly reduced on average by 33% compared to that of the control group. Somal size did not differ between the groups, suggesting that the integrity of survivor populations was not impacted. Reduced midbrain dopamine neuron number in fetally irradiated, adult monkeys indicates that radiation exposure during the critical period of neurogenesis results in an enduring reduction of this population and underscores the susceptibility of early neurodevelopmental processes to irreversible damage from environmental exposures.
Collapse
Affiliation(s)
- Lynn D Selemon
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, United States.
| | - Anita Begovic
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
28
|
Mabry SJ, McCollum LA, Farmer CB, Bloom ES, Roberts RC. Evidence for altered excitatory and inhibitory tone in the post-mortem substantia nigra in schizophrenia. World J Biol Psychiatry 2020; 21:339-356. [PMID: 31062628 PMCID: PMC6891153 DOI: 10.1080/15622975.2019.1615638] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 03/21/2019] [Accepted: 05/02/2019] [Indexed: 12/14/2022]
Abstract
Objectives: The substantia nigra (SN) receives glutamatergic and GABAergic inputs that regulate dopaminergic neuronal activity. Imaging studies have shown hyperactivity of the SN in schizophrenia (SZ) patients. We examined neurochemically defined inputs to the SN, synaptic density, and neuromelanin content that might contribute to or reflect this hyperexcitability.Methods: Glutamatergic axon terminals were identified by the immunohistochemical localisation of vGLUT1 and vGLUT2; GABA inputs were identified by the immunohistochemical localisation of GAD67. Neuromelanin granules are visible in unstained sections and thus were assessed in unstained sections. Optical densitometry was measured to assess the density of vGLUT1, vGLUT2 or GAD67 immunolabelled axon terminals and neuromelanin granules. Electron microscopy was used to quantify synaptic and mitochondrial density.Results: Compared to controls, SZ subjects had nonsignificant trends toward a decrease in vGLUT1, and an increase in both vGLUT2 and GAD67. vGLUT1 was negatively correlated with GAD67 in normal controls (NCs) and positively correlated in SZ subjects. A correlation of coefficient analysis showed a significant difference between the negative correlation in NCs and the positive correlation in SZ subjects. Frequency histograms showed the distribution of neuromelanin density was different in SZ subjects compared to NCs. Synaptic density data showed a decrease in inhibitory synapses in SZ subjects. Mitochondrial density was normal in SZ subjects.Conclusions: Synaptic density alterations and the lack of a positive correlation between GAD67 and vGLUT1 could contribute to hyperactivity in the SN.
Collapse
Affiliation(s)
- Samuel J. Mabry
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, 1720 7 Ave. South, Birmingham AL, 35294
| | - Lesley A. McCollum
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, 1720 7 Ave. South, Birmingham AL, 35294
| | - Charlene B. Farmer
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, 1720 7 Ave. South, Birmingham AL, 35294
| | - Emma S. Bloom
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, 1720 7 Ave. South, Birmingham AL, 35294
| | - Rosalinda C. Roberts
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, 1720 7 Ave. South, Birmingham AL, 35294
| |
Collapse
|
29
|
Reis-de-Oliveira G, Zuccoli GS, Fioramonte M, Schimitt A, Falkai P, Almeida V, Martins-de-Souza D. Digging deeper in the proteome of different regions from schizophrenia brains. J Proteomics 2020; 223:103814. [PMID: 32389842 DOI: 10.1016/j.jprot.2020.103814] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 04/21/2020] [Accepted: 05/05/2020] [Indexed: 02/07/2023]
Abstract
Schizophrenia is a psychiatric disorder that affects 21 million people worldwide. Despite several studies having been shown that some brain regions may play a critical role in the pathophysiology of schizophrenia, the molecular basis to explain this diversity is still lacking. The cerebellum (CER), caudate nucleus (CAU), and posterior cingulate cortex (PCC) are areas associated with negative and cognitive symptoms in schizophrenia. In this study, we performed shotgun proteomics of the aforementioned brain regions, collected postmortem from patients with schizophrenia and compared with the mentally healthy group. In addition, we performed a proteomic analysis of nuclear and mitochondrial fractions of these same regions. Our results presented 106, 727 and 135 differentially regulated proteins in the CAU, PCC, and CER, respectively. Pathway enrichment analysis revealed dysfunctions associated with synaptic processes in the CAU, transport in the CER, and in energy metabolism in the PCC. In all brain areas, we found that proteins related to oligodendrocytes and the metabolic processes were dysregulated in schizophrenia. SIGNIFICANCE: Schizophrenia is a complex and heterogeneous psychiatric disorder. Despite much research having been done to increase the knowledge about the role of each region in the pathophysiology of this disorder, the molecular mechanisms underlying it are still lacking. We performed shotgun proteomics in the postmortem cerebellum (CER), caudate nucleus (CAU) and posterior cingulate cortex (PCC) from patients with schizophrenia and compared with healthy controls. Our findings suggest that each aforementioned region presents dysregulations in specific molecular pathways, such as energy metabolism in the PCC, transport in the CER, and synaptic process in the CAU. Additionally, these areas presented dysfunctions in oligodendrocytes and metabolic processes. Our results may highlight future directions for the development of novel clinical approaches for specific therapeutic targets.
Collapse
Affiliation(s)
- G Reis-de-Oliveira
- Lab of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - G S Zuccoli
- Lab of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - M Fioramonte
- Lab of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - A Schimitt
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University (LMU), Munich, Germany; Laboratory of Neurosciences (LIM-27), Institute of Psychiatry, University of São Paulo, São Paulo, Brazil
| | - P Falkai
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University (LMU), Munich, Germany
| | - V Almeida
- Lab of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - D Martins-de-Souza
- Lab of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil; Experimental Medicine Research Cluster (EMRC), University of Campinas, Campinas, Brazil; D'Or Institute for Research and Education (IDOR), São Paulo, Brazil; Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBION), Conselho Nacional de Desenvolvimento Científico e Tecnológico, São Paulo, Brazil.
| |
Collapse
|
30
|
Duong P, Tenkorang MAA, Trieu J, McCuiston C, Rybalchenko N, Cunningham RL. Neuroprotective and neurotoxic outcomes of androgens and estrogens in an oxidative stress environment. Biol Sex Differ 2020; 11:12. [PMID: 32223745 PMCID: PMC7104511 DOI: 10.1186/s13293-020-0283-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 01/20/2020] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND The role of sex hormones on cellular function is unclear. Studies show androgens and estrogens are protective in the CNS, whereas other studies found no effects or damaging effects. Furthermore, sex differences have been observed in multiple oxidative stress-associated CNS disorders, such as Alzheimer's disease, depression, and Parkinson's disease. The goal of this study is to examine the relationship between sex hormones (i.e., androgens and estrogens) and oxidative stress on cell viability. METHODS N27 and PC12 neuronal and C6 glial phenotypic cell lines were used. N27 cells are female rat derived, whereas PC12 cells and C6 cells are male rat derived. These cells express estrogen receptors and the membrane-associated androgen receptor variant, AR45, but not the full-length androgen receptor. N27, PC12, and C6 cells were exposed to sex hormones either before or after an oxidative stressor to examine neuroprotective and neurotoxic properties, respectively. Estrogen receptor and androgen receptor inhibitors were used to determine the mechanisms mediating hormone-oxidative stress interactions on cell viability. Since the presence of AR45 in the human brain tissue was unknown, we examined the postmortem brain tissue from men and women for AR45 protein expression. RESULTS Neither androgens nor estrogens were protective against subsequent oxidative stress insults in glial cells. However, these hormones exhibited neuroprotective properties in neuronal N27 and PC12 cells via the estrogen receptor. Interestingly, a window of opportunity exists for sex hormone neuroprotection, wherein temporary hormone deprivation blocked neuroprotection by sex hormones. However, if sex hormones are applied following an oxidative stressor, they exacerbated oxidative stress-induced cell loss in neuronal and glial cells. CONCLUSIONS Sex hormone action on cell viability is dependent on the cellular environment. In healthy neuronal cells, sex hormones are protective against oxidative stress insults via the estrogen receptor, regardless of sex chromosome complement (XX, XY). However, in unhealthy (e.g., high oxidative stress) cells, sex hormones exacerbated oxidative stress-induced cell loss, regardless of cell type or sex chromosome complement. The non-genomic AR45 receptor, which is present in humans, mediated androgen's damaging effects, but it is unknown which receptor mediated estrogen's damaging effects. These differential effects of sex hormones that are dependent on the cellular environment, receptor profile, and cell type may mediate the observed sex differences in oxidative stress-associated CNS disorders.
Collapse
Affiliation(s)
- Phong Duong
- Department of Physiology and Anatomy, Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Mavis A A Tenkorang
- Department of Physiology and Anatomy, Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Jenny Trieu
- Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Clayton McCuiston
- Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Nataliya Rybalchenko
- Department of Physiology and Anatomy, Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Rebecca L Cunningham
- Department of Physiology and Anatomy, Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA. .,Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, 3400 Camp Bowie Boulevard, Fort Worth, TX, 76107, USA.
| |
Collapse
|
31
|
Decreased density of cholinergic interneurons in striatal territories in Williams syndrome. Brain Struct Funct 2020; 225:1019-1032. [PMID: 32189114 DOI: 10.1007/s00429-020-02055-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 02/27/2020] [Indexed: 12/22/2022]
Abstract
Williams syndrome (WS) is a rare neurodevelopmental disorder caused by the hemideletion of approximately 25-28 genes at 7q11.23. Its unusual social and cognitive phenotype is most strikingly characterized by the disinhibition of social behavior, in addition to reduced global IQ, with a relative sparing of language ability. Hypersociality and increased social approach behavior in WS may represent a unique inability to inhibit responses to specific social stimuli, which is likely associated with abnormalities of frontostriatal circuitry. The striatum is characterized by a diversity of interneuron subtypes, including inhibitory parvalbumin-positive interneurons (PV+) and excitatory cholinergic interneurons (Ch+). Animal model research has identified an important role for these specialized cells in regulating social approach behavior. Previous research in humans identified a depletion of interneuron subtypes associated with neuropsychiatric disorders. Here, we examined the density of PV+ and Ch+ interneurons in the striatum of 13 WS and neurotypical (NT) subjects. We found a significant reduction in the density of Ch+ interneurons in the medial caudate nucleus and nucleus accumbens, important regions receiving cortical afferents from the orbitofrontal and ventromedial prefrontal cortex, and circuitry involved in language and reward systems. No significant difference in the distribution of PV+ interneurons was found. The pattern of decreased Ch+ interneuron densities in WS differs from patterns of interneuron depletion found in other disorders.
Collapse
|
32
|
Tarcijonas G, Foran W, Haas GL, Luna B, Sarpal DK. Intrinsic Connectivity of the Globus Pallidus: An Uncharted Marker of Functional Prognosis in People With First-Episode Schizophrenia. Schizophr Bull 2020; 46:184-192. [PMID: 31150557 PMCID: PMC6942165 DOI: 10.1093/schbul/sbz034] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
There is growing evidence suggesting that abnormalities in cortical-basal ganglia circuitry may play a significant role in determining outcomes in schizophrenia. The globus pallidus (GP), a critical structure within this circuitry, unique in its role as a mediator of competing inputs through the striatum, has not been well characterized in schizophrenia. The following study examined functional interactions of the GP in individuals with first-episode schizophrenia (FES). To probe the large-scale intrinsic connectivity of the GP, resting-state fMRI scans were obtained from patients with FES and sex and age-matched healthy controls. Participants with FES were also evaluated after 6 months via the Strauss-Carpenter Outcomes Scale to assess overall functional trajectory. The GP was parcellated to generate seeds within its substructures, and connectivity maps were generated. Our FES cohort showed significantly lower functional connectivity between the left GP interna and a network of regions including the dorsolateral prefrontal cortex, caudate, and cerebellum at baseline. In addition, FES participants with lower overall scores of functioning at 6 months showed significantly decreased connectivity between the GP interna and the dorsal anterior cingulate and bilateral insula, all regions important for motivational salience. These results provide novel evidence for unique abnormalities in functional interactions of the GP with key prefrontal cortical regions in FES. Our findings also suggest that reduced prefrontal-pallidal connectivity may serve as a predictor of early functional outcome.
Collapse
Affiliation(s)
- Goda Tarcijonas
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
| | - William Foran
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
| | - Gretchen L Haas
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
| | - Beatriz Luna
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA,Department of Psychology, University of Pittsburgh, Pittsburgh, PA,Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA
| | - Deepak K Sarpal
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA,To whom correspondence should be addressed; Department of Psychiatry, University of Pittsburgh, 3501 Forbes Avenue, Suite 530, Pittsburgh, PA 15213, US; tel: 412-246-5618, fax: 412-246-5007, e-mail:
| |
Collapse
|
33
|
Jankowska A, Satała G, Partyka A, Wesołowska A, Bojarski AJ, Pawłowski M, Chłoń-Rzepa G. Discovery and Development of Non-Dopaminergic Agents for the Treatment of Schizophrenia: Overview of the Preclinical and Early Clinical Studies. Curr Med Chem 2019; 26:4885-4913. [PMID: 31291870 DOI: 10.2174/0929867326666190710172002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/11/2019] [Accepted: 06/14/2019] [Indexed: 02/05/2023]
Abstract
Schizophrenia is a chronic psychiatric disorder that affects about 1 in 100 people around the world and results in persistent emotional and cognitive impairments. Untreated schizophrenia leads to deterioration in quality of life and premature death. Although the clinical efficacy of dopamine D2 receptor antagonists against positive symptoms of schizophrenia supports the dopamine hypothesis of the disease, the resistance of negative and cognitive symptoms to these drugs implicates other systems in its pathophysiology. Many studies suggest that abnormalities in glutamate homeostasis may contribute to all three groups of schizophrenia symptoms. Scientific considerations also include disorders of gamma-aminobutyric acid-ergic and serotonergic neurotransmissions as well as the role of the immune system. The purpose of this review is to update the most recent reports on the discovery and development of non-dopaminergic agents that may reduce positive, negative, and cognitive symptoms of schizophrenia, and may be alternative to currently used antipsychotics. This review collects the chemical structures of representative compounds targeting metabotropic glutamate receptor, gamma-aminobutyric acid type A receptor, alpha 7 nicotinic acetylcholine receptor, glycine transporter type 1 and glycogen synthase kinase 3 as well as results of in vitro and in vivo studies indicating their efficacy in schizophrenia. Results of clinical trials assessing the safety and efficacy of the tested compounds have also been presented. Finally, attention has been paid to multifunctional ligands with serotonin receptor affinity or phosphodiesterase inhibitory activity as novel strategies in the search for dedicated medicines for patients with schizophrenia.
Collapse
Affiliation(s)
- Agnieszka Jankowska
- Department of Medicinal Chemistry, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland
| | - Grzegorz Satała
- Department of Medicinal Chemistry, Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Street, 31-343 Krakow, Poland
| | - Anna Partyka
- Department of Clinical Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland
| | - Anna Wesołowska
- Department of Clinical Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland
| | - Andrzej J Bojarski
- Department of Medicinal Chemistry, Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Street, 31-343 Krakow, Poland
| | - Maciej Pawłowski
- Department of Medicinal Chemistry, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland
| | - Grażyna Chłoń-Rzepa
- Department of Medicinal Chemistry, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland
| |
Collapse
|
34
|
Shukla DK, Chiappelli JJ, Sampath H, Kochunov P, Hare SM, Wisner K, Rowland LM, Hong LE. Aberrant Frontostriatal Connectivity in Negative Symptoms of Schizophrenia. Schizophr Bull 2019; 45:1051-1059. [PMID: 30576563 PMCID: PMC6737477 DOI: 10.1093/schbul/sby165] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Negative symptoms represent a distinct component of psychopathology in schizophrenia (SCZ) and are a stable construct over time. Although impaired frontostriatal connectivity has been frequently described in SCZ, its link with negative symptoms has not been carefully studied. We tested the hypothesis that frontostriatal connectivity at rest may be associated with the severity of negative symptoms in SCZ. Resting state functional connectivity (rsFC) data from 95 mostly medicated patients with SCZ and 139 healthy controls (HCs) were acquired. Negative symptoms were assessed using the Brief Negative Symptom Scale. The study analyzed voxel-wise rsFC between 9 frontal "seed regions" and the entire striatum, with the intention to reduce potential biases introduced by predefining any single frontal or striatal region. SCZ showed significantly reduced rsFC between the striatum and the right medial and lateral orbitofrontal cortex (OFC), lateral prefrontal cortex, and rostral anterior cingulate cortex compared with HCs. Further, rsFC between the striatum and the right medial OFC was significantly associated with negative symptom severity. The involved striatal regions were primarily at the ventral putamen. Our results support reduced frontostriatal functional connectivity in SCZ and implicate striatal connectivity with the right medial OFC in negative symptoms. This task-independent resting functional magnetic resonance imaging study showed that medial OFC-striatum functional connectivity is reduced in SCZ and associated with severity of negative symptoms. This finding supports a significant association between frontostriatal connectivity and negative symptoms and thus may provide a potential circuitry-level biomarker to study the neurobiological mechanisms of negative symptoms.
Collapse
Affiliation(s)
- Dinesh K Shukla
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD,To whom correspondence should be addressed; Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, PO Box 21247, Baltimore, MD 21228, US; tel: 410-402-6028, fax: 410-402-6077, e-mail:
| | - Joshua John Chiappelli
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD
| | - Hemalatha Sampath
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD
| | - Peter Kochunov
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD
| | - Stephanie M Hare
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD
| | - Krista Wisner
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD
| | - Laura M Rowland
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD
| | - L Elliot Hong
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD
| |
Collapse
|
35
|
Effects of Neonatal Administration of Memantine on Hippocampal Asymmetry and Working Memory Impairment Induced by Early Maternal Deprivation in Rats. NEUROPHYSIOLOGY+ 2019. [DOI: 10.1007/s11062-019-09799-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
36
|
Ye J, Ji F, Jiang D, Lin X, Chen G, Zhang W, Shan P, Zhang L, Zhuo C. Polymorphisms in Dopaminergic Genes in Schizophrenia and Their Implications in Motor Deficits and Antipsychotic Treatment. Front Neurosci 2019; 13:355. [PMID: 31057354 PMCID: PMC6479209 DOI: 10.3389/fnins.2019.00355] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 03/28/2019] [Indexed: 12/14/2022] Open
Abstract
Dopaminergic system dysfunction is involved in schizophrenia (SCZ) pathogenesis and can mediate SCZ-related motor disorders. Recent studies have gradually revealed that SCZ susceptibility and the associated motor symptoms can be mediated by genetic factors, including dopaminergic genes. More importantly, polymorphisms in these genes are associated with both antipsychotic drug sensitivity and adverse effects. The study of genetic polymorphisms in the dopaminergic system may help to optimize individualized drug strategies for SCZ patients. This review summarizes the current progress about the involvement of the dopamine system in SCZ-associated motor disorders and the motor-related adverse effects after antipsychotic treatment, with a special focus on polymorphisms in dopaminergic genes. We hypothesize that the genetic profile of the dopaminergic system mediates both SCZ-associated motor deficits associated and antipsychotic drug-related adverse effects. The study of dopaminergic gene polymorphisms may help to predict drug efficacy and decrease adverse effects, thereby optimizing treatment strategies.
Collapse
Affiliation(s)
- Jiaen Ye
- Department of Psychiatry, Wenzhou Seventh People's Hospital, Wenzhou, China
| | - Feng Ji
- Department of Psychiatry, College of Mental Health, Jining Medical University, Jining, China
| | - Deguo Jiang
- Department of Psychiatric-Neuroimaging-Genetics and Morbidity Laboratory (PNGC-Lab), Nankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center, Mental Health Teaching Hospital, Tianjin Medical University, Tianjin, China
| | - Xiaodong Lin
- Department of Psychiatry, Wenzhou Seventh People's Hospital, Wenzhou, China
| | - Guangdong Chen
- Department of Psychiatry, Wenzhou Seventh People's Hospital, Wenzhou, China
| | - Wei Zhang
- Department of Psychiatry, Wenzhou Seventh People's Hospital, Wenzhou, China
| | - Peiwei Shan
- Department of Psychiatry, Wenzhou Seventh People's Hospital, Wenzhou, China
| | - Li Zhang
- GHM Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Chuanjun Zhuo
- Department of Psychiatry, Wenzhou Seventh People's Hospital, Wenzhou, China.,Department of Psychiatry, College of Mental Health, Jining Medical University, Jining, China.,Department of Psychiatric-Neuroimaging-Genetics and Morbidity Laboratory (PNGC-Lab), Nankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center, Mental Health Teaching Hospital, Tianjin Medical University, Tianjin, China
| |
Collapse
|
37
|
Moussa-Tooks AB, Kim DJ, Bartolomeo LA, Purcell JR, Bolbecker AR, Newman SD, O’Donnell BF, Hetrick WP. Impaired Effective Connectivity During a Cerebellar-Mediated Sensorimotor Synchronization Task in Schizophrenia. Schizophr Bull 2019; 45:531-541. [PMID: 29800417 PMCID: PMC6483568 DOI: 10.1093/schbul/sby064] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Prominent conceptual models characterize schizophrenia as a dysconnectivity syndrome, with recent research focusing on the contributions of the cerebellum in this framework. The present study examined the role of the cerebellum and its effective connectivity to the cerebrum during sensorimotor synchronization in schizophrenia. Specifically, the role of the cerebellum in temporally coordinating cerebral motor activity was examined through path analysis. Thirty-one individuals diagnosed with schizophrenia and 40 healthy controls completed a finger-tapping fMRI task including tone-paced synchronization and self-paced continuation tapping at a 500 ms intertap interval (ITI). Behavioral data revealed shorter and more variable ITIs during self-paced continuation, greater clock (vs motor) variance, and greater force of tapping in the schizophrenia group. In a whole-brain analysis, groups showed robust activation of the cerebellum during self-paced continuation but not during tone-paced synchronization. However, effective connectivity analysis revealed decreased connectivity in individuals with schizophrenia between the cerebellum and primary motor cortex but increased connectivity between cerebellum and thalamus during self-paced continuation compared with healthy controls. These findings in schizophrenia indicate diminished temporal coordination of cerebral motor activity by cerebellum during the continuation tapping portion of sensorimotor synchronization. Taken together with the behavioral finding of greater temporal variability in schizophrenia, these effective connectivity results are consistent with structural and temporal models of dysconnectivity in the disorder.
Collapse
Affiliation(s)
| | - Dae-Jin Kim
- Department of Psychological & Brain Sciences, Indiana University, Bloomington, IN
| | | | - John R Purcell
- Department of Psychological & Brain Sciences, Indiana University, Bloomington, IN
| | - Amanda R Bolbecker
- Department of Psychological & Brain Sciences, Indiana University, Bloomington, IN,Larue D. Carter Memorial Hospital, Indianapolis, IN,Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN
| | - Sharlene D Newman
- Department of Psychological & Brain Sciences, Indiana University, Bloomington, IN,Imaging Research Facility, Indiana University College of Arts and Sciences, Bloomington, IN
| | - Brian F O’Donnell
- Department of Psychological & Brain Sciences, Indiana University, Bloomington, IN,Larue D. Carter Memorial Hospital, Indianapolis, IN,Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN
| | - William P Hetrick
- Department of Psychological & Brain Sciences, Indiana University, Bloomington, IN,Larue D. Carter Memorial Hospital, Indianapolis, IN,Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN,To whom correspondence should be addressed; Department of Psychological & Brain Sciences, Indiana University, 1101 E. 10th Street, Bloomington, IN 47405; tel: 812-855-2620, fax: 812-855-4691, e-mail:
| |
Collapse
|
38
|
Ramos-Miguel A, Barakauskas V, Alamri J, Miyauchi M, Barr AM, Beasley CL, Rosoklija G, Mann JJ, Dwork AJ, Moradian A, Morin GB, Honer WG. The SNAP25 Interactome in Ventromedial Caudate in Schizophrenia Includes the Mitochondrial Protein ARF1. Neuroscience 2019; 420:97-111. [PMID: 30610939 DOI: 10.1016/j.neuroscience.2018.12.045] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 12/21/2018] [Accepted: 12/24/2018] [Indexed: 12/11/2022]
Abstract
Abnormalities of SNAP25 (synaptosome-associated protein 25) amount and protein-protein interactions occur in schizophrenia, and may contribute to abnormalities of neurotransmitter release in patients. However, presynaptic terminal function depends on multiple subcellular mechanisms, including energy provided by mitochondria. To explore the SNAP25 interactome in schizophrenia, we immunoprecipitated SNAP25 along with interacting proteins from the ventromedial caudate of 15 cases of schizophrenia and 13 controls. Proteins were identified with mass spectrometry-based proteomics. As well as 15 SNARE- (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) associated proteins, we identified 17 mitochondria-associated and four other proteins. The mitochondrial small GTPase ARF1 (ADP-ribosylation factor 1) was identified in eight schizophrenia SNAP25 immunoprecipitates and none from controls (P = 0.004). Although the ARF1-SNAP25 interaction may be increased, immunoblotting demonstrated 21% lower ARF1-21 (21 kiloDaltons) in schizophrenia samples (P = 0.04). In contrast, the mitochondrial protein UQCRC1 (ubiquinol-cytochrome c reductase core protein 1) did not differ. Lower ARF1-21 levels were associated with the previously reported increased SNAP25-syntaxin interaction in schizophrenia (r = -0.39, P = 0.04). Additional immunoprecipitation studies confirmed the ARF1-21-SNAP25 interaction, independent of UQCRC1. Both ARF1 and SNAP25 were localized to synaptosomes. Confocal microscopy demonstrated co-localization of ARF1 and SNAP25, and further suggested fivefold enrichment of ARF1 in synaptosomes containing an excitatory marker (vesicular glutamate transporter) compared with synaptosomes containing an inhibitory marker (vesicular GABA transporter). The present findings suggest an association between abnormalities of SNARE proteins involved with vesicular neurotransmission and the mitochondrial protein ARF1 that may contribute to the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Alfredo Ramos-Miguel
- BC Mental Health and Addictions Research Institute, 938 West 28th Ave, Vancouver, BC V5Z 4H4, Canada; Department of Psychiatry, University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC V6T 2A1, Canada; Department of Pharmacology, University of the Basque Country, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Barrio Sarriena, s/n, 48940 Leioa, Biscay, Spain
| | - Vilte Barakauskas
- BC Mental Health and Addictions Research Institute, 938 West 28th Ave, Vancouver, BC V5Z 4H4, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, 2J9-4500 Oak St., Vancouver, BC V6H 3B1, Canada
| | - Jehan Alamri
- BC Mental Health and Addictions Research Institute, 938 West 28th Ave, Vancouver, BC V5Z 4H4, Canada
| | - Masatoshi Miyauchi
- BC Mental Health and Addictions Research Institute, 938 West 28th Ave, Vancouver, BC V5Z 4H4, Canada; Department of Psychiatry, University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC V6T 2A1, Canada
| | - Alasdair M Barr
- BC Mental Health and Addictions Research Institute, 938 West 28th Ave, Vancouver, BC V5Z 4H4, Canada; Department of Anesthesiology, Pharmacology, & Therapeutics, University of British Columbia, 2176 Health Sciences Mall Vancouver, BC V6T 1Z3, Canada
| | - Clare L Beasley
- BC Mental Health and Addictions Research Institute, 938 West 28th Ave, Vancouver, BC V5Z 4H4, Canada; Department of Psychiatry, University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC V6T 2A1, Canada
| | - Gorazd Rosoklija
- Department of Psychiatry, Columbia University, 1051 Riverside Drive, New York, NY 10032, USA
| | - J John Mann
- Department of Psychiatry, Columbia University, 1051 Riverside Drive, New York, NY 10032, USA
| | - Andrew J Dwork
- Department of Psychiatry, Columbia University, 1051 Riverside Drive, New York, NY 10032, USA
| | - Annie Moradian
- Department of Medical Genetics, University of British Columbia, C234-4500 Oak St., Vancouver, BC V6H 3B1, Canada
| | - Gregg B Morin
- Department of Medical Genetics, University of British Columbia, C234-4500 Oak St., Vancouver, BC V6H 3B1, Canada
| | - William G Honer
- BC Mental Health and Addictions Research Institute, 938 West 28th Ave, Vancouver, BC V5Z 4H4, Canada; Department of Psychiatry, University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC V6T 2A1, Canada.
| |
Collapse
|
39
|
Klein MO, Battagello DS, Cardoso AR, Hauser DN, Bittencourt JC, Correa RG. Dopamine: Functions, Signaling, and Association with Neurological Diseases. Cell Mol Neurobiol 2019; 39:31-59. [PMID: 30446950 DOI: 10.1007/s10571-018-0632-3] [Citation(s) in RCA: 512] [Impact Index Per Article: 85.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 11/02/2018] [Indexed: 02/07/2023]
Abstract
The dopaminergic system plays important roles in neuromodulation, such as motor control, motivation, reward, cognitive function, maternal, and reproductive behaviors. Dopamine is a neurotransmitter, synthesized in both central nervous system and the periphery, that exerts its actions upon binding to G protein-coupled receptors. Dopamine receptors are widely expressed in the body and function in both the peripheral and the central nervous systems. Dopaminergic signaling pathways are crucial to the maintenance of physiological processes and an unbalanced activity may lead to dysfunctions that are related to neurodegenerative diseases. Unveiling the neurobiology and the molecular mechanisms that underlie these illnesses may contribute to the development of new therapies that could promote a better quality of life for patients worldwide. In this review, we summarize the aspects of dopamine as a catecholaminergic neurotransmitter and discuss dopamine signaling pathways elicited through dopamine receptor activation in normal brain function. Furthermore, we describe the potential involvement of these signaling pathways in evoking the onset and progression of some diseases in the nervous system, such as Parkinson's, Schizophrenia, Huntington's, Attention Deficit and Hyperactivity Disorder, and Addiction. A brief description of new dopaminergic drugs recently approved and under development treatments for these ailments is also provided.
Collapse
Affiliation(s)
- Marianne O Klein
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, 05508-000, Brazil
| | - Daniella S Battagello
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, 05508-000, Brazil
| | - Ariel R Cardoso
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, 05508-000, Brazil
| | - David N Hauser
- Center for Translational Neuroscience, Sanford Burnham Prebys (SBP) Medical Discovery Institute, 10901 North Torrey Pines Rd., La Jolla, CA, 92037, USA
| | - Jackson C Bittencourt
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, 05508-000, Brazil.
- Center for Neuroscience and Behavior, Institute of Psychology, USP, São Paulo, Brazil.
| | - Ricardo G Correa
- Center for Translational Neuroscience, Sanford Burnham Prebys (SBP) Medical Discovery Institute, 10901 North Torrey Pines Rd., La Jolla, CA, 92037, USA.
| |
Collapse
|
40
|
Bariselli S, Fobbs WC, Creed MC, Kravitz AV. A competitive model for striatal action selection. Brain Res 2018; 1713:70-79. [PMID: 30300636 DOI: 10.1016/j.brainres.2018.10.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 10/03/2018] [Accepted: 10/05/2018] [Indexed: 12/20/2022]
Abstract
The direct and indirect pathway striatal medium spiny neurons (dMSNs and iMSNs) have long been linked to action selection, but the precise roles of these neurons in this process remain unclear. Here, we review different models of striatal pathway function, focusing on the classic "go/no-go" model which posits that dMSNs facilitate movement while iMSNs inhibit movement, and the "complementary" model, which argues that dMSNs facilitate the selection of specific actions while iMSNs inhibit potentially conflicting actions. We discuss the merits and shortcomings of these models and propose a "competitive" model to explain the contribution of these two pathways to behavior. The "competitive" model argues that rather than inhibiting conflicting actions, iMSNs are tuned to the same actions that dMSNs facilitate, and the two populations "compete" to determine the animal's behavioral response. This model provides a theoretical explanation for how these pathways work together to select actions. In addition, it provides a link between action selection and behavioral reinforcement, via modulating synaptic strength at inputs onto dMSNs and iMSNs. Finally, this model makes predictions about how imbalances in the activity of these pathways may underlie behavioral traits associated with psychiatric disorders. Understanding the roles of these striatal pathways in action selection may help to clarify the neuronal mechanisms of decision-making under normal and pathological conditions.
Collapse
Affiliation(s)
- S Bariselli
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, United States
| | - W C Fobbs
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, United States
| | - M C Creed
- Washington University in St Louis, Department of Anesthesiology, St Louis, MO, United States
| | - A V Kravitz
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, United States; National Institute on Drug Abuse, Baltimore, MD, United States.
| |
Collapse
|
41
|
Ledonne A, Mercuri NB. mGluR1-Dependent Long Term Depression in Rodent Midbrain Dopamine Neurons Is Regulated by Neuregulin 1/ErbB Signaling. Front Mol Neurosci 2018; 11:346. [PMID: 30327588 PMCID: PMC6174199 DOI: 10.3389/fnmol.2018.00346] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 09/04/2018] [Indexed: 11/22/2022] Open
Abstract
Increasing evidence demonstrates that the neurotrophic factor Neuregulin 1 (NRG1) and its receptors, ErbB tyrosine kinases, modulate midbrain dopamine (DA) transmission. We have previously reported that NRG1/ErbB signaling is essential for proper metabotropic glutamate receptors 1 (mGluR1) functioning in midbrain DA neurons, thus the functional interaction between ErbB receptors and mGluR1 regulates neuronal excitation and in vivo striatal DA release. While it is widely recognized that mGluR1 play a pivotal role in long-term modifications of synaptic transmission in several brain areas, specific mGluR1-dependent forms of synaptic plasticity in substantia nigra pars compacta (SNpc) DA neurons have not been described yet. Here, first we aimed to detect and characterize mGluR1-dependent glutamatergic long-term depression (LTD) in SNpc DA neurons. Second, we tested the hypothesis that endogenous ErbB signaling, by affecting mGluR1, fine-tunes glutamatergic synaptic plasticity in DA cells. We found that either pharmacological or synaptic activation of mGluR1 causes an LTD of AMPAR-mediated transmission in SNpc DA neurons from mice and rat slices, which is reliant on endogenous NRG1/ErbB signaling. Indeed, LTD is counteracted by a broad spectrum ErbB inhibitor. Moreover, the intracellular injection of pan-ErbB- or ErbB2 inhibitors inside DA neurons reduces mGluR1-dependent LTD, suggesting an involvement of ErbB2/ErbB4-containing receptors. Interestingly, exogenous NRG1 fosters LTD expression during minimal mGluRI activation. These results enlarge our cognizance on mGluR1 relevance in the induction of a novel form of long-term synaptic plasticity in SNpc DA neurons and describe a new NRG1/ErbB-dependent mechanism shaping glutamatergic transmission in DA cells. This might have important implications either in DA-dependent behaviors and learning/memory processes or in DA-linked diseases.
Collapse
Affiliation(s)
- Ada Ledonne
- Department of Experimental Neuroscience, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Nicola Biagio Mercuri
- Department of Experimental Neuroscience, IRCCS Santa Lucia Foundation, Rome, Italy.,Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
42
|
Huang MH, Chiu YC, Tsai CF. Aripiprazole in a Patient of PLA2G6-Associated Neurodegeneration With Psychosis. Clin Neuropharmacol 2018; 41:136-137. [PMID: 29916839 DOI: 10.1097/wnf.0000000000000284] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PLA2G6-associated neurodegeneration is a major subtype of neurodegeneration with brain iron accumulation. Patients with PLA2G6-associated neurodegeneration usually present degenerative motor symptoms with neuropsychiatric disturbance. This report presents a patient with PLA2G6-related dystonia-parkinsonism, who had neuropsychiatric symptoms preceding movement symptoms. Antipsychotic drug was prescribed, and he developed parkinsonism years later. Differential diagnosis between drug-induced parkinsonism and PLA2G6-related dystonia-parkinsonism could be challenging. PLA2G6 sequencing has to be considered facing a patient with adulthood parkinsonism, especially when it is associated with initial psychiatric symptoms. Our case highlights a common problem treating psychiatric symptoms in neurodegenerative diseases that lead to parkinsonism. The use of antipsychotics requires a special caution in these patients because it may cause extrapyramidal symptoms. Aripiprazole had therapeutic effect on both mood and psychotic symptoms and less impact on motor features, which may be considered an ideal pharmacological approach in treating these patients.
Collapse
|
43
|
Middlebrooks EH, Tuna IS, Grewal SS, Almeida L, Heckman MG, Lesser ER, Foote KD, Okun MS, Holanda VM. Segmentation of the Globus Pallidus Internus Using Probabilistic Diffusion Tractography for Deep Brain Stimulation Targeting in Parkinson Disease. AJNR Am J Neuroradiol 2018; 39:1127-1134. [PMID: 29700048 DOI: 10.3174/ajnr.a5641] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 02/24/2018] [Indexed: 01/13/2023]
Abstract
BACKGROUND AND PURPOSE Although globus pallidus internus deep brain stimulation is a widely accepted treatment for Parkinson disease, there is persistent variability in outcomes that is not yet fully understood. In this pilot study, we aimed to investigate the potential role of globus pallidus internus segmentation using probabilistic tractography as a supplement to traditional targeting methods. MATERIALS AND METHODS Eleven patients undergoing globus pallidus internus deep brain stimulation were included in this retrospective analysis. Using multidirection diffusion-weighted MR imaging, we performed probabilistic tractography at all individual globus pallidus internus voxels. Each globus pallidus internus voxel was then assigned to the 1 ROI with the greatest number of propagated paths. On the basis of deep brain stimulation programming settings, the volume of tissue activated was generated for each patient using a finite element method solution. For each patient, the volume of tissue activated within each of the 10 segmented globus pallidus internus regions was calculated and examined for association with a change in the Unified Parkinson Disease Rating Scale, Part III score before and after treatment. RESULTS Increasing volume of tissue activated was most strongly correlated with a change in the Unified Parkinson Disease Rating Scale, Part III score for the primary motor region (Spearman r = 0.74, P = .010), followed by the supplementary motor area/premotor cortex (Spearman r = 0.47, P = .15). CONCLUSIONS In this pilot study, we assessed a novel method of segmentation of the globus pallidus internus based on probabilistic tractography as a supplement to traditional targeting methods. Our results suggest that our method may be an independent predictor of deep brain stimulation outcome, and evaluation of a larger cohort or prospective study is warranted to validate these findings.
Collapse
Affiliation(s)
| | - I S Tuna
- Departments of Radiology (I.S.T.)
| | | | | | - M G Heckman
- Division of Biomedical Statistics and Informatics (M.G.H., E.R.L.), Mayo Clinic, Jacksonville, Florida
| | - E R Lesser
- Division of Biomedical Statistics and Informatics (M.G.H., E.R.L.), Mayo Clinic, Jacksonville, Florida
| | - K D Foote
- Neurosurgery (K.D.F.), University of Florida, Gainesville, Florida
| | | | - V M Holanda
- Center of Neurology and Neurosurgery Associates (V.M.H.), BP-A Beneficência Portuguesa de São Paulo, São Paulo, Brazil
| |
Collapse
|
44
|
Suzuki K, Kimura H. TAK-063, a novel PDE10A inhibitor with balanced activation of direct and indirect pathways, provides a unique opportunity for the treatment of schizophrenia. CNS Neurosci Ther 2018; 24:604-614. [PMID: 29318783 DOI: 10.1111/cns.12798] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 12/12/2017] [Accepted: 12/15/2017] [Indexed: 01/04/2023] Open
Abstract
The basal ganglia regulates motor, cognitive, and emotional behaviors. Dysfunction of dopamine system in this area is implicated in the pathophysiology of schizophrenia characterized by positive symptoms, negative symptoms, and cognitive deficits. Medium spiny neurons (MSNs) are principal output neurons of striatum in the basal ganglia. Similar to current antipsychotics with dopamine D2 receptor antagonism or partial agonism, phosphodiesterase 10A (PDE10A) inhibitors activate indirect pathway MSNs, leading to the expectation of therapeutic potential for the treatment of psychosis. PDE10A inhibitors also activate direct pathway MSNs which may be associated with cognitive functions. These pathways have competing effects on antipsychotic-like activities and extrapyramidal symptoms in rodents. Therefore, careful consideration of activation pattern of these pathways by a PDE10A inhibitor is critical to produce potent efficacy and superior safety profiles. In this review, we outline the pharmacological profile of TAK-063, a novel PDE10A selective inhibitor. Our study revealed that off-rates of PDE10A inhibitors may characterize their pharmacological profiles via regulation of each MSN pathway. TAK-063, with a faster off-rate property, could provide a unique opportunity as a novel therapeutic approach to treatment of psychosis and cognitive deficits in schizophrenia. TAK-063 also has a therapeutic potential in other basal ganglia disorders.
Collapse
Affiliation(s)
- Kazunori Suzuki
- CNS Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Haruhide Kimura
- CNS Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| |
Collapse
|
45
|
Zhu J, Zhuo C, Xu L, Liu F, Qin W, Yu C. Altered Coupling Between Resting-State Cerebral Blood Flow and Functional Connectivity in Schizophrenia. Schizophr Bull 2017; 43:1363-1374. [PMID: 28521048 PMCID: PMC5737873 DOI: 10.1093/schbul/sbx051] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Respective changes in resting-state cerebral blood flow (CBF) and functional connectivity in schizophrenia have been reported. However, their coupling alterations in schizophrenia remain largely unknown. METHODS 89 schizophrenia patients and 90 sex- and age-matched healthy controls underwent resting-state functional MRI to calculate functional connectivity strength (FCS) and arterial spin labeling imaging to compute CBF. The CBF-FCS coupling of the whole gray matter and the CBF/FCS ratio (the amount of blood supply per unit of connectivity strength) of each voxel were compared between the 2 groups. RESULTS Whole gray matter CBF-FCS coupling was decreased in schizophrenia patients relative to healthy controls. In schizophrenia patients, the decreased CBF/FCS ratio was predominantly located in cognitive- and emotional-related brain regions, including the dorsolateral prefrontal cortex, insula, hippocampus and thalamus, whereas an increased CBF/FCS ratio was mainly identified in the sensorimotor regions, including the putamen, and sensorimotor, mid-cingulate and visual cortices. CONCLUSION These findings suggest that the neurovascular decoupling in the brain may be a possible neuropathological mechanism of schizophrenia.
Collapse
Affiliation(s)
- Jiajia Zhu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Chuanjun Zhuo
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China,Department of Psychiatry Functional Neuroimaging Laboratory, Tianjin Mental Health Center, Tianjin Anding Hospital, Tianjin, China,Tianjin Anning Hospital, Tianjin, China
| | - Lixue Xu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Feng Liu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Wen Qin
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Chunshui Yu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China,To whom correspondence should be addressed; Department of Radiology, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300052, China; tel: +86-22-63062026, fax: +86-22-63062290, e-mail:
| |
Collapse
|
46
|
Abstract
PURPOSE A positron emission tomography (PET) tracer for the enzyme phosphodiesterase 10A (PDE10A) is desirable to guide the discovery and development of PDE10A inhibitors as potential therapeutics. The preclinical characterization of the PDE10A PET tracer [(11)C]MK-8193 is described. PROCEDURES In vitro binding studies with [(3)H]MK-8193 were conducted in rat, monkey, and human brain tissue. PET studies with [(11)C]MK-8193 were conducted in rats and rhesus monkeys at baseline and following administration of a PDE10A inhibitor. RESULTS [(3)H]MK-8193 is a high-affinity, selective PDE10A radioligand in rat, monkey, and human brain tissue. In vivo, [(11)C]MK-8193 displays rapid kinetics, low test-retest variability, and a large specific signal that is displaced by a structurally diverse PDE10A inhibitor, enabling the determination of pharmacokinetic/enzyme occupancy relationships. CONCLUSIONS [(11)C]MK-8193 is a useful PET tracer for the preclinical characterization of PDE10A therapeutic candidates in rat and monkey. Further evaluation of [(11)C]MK-8193 in humans is warranted.
Collapse
|
47
|
Pavăl D, Rad F, Rusu R, Niculae AŞ, Colosi HA, Dobrescu I, Dronca E. Low Retinal Dehydrogenase 1 (RALDH1) Level in Prepubertal Boys with Autism Spectrum Disorder: A Possible Link to Dopamine Dysfunction? CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2017; 15:229-236. [PMID: 28783931 PMCID: PMC5565080 DOI: 10.9758/cpn.2017.15.3.229] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 08/23/2016] [Accepted: 09/05/2016] [Indexed: 11/18/2022]
Abstract
Objective Retinal dehydrogenase 1 (RALDH1) is a cytosolic enzyme which acts both as a source of retinoic acid (RA) and as a detoxification enzyme. RALDH1 has key functions in the midbrain dopaminergic system, which influences motivation, cognition, and social behavior. Since dopamine has been increasingly linked to autism spectrum disorder (ASD), we asked whether RALDH1 could contribute to the autistic phenotype. Therefore, we investigated for the first time the levels of RALDH1 in autistic patients. To further assess the detoxification function of RALDH1, we also explored 4-hydroxynonenal protein adducts (4-HNE PAs) and reduced glutathione (GSH) levels. Moreover, considering the effect of testosterone on RALDH1 expression, we measured the second to fourth digit ratio (2D:4D ratio) for both hands, which reflects exposure to prenatal testosterone. Methods Male patients with ASD (n=18; age, 62.9±4.3 months) and healthy controls (n=13; age, 78.1±4.9 months) were examined. Erythrocyte RALDH1, serum 4-HNE PAs and erythrocyte GSH levels were measured using colorimetric assays, and digit lengths were measured using digital calipers. Results We found significantly lower (−42.9%) RALDH1 levels in autistic patients as compared to controls (p=0.032). However, there was no difference in 4-HNE PAs levels (p=0.368), GSH levels (p=0.586), or 2D:4D ratios (p=0.246 in the left hand, p=0.584 in the right hand) between healthy controls and autistic subjects. Conclusion We concluded that a subset of autistic patients had a low RALDH1 level. These results suggest that low RALDH1 levels could contribute to the autistic phenotype by reflecting a dopaminergic dysfunction.
Collapse
Affiliation(s)
- Denis Pavăl
- Department of Molecular Sciences, Faculty of Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Florina Rad
- Alexandru Obregia Psychiatry Hospital, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Răzvan Rusu
- Department of Molecular Sciences, Faculty of Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Alexandru-Ştefan Niculae
- Department of Molecular Sciences, Faculty of Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Horaţiu Alexandru Colosi
- Department of Medical Informatics and Biostatistics, Faculty of Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Iuliana Dobrescu
- Alexandru Obregia Psychiatry Hospital, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Eleonora Dronca
- Department of Molecular Sciences, Faculty of Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
48
|
Abboud R, Noronha C, Diwadkar VA. Motor system dysfunction in the schizophrenia diathesis: Neural systems to neurotransmitters. Eur Psychiatry 2017. [PMID: 28641214 DOI: 10.1016/j.eurpsy.2017.04.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Motor control is a ubiquitous aspect of human function, and from its earliest origins, abnormal motor control has been proposed as being central to schizophrenia. The neurobiological architecture of the motor system is well understood in primates and involves cortical and sub-cortical components including the primary motor cortex, supplementary motor area, dorsal anterior cingulate cortex, the prefrontal cortex, the basal ganglia, and cerebellum. Notably all of these regions are associated in some manner to the pathophysiology of schizophrenia. At the molecular scale, both dopamine and γ-Aminobutyric Acid (GABA) abnormalities have been associated with working memory dysfunction, but particularly relating to the basal ganglia and the prefrontal cortex respectively. As evidence from multiple scales (behavioral, regional and molecular) converges, here we provide a synthesis of the bio-behavioral relevance of motor dysfunction in schizophrenia, and its consistency across scales. We believe that the selective compendium we provide can supplement calls arguing for renewed interest in studying the motor system in schizophrenia. We believe that in addition to being a highly relevant target for the study of schizophrenia related pathways in the brain, such focus provides tractable behavioral probes for in vivo imaging studies in the illness. Our assessment is that the motor system is a highly valuable research domain for the study of schizophrenia.
Collapse
Affiliation(s)
- R Abboud
- College of Osteopathic Medicine, Michigan State University Lansing, MI, USA
| | - C Noronha
- School of Medicine, University of Michigan, Ann Arbor, MI, USA
| | - V A Diwadkar
- Department of Psychiatry and Behavioral Neuroscience, Wayne State University School of Medicine, Suite 5A, Tolan Park Medical Building, 3901 Chrysler Service Drive, 48201 Detroit, MI, USA.
| |
Collapse
|
49
|
Zhuo C, Zhu J, Qin W, Qu H, Ma X, Yu C. Cerebral blood flow alterations specific to auditory verbal hallucinations in schizophrenia. Br J Psychiatry 2017; 210:209-215. [PMID: 28104737 DOI: 10.1192/bjp.bp.115.174961] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 09/25/2016] [Accepted: 10/07/2016] [Indexed: 01/17/2023]
Abstract
BackgroundAuditory verbal hallucinations (AVHs) have been associated with deficits in auditory and speech-related networks. However, the resting-state cerebral blood flow (CBF) alterations specific to AVHs in schizophrenia remain unknown.AimsTo explore AVH-related CBF alterations in individuals with schizophrenia.MethodIn total, 35 individuals with schizophrenia with AVHs, 41 individuals with schizophrenia without AVHs and 50 controls underwent arterial spin labelling magnetic resonance imaging. The CBF differences were voxel-wise compared across the three groups.ResultsWe found AVH-specific CBF increase in the right superior temporal gyrus and caudate, and AVH-specific CBF decrease in the bilateral occipital and left parietal cortices. We also observed consistent CBF changes in both schizophrenia subgroups (i.e. those with and without AVHs) including decreased CBF in the bilateral occipital regions, the left lateral prefrontal and insular cortices, and the right anterior cingulate cortex and increased CBF in the bilateral lateral temporal regions and putamen, the left middle cingulate cortex and the right thalamus.ConclusionsThe AVH-specific CBF increases in the auditory and striatal areas and CBF reductions in the visual and parietal areas suggest that there exists a CBF redistribution associated with AVHs.
Collapse
Affiliation(s)
- Chuanjun Zhuo
- Chuanjun Zhuo, MD, PhD, Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, and Department of Psychiatry Functional Neuroimaging Laboratory, Tianjin Mental Health Center, Tianjin Anding Hospital, Tianjin; Jiajia Zhu, MD, Wen Qin, MD, Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, China; Hongru Qu, MD, Xiaolei Ma, MD, Department of Psychiatry, Tianjin Anning Hospital, Tianjin, China; Chunshui Yu, MD, Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Jiajia Zhu
- Chuanjun Zhuo, MD, PhD, Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, and Department of Psychiatry Functional Neuroimaging Laboratory, Tianjin Mental Health Center, Tianjin Anding Hospital, Tianjin; Jiajia Zhu, MD, Wen Qin, MD, Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, China; Hongru Qu, MD, Xiaolei Ma, MD, Department of Psychiatry, Tianjin Anning Hospital, Tianjin, China; Chunshui Yu, MD, Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Wen Qin
- Chuanjun Zhuo, MD, PhD, Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, and Department of Psychiatry Functional Neuroimaging Laboratory, Tianjin Mental Health Center, Tianjin Anding Hospital, Tianjin; Jiajia Zhu, MD, Wen Qin, MD, Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, China; Hongru Qu, MD, Xiaolei Ma, MD, Department of Psychiatry, Tianjin Anning Hospital, Tianjin, China; Chunshui Yu, MD, Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Hongru Qu
- Chuanjun Zhuo, MD, PhD, Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, and Department of Psychiatry Functional Neuroimaging Laboratory, Tianjin Mental Health Center, Tianjin Anding Hospital, Tianjin; Jiajia Zhu, MD, Wen Qin, MD, Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, China; Hongru Qu, MD, Xiaolei Ma, MD, Department of Psychiatry, Tianjin Anning Hospital, Tianjin, China; Chunshui Yu, MD, Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaolei Ma
- Chuanjun Zhuo, MD, PhD, Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, and Department of Psychiatry Functional Neuroimaging Laboratory, Tianjin Mental Health Center, Tianjin Anding Hospital, Tianjin; Jiajia Zhu, MD, Wen Qin, MD, Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, China; Hongru Qu, MD, Xiaolei Ma, MD, Department of Psychiatry, Tianjin Anning Hospital, Tianjin, China; Chunshui Yu, MD, Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Chunshui Yu
- Chuanjun Zhuo, MD, PhD, Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, and Department of Psychiatry Functional Neuroimaging Laboratory, Tianjin Mental Health Center, Tianjin Anding Hospital, Tianjin; Jiajia Zhu, MD, Wen Qin, MD, Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, China; Hongru Qu, MD, Xiaolei Ma, MD, Department of Psychiatry, Tianjin Anning Hospital, Tianjin, China; Chunshui Yu, MD, Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
50
|
Bernard JA, Russell CE, Newberry RE, Goen JR, Mittal VA. Patients with schizophrenia show aberrant patterns of basal ganglia activation: Evidence from ALE meta-analysis. Neuroimage Clin 2017; 14:450-463. [PMID: 28275545 PMCID: PMC5328905 DOI: 10.1016/j.nicl.2017.01.034] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 12/23/2016] [Accepted: 01/31/2017] [Indexed: 12/29/2022]
Abstract
The diverse circuits and functional contributions of the basal ganglia, coupled with known differences in dopaminergic function in patients with schizophrenia, suggest they may be an important contributor to the etiology of the hallmark symptoms and cognitive dysfunction experienced by these patients. Using activation-likelihood-estimation meta-analysis of functional imaging research, we investigated differences in activation patterns in the basal ganglia in patients with schizophrenia, relative to healthy controls across task domains. This analysis included 42 functional neuroimaging studies, representing a variety of behavioral domains that have been linked to basal ganglia function in prior work. We provide important new information about the functional activation patterns and functional topography of the basal ganglia for different task domains in healthy controls. Crucially however, we demonstrate that across task domains, patients with schizophrenia show markedly decreased activation in the basal ganglia relative to healthy controls. Our results provide further support for basal ganglia dysfunction in patients with schizophrenia, and the broad dysfunction across task domains may contribute to the symptoms and cognitive deficits associated with schizophrenia.
Collapse
Affiliation(s)
- Jessica A. Bernard
- Department of Psychology, Texas A&M University, United States
- Texas A&M Institute for Neuroscience, Texas A&M University, United States
| | - Courtney E. Russell
- Department of Psychology & Neuroscience, University of Colorado Boulder, United States
| | - Raeana E. Newberry
- Department of Psychology & Neuroscience, University of Colorado Boulder, United States
| | - James R.M. Goen
- Department of Psychology, Texas A&M University, United States
| | - Vijay A. Mittal
- Department of Psychology, Northwestern University, United States
- Department of Psychiatry, Northwestern University, United States
- Institute for Policy Research, Northwestern University, United States
- Department of Medical Social Sciences, Northwestern University, United States
| |
Collapse
|