1
|
Bracher KM, Wohlschlaeger A, Koch K, Knolle F. Cognitive subgroups of affective and non-affective psychosis show differences in medication and cortico-subcortical brain networks. Sci Rep 2024; 14:20314. [PMID: 39223185 PMCID: PMC11369100 DOI: 10.1038/s41598-024-71316-3] [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: 03/29/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024] Open
Abstract
Cognitive deficits are prevalent in individuals with psychosis and are associated with neurobiological changes, potentially serving as an endophenotype for psychosis. Using the HCP-Early-Psychosis-dataset (n = 226), we aimed to investigate cognitive subtypes (deficit/intermediate/spared) through data-driven clustering in affective (AP) and non-affective psychosis patients (NAP) and controls (HC). We explored differences between three clusters in symptoms, cognition, medication, and grey matter volume. Applying principal component analysis, we selected features for clustering. Features that explained most variance were scores for intelligence, verbal recognition and comprehension, auditory attention, working memory, reasoning and executive functioning. Fuzzy K-Means clustering on those features revealed that the subgroups significantly varied in cognitive impairment, clinical symptoms, and, importantly, also in medication and grey matter volume in fronto-parietal and subcortical networks. The spared cluster (86%HC, 37%AP, 17%NAP) exhibited unimpaired cognition, lowest symptoms/medication, and grey matter comparable to controls. The deficit cluster (4%HC, 10%AP, 47%NAP) had impairments across all domains, highest symptoms scores/medication dosage, and pronounced grey matter alterations. The intermediate deficit cluster (11%HC, 54%AP, 36%NAP) showed fewer deficits than the second cluster, but similar symptoms/medication/grey matter to the spared cluster. Controlling for medication, cognitive scores correlated with grey matter changes and negative symptoms across all patients. Our findings generally emphasize the interplay between cognition, brain structure, symptoms, and medication in AP and NAP, and specifically suggest a possible mediating role of cognition, highlighting the potential of screening cognitive changes to aid tailoring treatments and interventions.
Collapse
Affiliation(s)
- Katharina M Bracher
- Division of Neurobiology, Faculty of Biology, LMU Munich, 82152, Martinsried, Germany
| | - Afra Wohlschlaeger
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Kathrin Koch
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Franziska Knolle
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Technical University of Munich, Munich, Germany.
| |
Collapse
|
2
|
Mana L, Schwartz-Pallejà M, Vila-Vidal M, Deco G. Overview on cognitive impairment in psychotic disorders: From impaired microcircuits to dysconnectivity. Schizophr Res 2024; 269:132-143. [PMID: 38788432 DOI: 10.1016/j.schres.2024.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024]
Abstract
Schizophrenia's cognitive deficits, often overshadowed by positive symptoms, significantly contribute to the disorder's morbidity. Increasing attention highlights these deficits as reflections of neural circuit dysfunction across various cortical regions. Numerous connectivity alterations linked to cognitive symptoms in psychotic disorders have been reported, both at the macroscopic and microscopic level, emphasizing the potential role of plasticity and microcircuits impairment during development and later stages. However, the heterogeneous clinical presentation of cognitive impairment and diverse connectivity findings pose challenges in summarizing them into a cohesive picture. This review aims to synthesize major cognitive alterations, recent insights into network structural and functional connectivity changes and proposed mechanisms and microcircuit alterations underpinning these symptoms, particularly focusing on neurodevelopmental impairment, E/I balance, and sleep disturbances. Finally, we will also comment on some of the most recent and promising therapeutic approaches that aim to target these mechanisms to address cognitive symptoms. Through this comprehensive exploration, we strive to provide an updated and nuanced overview of the multiscale connectivity impairment underlying cognitive impairment in psychotic disorders.
Collapse
Affiliation(s)
- L Mana
- Center for Brain and Cognition, Computational Neuroscience Group, Department of Information and Communication Technologies, Universitat Pompeu Fabra, Roc Boronat 138, Barcelona 08018, Spain.
| | - M Schwartz-Pallejà
- Center for Brain and Cognition, Computational Neuroscience Group, Department of Information and Communication Technologies, Universitat Pompeu Fabra, Roc Boronat 138, Barcelona 08018, Spain; Department of Experimental and Health Science, Universitat Pompeu Fabra, Roc Boronat 138, Barcelona 08018, Spain; Eurecat, Technology Center of Catalonia, Multimedia Technologies, Barcelona, Spain.
| | - M Vila-Vidal
- Center for Brain and Cognition, Computational Neuroscience Group, Department of Information and Communication Technologies, Universitat Pompeu Fabra, Roc Boronat 138, Barcelona 08018, Spain; Computational Biology and Complex Systems Group, Department of Physics, Universitat Politècnica de Catalunya, Barcelona, Spain.
| | - G Deco
- Center for Brain and Cognition, Computational Neuroscience Group, Department of Information and Communication Technologies, Universitat Pompeu Fabra, Roc Boronat 138, Barcelona 08018, Spain; Institució Catalana de la Recerca i Estudis Avançats (ICREA), Passeig Lluís Companys 23, Barcelona 08010, Spain.
| |
Collapse
|
3
|
Haikonen J, Szrinivasan R, Ojanen S, Rhee JK, Ryazantseva M, Sulku J, Zumaraite G, Lauri SE. GluK1 kainate receptors are necessary for functional maturation of parvalbumin interneurons regulating amygdala circuit function. Mol Psychiatry 2024:10.1038/s41380-024-02641-2. [PMID: 38942774 DOI: 10.1038/s41380-024-02641-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 06/30/2024]
Abstract
Parvalbumin expressing interneurons (PV INs) are key players in the local inhibitory circuits and their developmental maturation coincides with the onset of adult-type network dynamics in the brain. Glutamatergic signaling regulates emergence of the unique PV IN phenotype, yet the receptor mechanisms involved are not fully understood. Here we show that GluK1 subunit containing kainate receptors (KARs) are necessary for development and maintenance of the neurochemical and functional properties of PV INs in the lateral and basal amygdala (BLA). Ablation of GluK1 expression specifically from PV INs resulted in low parvalbumin expression and loss of characteristic high firing rate throughout development. In addition, we observed reduced spontaneous excitatory synaptic activity at adult GluK1 lacking PV INs. Intriguingly, inactivation of GluK1 expression in adult PV INs was sufficient to abolish their high firing rate and to reduce PV expression levels, suggesting a role for GluK1 in dynamic regulation of PV IN maturation state. The PV IN dysfunction in the absence of GluK1 perturbed the balance between evoked excitatory vs. inhibitory synaptic inputs and long-term potentiation (LTP) in LA principal neurons, and resulted in aberrant development of the resting-state functional connectivity between mPFC and BLA. Behaviorally, the absence of GluK1 from PV INs associated with hyperactivity and increased fear of novelty. These results indicate a critical role for GluK1 KARs in regulation of PV IN function across development and suggest GluK1 as a potential therapeutic target for pathologies involving PV IN malfunction.
Collapse
Affiliation(s)
- Joni Haikonen
- HiLife Neuroscience Center and Molecular and Integrative Biosciences Research Programme, University of Helsinki, Helsinki, Finland
| | - Rakenduvadhana Szrinivasan
- HiLife Neuroscience Center and Molecular and Integrative Biosciences Research Programme, University of Helsinki, Helsinki, Finland
| | - Simo Ojanen
- HiLife Neuroscience Center and Molecular and Integrative Biosciences Research Programme, University of Helsinki, Helsinki, Finland
| | - Jun Kyu Rhee
- HiLife Neuroscience Center and Molecular and Integrative Biosciences Research Programme, University of Helsinki, Helsinki, Finland
| | - Maria Ryazantseva
- HiLife Neuroscience Center and Molecular and Integrative Biosciences Research Programme, University of Helsinki, Helsinki, Finland
| | - Janne Sulku
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Gabija Zumaraite
- HiLife Neuroscience Center and Molecular and Integrative Biosciences Research Programme, University of Helsinki, Helsinki, Finland
| | - Sari E Lauri
- HiLife Neuroscience Center and Molecular and Integrative Biosciences Research Programme, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
4
|
Xu F, Ma J, Wang W, Li H. A longitudinal study of the brain structure network changes in HIV patients with ANI: combined VBM with SCN. Front Neurol 2024; 15:1388616. [PMID: 38694776 PMCID: PMC11061470 DOI: 10.3389/fneur.2024.1388616] [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: 02/23/2024] [Accepted: 04/04/2024] [Indexed: 05/04/2024] Open
Abstract
Background Despite the widespread adoption of combination antiretroviral therapy (cART) in managing HIV, the virus's impact on the brain structure of patients remains significant. This study aims to longitudinally explore the persistent effects of HIV on brain structure, focusing on changes in gray matter volume (GMV) and structural covariance network (SCN) among patients at the Asymptomatic Neurocognitive Impairment (ANI) stage. Methods This research involved 45 HIV patients diagnosed with ANI and 45 demographically matched healthy controls (HCs). The participants were observed over a 1.5-year period. Differences in GMV between groups were analyzed using voxel-based morphometry (VBM), while the graph theory model facilitated the establishment of topological metrics for assessing network indices. These differences were evaluated using two-sample t-tests and paired-sample t-tests, applying the network-based statistics method. Additionally, the study examined correlations between GMV and cognitive performance, as well as clinical variables. Results Compared with HCs, HIV patients demonstrated reduced GMV in the right middle temporal gyrus and left middle frontal gyrus (FWE, p < 0.05), along with decreased betweenness centrality (BC) in the left anterior cingulate and paracingulate cortex. Conversely, an increase in the clustering coefficient (Cp) was observed (FDR, p < 0.05). During the follow-up period, a decline in GMV in the right fusiform gyrus (FWE, p < 0.05) and a reduction in node efficiency (Ne) in the triangular part of the inferior frontal gyrus were noted compared with baseline measurements (FDR, p < 0.05). The SCN of HIV patients exhibited small-world properties across most sparsity levels (Sigma >1), and area under the curve (AUC) analysis revealed no significant statistical differences between groups. Conclusion The findings suggest that despite the administration of combination antiretroviral therapy (cART), HIV continues to exert slow and sustained damage on brain structures. However, when compared to HCs, the small-world properties of the patients' SCNs did not significantly differ, and the clustering coefficient, indicative of the overall information-processing capacity of the brain network, was slightly elevated in HIV patients. This elevation may relate to compensatory effects of brain area functions, the impact of cART, functional reorganization, or inflammatory responses.
Collapse
Affiliation(s)
| | | | | | - Hongjun Li
- Department of Radiology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
5
|
Negrón-Oyarzo I, Dib T, Chacana-Véliz L, López-Quilodrán N, Urrutia-Piñones J. Large-scale coupling of prefrontal activity patterns as a mechanism for cognitive control in health and disease: evidence from rodent models. Front Neural Circuits 2024; 18:1286111. [PMID: 38638163 PMCID: PMC11024307 DOI: 10.3389/fncir.2024.1286111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 03/11/2024] [Indexed: 04/20/2024] Open
Abstract
Cognitive control of behavior is crucial for well-being, as allows subject to adapt to changing environments in a goal-directed way. Changes in cognitive control of behavior is observed during cognitive decline in elderly and in pathological mental conditions. Therefore, the recovery of cognitive control may provide a reliable preventive and therapeutic strategy. However, its neural basis is not completely understood. Cognitive control is supported by the prefrontal cortex, structure that integrates relevant information for the appropriate organization of behavior. At neurophysiological level, it is suggested that cognitive control is supported by local and large-scale synchronization of oscillatory activity patterns and neural spiking activity between the prefrontal cortex and distributed neural networks. In this review, we focus mainly on rodent models approaching the neuronal origin of these prefrontal patterns, and the cognitive and behavioral relevance of its coordination with distributed brain systems. We also examine the relationship between cognitive control and neural activity patterns in the prefrontal cortex, and its role in normal cognitive decline and pathological mental conditions. Finally, based on these body of evidence, we propose a common mechanism that may underlie the impaired cognitive control of behavior.
Collapse
Affiliation(s)
- Ignacio Negrón-Oyarzo
- Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Tatiana Dib
- Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Lorena Chacana-Véliz
- Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Programa de Doctorado en Ciencias Mención en Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Nélida López-Quilodrán
- Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Programa de Doctorado en Ciencias Mención en Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Jocelyn Urrutia-Piñones
- Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Programa de Doctorado en Ciencias Mención en Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
6
|
Laricchiuta D, Papi M, Decandia D, Panuccio A, Cutuli D, Peciccia M, Mazzeschi C, Petrosini L. The role of glial cells in mental illness: a systematic review on astroglia and microglia as potential players in schizophrenia and its cognitive and emotional aspects. Front Cell Neurosci 2024; 18:1358450. [PMID: 38419655 PMCID: PMC10899480 DOI: 10.3389/fncel.2024.1358450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 01/29/2024] [Indexed: 03/02/2024] Open
Abstract
Schizophrenia is a complex and severe mental disorder that affects approximately 1% of the global population. It is characterized by a wide range of symptoms, including delusions, hallucinations, disorganized speech and behavior, and cognitive impairment. Recent research has suggested that the immune system dysregulation may play a significant role in the pathogenesis of schizophrenia, and glial cells, such as astroglia and microglia known to be involved in neuroinflammation and immune regulation, have emerged as potential players in this process. The aim of this systematic review is to summarize the glial hallmarks of schizophrenia, choosing as cellular candidate the astroglia and microglia, and focusing also on disease-associated psychological (cognitive and emotional) changes. We conducted a systematic review following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. We searched PubMed, Scopus, and Web of Science for articles that investigated the differences in astroglia and microglia in patients with schizophrenia, published in the last 5 years. The present systematic review indicates that changes in the density, morphology, and functioning of astroglia and microglia may be involved in the development of schizophrenia. The glial alterations may contribute to the pathogenesis of schizophrenia by dysregulating neurotransmission and immune responses, worsening cognitive capabilities. The complex interplay of astroglial and microglial activation, genetic/epigenetic variations, and cognitive assessments underscores the intricate relationship between biological mechanisms, symptomatology, and cognitive functioning in schizophrenia.
Collapse
Affiliation(s)
- Daniela Laricchiuta
- Department of Philosophy, Social Sciences and Education, University of Perugia, Perugia, Italy
| | - Martina Papi
- Department of Philosophy, Social Sciences and Education, University of Perugia, Perugia, Italy
| | - Davide Decandia
- Laboratory of Experimental and Behavioral Neurophysiology, IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Psychology, University Sapienza of Rome, Rome, Italy
| | - Anna Panuccio
- Laboratory of Experimental and Behavioral Neurophysiology, IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Psychology, University Sapienza of Rome, Rome, Italy
| | - Debora Cutuli
- Laboratory of Experimental and Behavioral Neurophysiology, IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Psychology, University Sapienza of Rome, Rome, Italy
| | - Maurizio Peciccia
- Department of Philosophy, Social Sciences and Education, University of Perugia, Perugia, Italy
| | - Claudia Mazzeschi
- Department of Philosophy, Social Sciences and Education, University of Perugia, Perugia, Italy
| | - Laura Petrosini
- Laboratory of Experimental and Behavioral Neurophysiology, IRCCS Santa Lucia Foundation, Rome, Italy
| |
Collapse
|
7
|
Thakuri DS, Bhattarai P, Wong DF, Chand GB. Dysregulated Salience Network Control over Default-Mode and Central-Executive Networks in Schizophrenia Revealed Using Stochastic Dynamical Causal Modeling. Brain Connect 2024; 14:70-79. [PMID: 38164105 PMCID: PMC10890948 DOI: 10.1089/brain.2023.0054] [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] [Indexed: 01/03/2024] Open
Abstract
Introduction: Neuroimaging studies suggest that the human brain consists of intrinsically organized, large-scale neural networks. Among these networks, the interplay among the default-mode network (DMN), salience network (SN), and central-executive network (CEN) has been widely used to understand the functional interaction patterns in health and disease. This triple network model suggests that the SN causally controls over the DMN and CEN in healthy individuals. This interaction is often referred to as SN's dynamic regulating mechanism. However, such interactions are not well understood in individuals with schizophrenia. Methods: In this study, we leveraged resting-state functional magnetic resonance imaging data from schizophrenia (n = 67) and healthy controls (n = 81) and evaluated the directional functional interactions among DMN, SN, and CEN using stochastic dynamical causal modeling methodology. Results: In healthy controls, our analyses replicated previous findings that SN regulates DMN and CEN activities (Mann-Whitney U test; p < 10-8). In schizophrenia, however, our analyses revealed a disrupted SN-based controlling mechanism over the DMN and CEN (Mann-Whitney U test; p < 10-16). Conclusions: These results indicate that the disrupted controlling mechanism of SN over the other two neural networks may be a candidate neuroimaging phenotype in schizophrenia.
Collapse
Affiliation(s)
- Deepa S. Thakuri
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
- Departments of Medicine and Radiology, University of Missouri School of Medicine, Columbia, Missouri, USA
| | - Puskar Bhattarai
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Dean F. Wong
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
- Departments of Neuroscience, Psychiatry, and Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
- Imaging Core, Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ganesh B. Chand
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
- Imaging Core, Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, USA
- Institute of Clinical and Translational Sciences, Washington University School of Medicine, St. Louis, Missouri, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
8
|
Zhang L, Shi W, Liu J, Chen K, Zhang G, Zhang S, Cong B, Li Y. Interleukin 6 (IL-6) Regulates GABAA Receptors in the Dorsomedial Hypothalamus Nucleus (DMH) through Activation of the JAK/STAT Pathway to Affect Heart Rate Variability in Stressed Rats. Int J Mol Sci 2023; 24:12985. [PMID: 37629166 PMCID: PMC10455568 DOI: 10.3390/ijms241612985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/09/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
The dorsomedial hypothalamus nucleus (DMH) is an important component of the autonomic nervous system and plays a critical role in regulating the sympathetic outputs of the heart. Stress alters the neuronal activity of the DMH, affecting sympathetic outputs and triggering heart rate variability. However, the specific molecular mechanisms behind stress leading to abnormal DMH neuronal activity have still not been fully elucidated. Therefore, in the present study, we successfully constructed a stressed rat model and used it to investigate the potential molecular mechanisms by which IL-6 regulates GABAA receptors in the DMH through activation of the JAK/STAT pathway and thus affects heart rate variability in rats. By detecting the c-Fos expression of neurons in the DMH and electrocardiogram (ECG) changes in rats, we clarified the relationship between abnormal DMH neuronal activity and heart rate variability in stressed rats. Then, using ELISA, immunohistochemical staining, Western blotting, RT-qPCR, and RNAscope, we further explored the correlation between the IL-6/JAK/STAT signaling pathway and GABAA receptors. The data showed that an increase in IL-6 induced by stress inhibited GABAA receptors in DMH neurons by activating the JAK/STAT signaling pathway, while specific inhibition of the JAK/STAT signaling pathway using AG490 obviously reduced DMH neuronal activity and improved heart rate variability in rats. These findings suggest that IL-6 regulates the expression of GABAA receptors via the activation of the JAK/STAT pathway in the DMH, which may be an important cause of heart rate variability in stressed rats.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Bin Cong
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Department of Forensic Medicine, Hebei Medical University, Shijiazhuang 050017, China; (L.Z.); (W.S.); (J.L.); (K.C.); (G.Z.); (S.Z.)
| | - Yingmin Li
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Department of Forensic Medicine, Hebei Medical University, Shijiazhuang 050017, China; (L.Z.); (W.S.); (J.L.); (K.C.); (G.Z.); (S.Z.)
| |
Collapse
|
9
|
Rukh S, Meechan DW, Maynard TM, Lamantia AS. Out of Line or Altered States? Neural Progenitors as a Target in a Polygenic Neurodevelopmental Disorder. Dev Neurosci 2023; 46:1-21. [PMID: 37231803 DOI: 10.1159/000530898] [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: 02/15/2023] [Accepted: 04/19/2023] [Indexed: 05/27/2023] Open
Abstract
The genesis of a mature complement of neurons is thought to require, at least in part, precursor cell lineages in which neural progenitors have distinct identities recognized by exclusive expression of one or a few molecular markers. Nevertheless, limited progenitor types distinguished by specific markers and lineal progression through such subclasses cannot easily yield the magnitude of neuronal diversity in most regions of the nervous system. The late Verne Caviness, to whom this edition of Developmental Neuroscience is dedicated, recognized this mismatch. In his pioneering work on the histogenesis of the cerebral cortex, he acknowledged the additional flexibility required to generate multiple classes of cortical projection and interneurons. This flexibility may be accomplished by establishing cell states in which levels rather than binary expression or repression of individual genes vary across each progenitor's shared transcriptome. Such states may reflect local, stochastic signaling via soluble factors or coincidence of cell surface ligand/receptor pairs in subsets of neighboring progenitors. This probabilistic, rather than determined, signaling could modify transcription levels via multiple pathways within an apparently uniform population of progenitors. Progenitor states, therefore, rather than lineal relationships between types may underlie the generation of neuronal diversity in most regions of the nervous system. Moreover, mechanisms that influence variation required for flexible progenitor states may be targets for pathological changes in a broad range of neurodevelopmental disorders, especially those with polygenic origins.
Collapse
Affiliation(s)
- Shah Rukh
- Fralin Biomedical Research Institute, Virginia Tech Carilion School of Medicine, Roanoke, Virginia, USA
| | - Daniel W Meechan
- Fralin Biomedical Research Institute, Virginia Tech Carilion School of Medicine, Roanoke, Virginia, USA
| | - Thomas M Maynard
- Fralin Biomedical Research Institute, Virginia Tech Carilion School of Medicine, Roanoke, Virginia, USA
| | - Anthony-Samuel Lamantia
- Fralin Biomedical Research Institute, Virginia Tech Carilion School of Medicine, Roanoke, Virginia, USA
- Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia, USA
| |
Collapse
|
10
|
De Simone G, Mazza B, Vellucci L, Barone A, Ciccarelli M, de Bartolomeis A. Schizophrenia Synaptic Pathology and Antipsychotic Treatment in the Framework of Oxidative and Mitochondrial Dysfunction: Translational Highlights for the Clinics and Treatment. Antioxidants (Basel) 2023; 12:antiox12040975. [PMID: 37107350 PMCID: PMC10135787 DOI: 10.3390/antiox12040975] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/05/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Schizophrenia is a worldwide mental illness characterized by alterations at dopaminergic and glutamatergic synapses resulting in global dysconnectivity within and between brain networks. Impairments in inflammatory processes, mitochondrial functions, energy expenditure, and oxidative stress have been extensively associated with schizophrenia pathophysiology. Antipsychotics, the mainstay of schizophrenia pharmacological treatment and all sharing the common feature of dopamine D2 receptor occupancy, may affect antioxidant pathways as well as mitochondrial protein levels and gene expression. Here, we systematically reviewed the available evidence on antioxidants' mechanisms in antipsychotic action and the impact of first- and second-generation compounds on mitochondrial functions and oxidative stress. We further focused on clinical trials addressing the efficacy and tolerability of antioxidants as an augmentation strategy of antipsychotic treatment. EMBASE, Scopus, and Medline/PubMed databases were interrogated. The selection process was conducted in respect of the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) criteria. Several mitochondrial proteins involved in cell viability, energy metabolism, and regulation of oxidative systems were reported to be significantly modified by antipsychotic treatment with differences between first- and second-generation drugs. Finally, antioxidants may affect cognitive and psychotic symptoms in patients with schizophrenia, and although the evidence is only preliminary, the results indicate that further studies are warranted.
Collapse
Affiliation(s)
- Giuseppe De Simone
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences, and Dentistry, University Medical School of Naples "Federico II", Via Pansini 5, 80131 Naples, Italy
| | - Benedetta Mazza
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences, and Dentistry, University Medical School of Naples "Federico II", Via Pansini 5, 80131 Naples, Italy
| | - Licia Vellucci
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences, and Dentistry, University Medical School of Naples "Federico II", Via Pansini 5, 80131 Naples, Italy
| | - Annarita Barone
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences, and Dentistry, University Medical School of Naples "Federico II", Via Pansini 5, 80131 Naples, Italy
| | - Mariateresa Ciccarelli
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences, and Dentistry, University Medical School of Naples "Federico II", Via Pansini 5, 80131 Naples, Italy
| | - Andrea de Bartolomeis
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences, and Dentistry, University Medical School of Naples "Federico II", Via Pansini 5, 80131 Naples, Italy
- UNESCO Chair on Health Education and Sustainable Development, University of Naples "Federico II", 80131 Naples, Italy
| |
Collapse
|
11
|
Moya NA, Yun S, Fleps SW, Martin MM, Nadel JA, Beutler LR, Zweifel LS, Parker JG. The effect of selective nigrostriatal dopamine excess on behaviors linked to the cognitive and negative symptoms of schizophrenia. Neuropsychopharmacology 2023; 48:690-699. [PMID: 36380221 PMCID: PMC9938164 DOI: 10.1038/s41386-022-01492-1] [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: 08/21/2022] [Revised: 10/16/2022] [Accepted: 10/25/2022] [Indexed: 11/16/2022]
Abstract
Excess dopamine release in the dorsal striatum (DS) is linked to psychosis. Antipsychotics are thought to work by blocking striatal D2 dopamine receptors, but they lack efficacy for the negative and cognitive symptoms of schizophrenia. These observations and the fact that increasing brain-wide dopamine improves cognition have fueled the dogma that excess dopamine is not involved in negative and cognitive symptoms. However, this idea has never been explicitly tested with DS-pathway specificity. To determine if excess DS dopamine is involved in cognitive and negative symptoms, we selectively re-expressed excitatory TRPV1 receptors in DS-projecting dopamine neurons of Trpv1 knockout mice. We treated these mice with capsaicin (TRPV1 agonist) to selectively activate these neurons, validated this approach with fiber photometry, and assessed its effects on social interaction and working memory, behavioral constructs related to negative and cognitive symptoms. We combined this manipulation with antipsychotic treatment (haloperidol) and compared it to brain-wide dopamine release via amphetamine treatment. We found that selectively activating DS-projecting dopamine neurons increased DS (but not cortical) dopamine release and increased locomotor activity. Surprisingly, this manipulation also impaired social interaction and working memory. Haloperidol normalized locomotion, but only partially rescued working memory and had no effect on social interaction. By contrast, amphetamine increased locomotion but did not impair social interaction or working memory. These results suggest that excess dopamine release, when restricted to the DS, causes behavioral deficits linked to negative and cognitive symptoms. Future therapies should address this disregarded role for excess striatal dopamine in the treatment-resistant symptoms of psychosis.
Collapse
Affiliation(s)
- Nicolette A Moya
- Departments of Neuroscience and Pharmacology, Northwestern University, Chicago, IL, USA
| | - Seongsik Yun
- Departments of Neuroscience and Pharmacology, Northwestern University, Chicago, IL, USA
| | - Stefan W Fleps
- Departments of Neuroscience and Pharmacology, Northwestern University, Chicago, IL, USA
| | - Madison M Martin
- Departments of Neuroscience and Pharmacology, Northwestern University, Chicago, IL, USA
| | - Jacob A Nadel
- Departments of Neuroscience and Pharmacology, Northwestern University, Chicago, IL, USA
| | - Lisa R Beutler
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University, Chicago, IL, USA
| | - Larry S Zweifel
- Departments of Psychiatry and Behavioral Sciences and Pharmacology, University of Washington, Seattle, WA, USA
| | - Jones G Parker
- Departments of Neuroscience and Pharmacology, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
12
|
Prieto-Alcántara M, Ibáñez-Molina A, Crespo-Cobo Y, Molina R, Soriano MF, Iglesias-Parro S. Alpha and gamma EEG coherence during on-task and mind wandering states in schizophrenia. Clin Neurophysiol 2023; 146:21-29. [PMID: 36495599 DOI: 10.1016/j.clinph.2022.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 10/12/2022] [Accepted: 11/13/2022] [Indexed: 11/27/2022]
Abstract
OBJECTIVE Electroencephalographic (EEG) coherence is one of the most relevant physiological measures used to detect abnormalities in patients with schizophrenia. The present study applies a task-related EEG coherence approach to understand cognitive processing in patients with schizophrenia and healthy controls. METHODS EEG coherence for alpha and gamma frequency bands was analyzed in a group of patients with schizophrenia and a group of healthy controls during the performance of an ecological task of sustained attention. We compared EEG coherence when participants presented externally directed cognitive states (On-Task) and when they presented cognitive distraction episodes (Mind-Wandering). RESULTS Results reflect cortical differences between groups (higher coherence for schizophrenia in the frontocentral and fronto-temporal regions, and higher coherence for healthy-controls in the postero-central regions), especially in the On-Task condition for the alpha band, compared to Mind-Wandering episodes. Few individual differences in gamma coherence were found. CONCLUSIONS The current study provides evidence of neurophysiological differences underlying different cognitive states in schizophrenia and healthy controls. SIGNIFICANCE Differences between groups may reflect inhibitory processes necessary for the successful processing of information, especially in the alpha band, given its role in cortical inhibition processes. Patients may activate compensatory inhibitory mechanisms when performing the task, reflected in increased coherence in fronto-temporal regions.
Collapse
Affiliation(s)
| | | | | | - Rosa Molina
- Psychology Department, University of Jaén, 23071 Jaén, Spain
| | | | | |
Collapse
|
13
|
Angulo Salavarria MM, Dell’Amico C, D’Agostino A, Conti L, Onorati M. Cortico-thalamic development and disease: From cells, to circuits, to schizophrenia. Front Neuroanat 2023; 17:1130797. [PMID: 36935652 PMCID: PMC10019505 DOI: 10.3389/fnana.2023.1130797] [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: 12/23/2022] [Accepted: 02/09/2023] [Indexed: 03/06/2023] Open
Abstract
The human brain is the most complex structure generated during development. Unveiling the ontogenesis and the intrinsic organization of specific neural networks may represent a key to understanding the physio-pathological aspects of different brain areas. The cortico-thalamic and thalamo-cortical (CT-TC) circuits process and modulate essential tasks such as wakefulness, sleep and memory, and their alterations may result in neurodevelopmental and psychiatric disorders. These pathologies are reported to affect specific neural populations but may also broadly alter physiological connections and thus dysregulate brain network generation, communication, and function. More specifically, the CT-TC system is reported to be severely affected in disorders impacting superior brain functions, such as schizophrenia (SCZ), bipolar disorder, autism spectrum disorders or epilepsy. In this review, the focus will be on CT development, and the models exploited to uncover and comprehend its molecular and cellular mechanisms. In parallel to animal models, still fundamental to unveil human neural network establishment, advanced in vitro platforms, such as brain organoids derived from human pluripotent stem cells, will be discussed. Indeed, organoids and assembloids represent unique tools to study and accelerate fundamental research in CT development and its dysfunctions. We will then discuss recent cutting-edge contributions, including in silico approaches, concerning ontogenesis, specification, and function of the CT-TC circuitry that generates connectivity maps in physiological and pathological conditions.
Collapse
Affiliation(s)
| | - Claudia Dell’Amico
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, Pisa, Italy
| | - Armando D’Agostino
- Department of Health Sciences, University of Milan, Milan, Italy
- Department of Mental Health and Addictions, ASST Santi Paolo e Carlo, Milan, Italy
| | - Luciano Conti
- Department of Cellular, Computational, and Integrative Biology, University of Trento, Trento, Italy
| | - Marco Onorati
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, Pisa, Italy
- *Correspondence: Marco Onorati,
| |
Collapse
|
14
|
Micale V, Di Bartolomeo M, Di Martino S, Stark T, Dell'Osso B, Drago F, D'Addario C. Are the epigenetic changes predictive of therapeutic efficacy for psychiatric disorders? A translational approach towards novel drug targets. Pharmacol Ther 2023; 241:108279. [PMID: 36103902 DOI: 10.1016/j.pharmthera.2022.108279] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 09/01/2022] [Accepted: 09/01/2022] [Indexed: 02/06/2023]
Abstract
The etiopathogenesis of mental disorders is not fully understood and accumulating evidence support that clinical symptomatology cannot be assigned to a single gene mutation, but it involves several genetic factors. More specifically, a tight association between genes and environmental risk factors, which could be mediated by epigenetic mechanisms, may play a role in the development of mental disorders. Several data suggest that epigenetic modifications such as DNA methylation, post-translational histone modification and interference of microRNA (miRNA) or long non-coding RNA (lncRNA) may modify the severity of the disease and the outcome of the therapy. Indeed, the study of these mechanisms may help to identify patients particularly vulnerable to mental disorders and may have potential utility as biomarkers to facilitate diagnosis and treatment of psychiatric disorders. This article summarizes the most relevant preclinical and human data showing how epigenetic modifications can be central to the therapeutic efficacy of antidepressant and/or antipsychotic agents, as possible predictor of drugs response.
Collapse
Affiliation(s)
- Vincenzo Micale
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy.
| | - Martina Di Bartolomeo
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Serena Di Martino
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy
| | - Tibor Stark
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czech Republic; Scientific Core Unit Neuroimaging, Max Planck Institute of Psychiatry, Munich, Germany
| | - Bernardo Dell'Osso
- Department of Biomedical and Clinical Sciences 'Luigi Sacco', University of Milan, Milan, Italy, Department of Mental Health, ASST Fatebenefratelli-Sacco, Milan, Italy; "Aldo Ravelli" Research Center for Neurotechnology and Experimental Brain Therapeutics, Department of Health Sciences, University of Milan Medical School, Milan, Italy; Department of Psychiatry and Behavioral Sciences, Stanford University, CA, USA
| | - Filippo Drago
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy.
| | - Claudio D'Addario
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy; Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
15
|
Dong J, Chen W, Liu N, Chang S, Zhu W, Kang J. NRG1 knockdown rescues PV interneuron GABAergic maturation deficits and schizophrenia behaviors in fetal growth restriction mice. Cell Death Dis 2022; 8:476. [PMID: 36460658 PMCID: PMC9718849 DOI: 10.1038/s41420-022-01271-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 11/22/2022] [Accepted: 11/22/2022] [Indexed: 12/04/2022]
Abstract
Schizophrenia is a highly debilitating mental disorder, those who experienced fetal growth restriction (FGR) in the early stage of life have a greater probability of schizophrenia. In this study, FGR mice showed hyperactivity in locomotor activity test, sociability dysfunction in three chamber test and nesting social behavior tests, cognition decline in Morris water maze and impaired sensory motor gating function in prepulse inhibition test. Mechanistic studies indicated that the number of parvalbumin (PV) interneuron was significantly reduced in FGR mouse media prefrontal cortex (mPFC). And the mRNA and protein level of neuregulin 1(NRG1), which is a critical schizophrenia gene, increased significantly in FGR mouse mPFC. Furthermore, NRG1 knockdown in FGR mouse mPFC improved PV interneuron GABAergic maturation and rescued schizophrenia behaviors including hyperactivity, social novelty defects, cognition decline, and sensorimotor gating deficits in FGR mice. This study indicates that mPFC NRG1 upregulation is one of the main causes of FGR-induced schizophrenia, which leads to significant reduction of PV interneuron number in mPFC. NRG1 knockdown in mPFC significantly rescues schizophrenia behaviors in FGR mouse. This study thus provides a potential effective therapy target or strategy for schizophrenia patients induced by FGR.
Collapse
Affiliation(s)
- Jianfeng Dong
- grid.24516.340000000123704535Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Institute for Advanced Study, Tongji University, Shanghai, China
| | - Wen Chen
- grid.24516.340000000123704535Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Institute for Advanced Study, Tongji University, Shanghai, China
| | - Nana Liu
- grid.24516.340000000123704535Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Institute for Advanced Study, Tongji University, Shanghai, China
| | - Shujuan Chang
- grid.24516.340000000123704535Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Institute for Advanced Study, Tongji University, Shanghai, China
| | - Wei Zhu
- grid.24516.340000000123704535Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Institute for Advanced Study, Tongji University, Shanghai, China
| | - Jiuhong Kang
- grid.24516.340000000123704535Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Institute for Advanced Study, Tongji University, Shanghai, China
| |
Collapse
|
16
|
Associations between polygenic risk, negative symptoms, and functional connectome topology during a working memory task in early-onset schizophrenia. SCHIZOPHRENIA 2022; 8:54. [PMID: 35853905 PMCID: PMC9261080 DOI: 10.1038/s41537-022-00260-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 05/12/2022] [Indexed: 11/18/2022]
Abstract
Working memory (WM) deficit in schizophrenia is thought to arise from a widespread neural inefficiency. However, we do not know if this deficit results from the illness-related genetic risk and influence the symptom burden in various domains, especially in patients who have an early onset illness. We used graph theory to examine the topology of the functional connectome in 99 subjects (27 early-onset schizophrenia (EOS), 24 asymptomatic siblings, and 48 healthy subjects) during an n-back task, and calculated their polygenic risk score (PRS) for susceptibility to schizophrenia. Linear regression analysis was used to test associations of the PRS, clinical symptoms, altered connectomic properties, and WM accuracy in EOS. Indices of small-worldness and segregation were elevated in EOS during the WM task compared with the other two groups; these connectomic aberrations correlated with increased PRS and negative symptoms. In patients with higher polygenic risk, WM performance was lower only when both the connectomic aberrations and the burden of negative symptoms were higher. Negative symptoms had a stronger moderating role in this relationship. Our findings suggest that the aberrant connectomic topology is a feature of WM task performance in schizophrenia; this relates to higher polygenic risk score as well as higher burden of negative symptoms. The deleterious effects of polygenic risk on cognition are played out via its effects on the functional connectome, as well as negative symptoms.
Collapse
|
17
|
Armenta-Resendiz M, Assali A, Tsvetkov E, Cowan CW, Lavin A. Repeated methamphetamine administration produces cognitive deficits through augmentation of GABAergic synaptic transmission in the prefrontal cortex. Neuropsychopharmacology 2022; 47:1816-1825. [PMID: 35788684 PMCID: PMC9372065 DOI: 10.1038/s41386-022-01371-9] [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: 05/02/2022] [Revised: 06/14/2022] [Accepted: 06/22/2022] [Indexed: 11/08/2022]
Abstract
Methamphetamine (METH) abuse is associated with the emergence of cognitive deficits and hypofrontality, a pathophysiological marker of many neuropsychiatric disorders that is produced by altered balance of local excitatory and inhibitory synaptic transmission. However, there is a dearth of information regarding the cellular and synaptic mechanisms underlying METH-induced cognitive deficits and associated hypofrontal states. Using PV-Cre transgenic rats that went through a METH sensitization regime or saline (SAL) followed by 7-10 days of home cage abstinence combined with cognitive tests, chemogenetic experiments, and whole-cell patch recordings on the prelimbic prefrontal cortex (PFC), we investigated the cellular and synaptic mechanisms underlying METH-induce hypofrontality. We report here that repeated METH administration in rats produces deficits in working memory and increases in inhibitory synaptic transmission onto pyramidal neurons in the PFC. The increased PFC inhibition is detected by an increase in spontaneous and evoked inhibitory postsynaptic synaptic currents (IPSCs), an increase in GABAergic presynaptic function, and a shift in the excitatory-inhibitory balance onto PFC deep-layer pyramidal neurons. We find that pharmacological blockade of D1 dopamine receptor function reduces the METH-induced augmentation of IPSCs, suggesting a critical role for D1 dopamine signaling in METH-induced hypofrontality. In addition, repeated METH administration increases the intrinsic excitability of parvalbumin-positive fast spiking interneurons (PV + FSIs), a key local interneuron population in PFC that contributes to the control of inhibitory tone. Using a cell type-specific chemogenetic approach, we show that increasing PV + FSIs activity in the PFC is necessary and sufficient to cause deficits in temporal order memory similar to those induced by METH. Conversely, reducing PV + FSIs activity in the PFC of METH-exposed rats rescues METH-induced temporal order memory deficits. Together, our findings reveal that repeated METH exposure increases PFC inhibitory tone through a D1 dopamine signaling-dependent potentiation of inhibitory synaptic transmission, and that reduction of PV + FSIs activity can rescue METH-induced cognitive deficits, suggesting a potential therapeutic approach to treating cognitive symptoms in patients suffering from METH use disorder.
Collapse
Affiliation(s)
| | - Ahlem Assali
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Evgeny Tsvetkov
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Christopher W Cowan
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Antonieta Lavin
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
18
|
de Bartolomeis A, Barone A, Vellucci L, Mazza B, Austin MC, Iasevoli F, Ciccarelli M. Linking Inflammation, Aberrant Glutamate-Dopamine Interaction, and Post-synaptic Changes: Translational Relevance for Schizophrenia and Antipsychotic Treatment: a Systematic Review. Mol Neurobiol 2022; 59:6460-6501. [PMID: 35963926 PMCID: PMC9463235 DOI: 10.1007/s12035-022-02976-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 07/24/2022] [Indexed: 12/16/2022]
Abstract
Evidence from clinical, preclinical, and post-mortem studies supports the inflammatory/immune hypothesis of schizophrenia pathogenesis. Less evident is the link between the inflammatory background and two well-recognized functional and structural findings of schizophrenia pathophysiology: the dopamine-glutamate aberrant interaction and the alteration of dendritic spines architecture, both believed to be the “quantal” elements of cortical-subcortical dysfunctional network. In this systematic review, we tried to capture the major findings linking inflammation, aberrant glutamate-dopamine interaction, and post-synaptic changes under a direct and inverse translational perspective, a paramount picture that at present is lacking. The inflammatory effects on dopaminergic function appear to be bidirectional: the inflammation influences dopamine release, and dopamine acts as a regulator of discrete inflammatory processes involved in schizophrenia such as dysregulated interleukin and kynurenine pathways. Furthermore, the link between inflammation and glutamate is strongly supported by clinical studies aimed at exploring overactive microglia in schizophrenia patients and maternal immune activation models, indicating impaired glutamate regulation and reduced N-methyl-D-aspartate receptor (NMDAR) function. In addition, an inflammatory/immune-induced alteration of post-synaptic density scaffold proteins, crucial for downstream NMDAR signaling and synaptic efficacy, has been demonstrated. According to these findings, a significant increase in plasma inflammatory markers has been found in schizophrenia patients compared to healthy controls, associated with reduced cortical integrity and functional connectivity, relevant to the cognitive deficit of schizophrenia. Finally, the link between altered inflammatory/immune responses raises relevant questions regarding potential new therapeutic strategies specifically for those forms of schizophrenia that are resistant to canonical antipsychotics or unresponsive to clozapine.
Collapse
Affiliation(s)
- Andrea de Bartolomeis
- Laboratory of Molecular and Translational Psychiatry, University School of Medicine of Naples Federico II, Naples, Italy. .,Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, University School of Medicine of Naples Federico II, Naples, Italy.
| | - Annarita Barone
- Laboratory of Molecular and Translational Psychiatry, University School of Medicine of Naples Federico II, Naples, Italy.,Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, University School of Medicine of Naples Federico II, Naples, Italy
| | - Licia Vellucci
- Laboratory of Molecular and Translational Psychiatry, University School of Medicine of Naples Federico II, Naples, Italy.,Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, University School of Medicine of Naples Federico II, Naples, Italy
| | - Benedetta Mazza
- Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, University School of Medicine of Naples Federico II, Naples, Italy
| | - Mark C Austin
- Clinical Psychopharmacology Program, College of Pharmacy, Idaho State University (ISU), Pocatello, ID, USA
| | - Felice Iasevoli
- Laboratory of Molecular and Translational Psychiatry, University School of Medicine of Naples Federico II, Naples, Italy.,Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, University School of Medicine of Naples Federico II, Naples, Italy
| | - Mariateresa Ciccarelli
- Laboratory of Molecular and Translational Psychiatry, University School of Medicine of Naples Federico II, Naples, Italy.,Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, University School of Medicine of Naples Federico II, Naples, Italy
| |
Collapse
|
19
|
Harrison PJ, Mould A, Tunbridge EM. New drug targets in psychiatry: Neurobiological considerations in the genomics era. Neurosci Biobehav Rev 2022; 139:104763. [PMID: 35787892 DOI: 10.1016/j.neubiorev.2022.104763] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 05/15/2022] [Accepted: 06/14/2022] [Indexed: 01/11/2023]
Abstract
After a period of withdrawal, pharmaceutical companies have begun to reinvest in neuropsychiatric disorders, due to improvements in our understanding of these disorders, stimulated in part by genomic studies. However, translating this information into disease insights and ultimately into tractable therapeutic targets is a major challenge. Here we consider how different sources of information might be integrated to guide this process. We review how an understanding of neurobiology has been used to advance therapeutic candidates identified in the pre-genomic era, using catechol-O-methyltransferase (COMT) as an exemplar. We then contrast with ZNF804A, the first genome-wide significant schizophrenia gene, and draw on some of the lessons that these and other examples provide. We highlight that, at least in the short term, the translation of potential targets for which there is orthogonal neurobiological support is likely to be more straightforward and productive than that those relying solely on genomic information. Although we focus here on information from genomic studies of schizophrenia, the points are broadly applicable across major psychiatric disorders and their symptoms.
Collapse
Affiliation(s)
- Paul J Harrison
- Department of Psychiatry, University of Oxford, Oxford, UK; Oxford Health NHS Foundation Trust, Oxford, UK
| | - Arne Mould
- Department of Psychiatry, University of Oxford, Oxford, UK; Oxford Health NHS Foundation Trust, Oxford, UK
| | - Elizabeth M Tunbridge
- Department of Psychiatry, University of Oxford, Oxford, UK; Oxford Health NHS Foundation Trust, Oxford, UK.
| |
Collapse
|
20
|
Ueno H, Takahashi Y, Murakami S, Wani K, Matsumoto Y, Okamoto M, Ishihara T. Fingolimod increases parvalbumin-positive neurons in adult mice. IBRO Neurosci Rep 2022; 13:96-106. [PMID: 36590091 PMCID: PMC9795291 DOI: 10.1016/j.ibneur.2022.06.005] [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: 01/25/2022] [Revised: 06/28/2022] [Accepted: 06/29/2022] [Indexed: 01/04/2023] Open
Abstract
In recent years, it has been shown that central nervous system agents, such as antidepressants and antiepileptic drugs, reopen a critical period in mature animals. Fingolimod, which is used for the treatment of multiple sclerosis, also restores neuroplasticity. In this study, we investigated the effects of parvalbumin (PV)-positive neurons and perineuronal nets (PNN) on fingolimod administration with respect to neuroplasticity. Fingolimod was chronically administered intraperitoneally to mature mice. PV-positive neurons and PNN in the hippocampus, prefrontal cortex, and somatosensory cortex were analyzed. An increase in PV-positive neurons was observed in the hippocampus, prefrontal cortex, and somatosensory cortex of the fingolimod-treated mice. An increase in Wisteria floribunda agglutinin-positive PNN was confirmed in mice treated with fingolimod in the somatosensory cortex only. Fingolimod increased the density of PV-positive neurons in the brains of mature mice. The results indicate that fingolimod may change the critical period in mature animals.
Collapse
Key Words
- CNS, central nervous system
- ECM, extracellular matrix
- Fingolimod
- GAD67, anti-glutamic acid decarboxylase
- GFAP, glial fibrillary acidic protein
- Hippocampus
- IL, infralimbic cortex
- NIH, National Institutes of Health, PBS, phosphate-buffered saline
- PL, prelimbic cortex
- PNN, perineuronal net
- PV neurons, parvalbumin-expressing interneurons
- Parvalbumin
- Perineuronal nets
- Prefrontal cortex
- Somatosensory cortex
- WFA, Wisteria floribunda agglutinin
- dAC, dorsal anterior cingulate cortex
Collapse
Affiliation(s)
- Hiroshi Ueno
- Department of Medical Technology, Kawasaki University of Medical Welfare, Kurashiki 701–0193, Japan,Correspondence to: Department of Medical Technology, Kawasaki University of Medical Welfare, 288 Matsushima, Kurashiki, Okayama 701–0193, Japan, 193.
| | - Yu Takahashi
- Department of Psychiatry, Kawasaki Medical School, Kurashiki 701–0192, Japan
| | - Shinji Murakami
- Department of Psychiatry, Kawasaki Medical School, Kurashiki 701–0192, Japan
| | - Kenta Wani
- Department of Psychiatry, Kawasaki Medical School, Kurashiki 701–0192, Japan
| | - Yosuke Matsumoto
- Department of Neuropsychiatry, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700–8558, Japan
| | - Motoi Okamoto
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, Okayama 700–8558, Japan
| | - Takeshi Ishihara
- Department of Psychiatry, Kawasaki Medical School, Kurashiki 701–0192, Japan
| |
Collapse
|
21
|
Sultan FA, Sawaya BE. Gadd45 in Neuronal Development, Function, and Injury. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1360:117-148. [PMID: 35505167 DOI: 10.1007/978-3-030-94804-7_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The growth arrest and DNA damage-inducible (Gadd) 45 proteins have been associated with numerous cellular mechanisms including cell cycle control, DNA damage sensation and repair, genotoxic stress, neoplasia, and molecular epigenetics. The genes were originally identified in in vitro screens of irradiation- and interleukin-induced transcription and have since been implicated in a host of normal and aberrant central nervous system processes. These include early and postnatal development, injury, cancer, memory, aging, and neurodegenerative and psychiatric disease states. The proteins act through a variety of molecular signaling cascades including the MAPK cascade, cell cycle control mechanisms, histone regulation, and epigenetic DNA demethylation. In this review, we provide a comprehensive discussion of the literature implicating each of the three members of the Gadd45 family in these processes.
Collapse
Affiliation(s)
- Faraz A Sultan
- Department of Psychiatry, Rush University, Chicago, IL, USA.
| | - Bassel E Sawaya
- Molecular Studies of Neurodegenerative Diseases Lab, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.,FELS Cancer Institute for Personalized Medicine Institute, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.,Departments of Neurology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.,Cancer and Cell Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.,Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| |
Collapse
|
22
|
de Oliveira Figueiredo EC, Bondiolotti BM, Laugeray A, Bezzi P. Synaptic Plasticity Dysfunctions in the Pathophysiology of 22q11 Deletion Syndrome: Is There a Role for Astrocytes? Int J Mol Sci 2022; 23:ijms23084412. [PMID: 35457231 PMCID: PMC9028090 DOI: 10.3390/ijms23084412] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/14/2022] [Accepted: 04/15/2022] [Indexed: 01/01/2023] Open
Abstract
The 22q11 deletion syndrome (DS) is the most common microdeletion syndrome in humans and gives a high probability of developing psychiatric disorders. Synaptic and neuronal malfunctions appear to be at the core of the symptoms presented by patients. In fact, it has long been suggested that the behavioural and cognitive impairments observed in 22q11DS are probably due to alterations in the mechanisms regulating synaptic function and plasticity. Often, synaptic changes are related to structural and functional changes observed in patients with cognitive dysfunctions, therefore suggesting that synaptic plasticity has a crucial role in the pathophysiology of the syndrome. Most interestingly, among the genes deleted in 22q11DS, six encode for mitochondrial proteins that, in mouse models, are highly expressed just after birth, when active synaptogenesis occurs, therefore indicating that mitochondrial processes are strictly related to synapse formation and maintenance of a correct synaptic signalling. Because correct synaptic functioning, not only requires correct neuronal function and metabolism, but also needs the active contribution of astrocytes, we summarize in this review recent studies showing the involvement of synaptic plasticity in the pathophysiology of 22q11DS and we discuss the relevance of mitochondria in these processes and the possible involvement of astrocytes.
Collapse
Affiliation(s)
| | - Bianca Maria Bondiolotti
- Department of Fundamental Neurosciences, University of Lausanne, 1005 Lausanne, Switzerland; (E.C.d.O.F.); (B.M.B.); (A.L.)
| | - Anthony Laugeray
- Department of Fundamental Neurosciences, University of Lausanne, 1005 Lausanne, Switzerland; (E.C.d.O.F.); (B.M.B.); (A.L.)
| | - Paola Bezzi
- Department of Fundamental Neurosciences, University of Lausanne, 1005 Lausanne, Switzerland; (E.C.d.O.F.); (B.M.B.); (A.L.)
- Department of Pharmacology and Physiology, University of Rome Sapienza, 00185 Rome, Italy
- Correspondence: or
| |
Collapse
|
23
|
Cruz-Santos M, Cardo LF, Li M. A Novel LHX6 Reporter Cell Line for Tracking Human iPSC-Derived Cortical Interneurons. Cells 2022; 11:cells11050853. [PMID: 35269475 PMCID: PMC8909769 DOI: 10.3390/cells11050853] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/16/2022] [Accepted: 02/25/2022] [Indexed: 12/15/2022] Open
Abstract
GABAergic interneurons control the neural circuitry and network activity in the brain. The dysfunction of cortical interneurons, especially those derived from the medial ganglionic eminence, contributes to neurological disease states. Pluripotent stem cell-derived interneurons provide a powerful tool for understanding the etiology of neuropsychiatric disorders, as well as having the potential to be used as medicine in cell therapy for neurological conditions such as epilepsy. Although large numbers of interneuron progenitors can be readily induced in vitro, the generation of defined interneuron subtypes remains inefficient. Using CRISPR/Cas9-assisted homologous recombination in hPSCs, we inserted the coding sequence of mEmerald and mCherry fluorescence protein, respectively, downstream that of the LHX6, a gene required for, and a marker of medial ganglionic eminence (MGE)-derived cortical interneurons. Upon differentiation of the LHX6-mEmerald and LHX6-mCherry hPSCs towards the MGE fate, both reporters exhibited restricted expression in LHX6+ MGE derivatives of hPSCs. Moreover, the reporter expression responded to changes of interneuron inductive cues. Thus, the LHX6-reporter lines represent a valuable tool to identify molecules controlling human interneuron development and design better interneuron differentiation protocols as well as for studying risk genes associated with interneuronopathies.
Collapse
Affiliation(s)
- Maria Cruz-Santos
- Neuroscience and Mental Health Research Institute, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK; (M.C.-S.); (L.F.C.)
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
| | - Lucia Fernandez Cardo
- Neuroscience and Mental Health Research Institute, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK; (M.C.-S.); (L.F.C.)
| | - Meng Li
- Neuroscience and Mental Health Research Institute, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK; (M.C.-S.); (L.F.C.)
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
- Correspondence:
| |
Collapse
|
24
|
Identification of TGFβ signaling as a regulator of interneuron neurogenesis in a human pluripotent stem cell model. Neuronal Signal 2021; 5:NS20210020. [PMID: 34956651 PMCID: PMC8661503 DOI: 10.1042/ns20210020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 11/17/2022] Open
Abstract
Cortical interneurons are GABAergic inhibitory cells that connect locally in the neocortex and play a pivotal role in shaping cortical network activities. Dysfunction of these cells is believed to lead to runaway excitation underlying seizure-based diseases, such as epilepsy, autism and schizophrenia. There is a growing interest in using cortical interneurons derived from human pluripotent stem cells for understanding their complex development and for modeling neuropsychiatric diseases. Here, we report the identification of a novel role of transforming growth factor β (TGFβ) signaling in modulating interneuron progenitor maintenance and neuronal differentiation. TGFβ signaling inhibition suppresses terminal differentiation of interneuron progenitors, while exogenous TGFβ3 accelerates the transition of progenitors into postmitotic neurons. We provide evidence that TGFb signaling exerts this function via regulating cell cycle length of the NKX2.1+ neural progenitors. Together, the present study represents a useful platform for studying human interneuron development and interneuron-associated neurological diseases with human pluripotent stem cells.
Collapse
|
25
|
Liu Y, Ouyang P, Zheng Y, Mi L, Zhao J, Ning Y, Guo W. A Selective Review of the Excitatory-Inhibitory Imbalance in Schizophrenia: Underlying Biology, Genetics, Microcircuits, and Symptoms. Front Cell Dev Biol 2021; 9:664535. [PMID: 34746116 PMCID: PMC8567014 DOI: 10.3389/fcell.2021.664535] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 09/27/2021] [Indexed: 12/29/2022] Open
Abstract
Schizophrenia is a chronic disorder characterized by specific positive and negative primary symptoms, social behavior disturbances and cognitive deficits (e.g., impairment in working memory and cognitive flexibility). Mounting evidence suggests that altered excitability and inhibition at the molecular, cellular, circuit and network level might be the basis for the pathophysiology of neurodevelopmental and neuropsychiatric disorders such as schizophrenia. In the past decades, human and animal studies have identified that glutamate and gamma-aminobutyric acid (GABA) neurotransmissions are critically involved in several cognitive progresses, including learning and memory. The purpose of this review is, by analyzing emerging findings relating to the balance of excitatory and inhibitory, ranging from animal models of schizophrenia to clinical studies in patients with early onset, first-episode or chronic schizophrenia, to discuss how the excitatory-inhibitory imbalance may relate to the pathophysiology of disease phenotypes such as cognitive deficits and negative symptoms, and highlight directions for appropriate therapeutic strategies.
Collapse
Affiliation(s)
- Yi Liu
- National Clinical Research Center on Mental Disorders and Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China.,Department of Psychiatry, The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China
| | - Pan Ouyang
- National Clinical Research Center on Mental Disorders and Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yingjun Zheng
- Department of Psychiatry, The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China
| | - Lin Mi
- Department of Psychiatry, The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jingping Zhao
- National Clinical Research Center on Mental Disorders and Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yuping Ning
- Department of Psychiatry, The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China.,The First School of Clinical Medical University, Guangzhou, China
| | - Wenbin Guo
- National Clinical Research Center on Mental Disorders and Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
26
|
Yang J, Huber L, Yu Y, Bandettini PA. Linking cortical circuit models to human cognition with laminar fMRI. Neurosci Biobehav Rev 2021; 128:467-478. [PMID: 34245758 DOI: 10.1016/j.neubiorev.2021.07.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 06/29/2021] [Accepted: 07/05/2021] [Indexed: 10/20/2022]
Abstract
Laboratory animal research has provided significant knowledge into the function of cortical circuits at the laminar level, which has yet to be fully leveraged towards insights about human brain function on a similar spatiotemporal scale. The use of functional magnetic resonance imaging (fMRI) in conjunction with neural models provides new opportunities to gain important insights from current knowledge. During the last five years, human studies have demonstrated the value of high-resolution fMRI to study laminar-specific activity in the human brain. This is mostly performed at ultra-high-field strengths (≥ 7 T) and is known as laminar fMRI. Advancements in laminar fMRI are beginning to open new possibilities for studying questions in basic cognitive neuroscience. In this paper, we first review recent methodological advances in laminar fMRI and describe recent human laminar fMRI studies. Then, we discuss how the use of laminar fMRI can help bridge the gap between cortical circuit models and human cognition.
Collapse
Affiliation(s)
- Jiajia Yang
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, Japan; Section on Functional Imaging Methods, National Institute of Mental Health, Bethesda, MD, USA.
| | - Laurentius Huber
- MR-Methods Group, Faculty of Psychology and Neuroscience, University of Maastricht, Maastricht, the Netherlands
| | - Yinghua Yu
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, Japan; Section on Functional Imaging Methods, National Institute of Mental Health, Bethesda, MD, USA
| | - Peter A Bandettini
- Section on Functional Imaging Methods, National Institute of Mental Health, Bethesda, MD, USA; Functional MRI Core Facility, National Institute of Mental Health, Bethesda, MD, USA
| |
Collapse
|
27
|
Siddiqi F, Trakimas AL, Joseph DJ, Lippincott ML, Marsh ED, Wolfe JH. Islet1 Precursors Contribute to Mature Interneuron Subtypes in Mouse Neocortex. Cereb Cortex 2021; 31:5206-5224. [PMID: 34228108 DOI: 10.1093/cercor/bhab152] [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: 01/16/2020] [Revised: 05/07/2021] [Accepted: 05/08/2021] [Indexed: 11/15/2022] Open
Abstract
Cortical interneurons (GABAergic cells) arise during embryogenesis primarily from the medial and caudal ganglionic eminences (MGE and CGE, respectively) with a small population generated from the preoptic area (POA). Progenitors from the lateral ganglionic eminence (LGE) are thought to only generate GABAergic medium spiny neurons that populate the striatum and project to the globus pallidus. Here, we report evidence that neuronal precursors that express the LGE-specific transcription factor Islet1 (Isl1) can give rise to a small population of cortical interneurons. Lineage tracing and homozygous deletion of Nkx2.1 in Isl1 fate-mapped mice showed that neighboring MGE/POA-specific Nkx2.1 cells and LGE-specific Isl1 cells make both common and distinct lineal contributions towards cortical interneuron fate. Although the majority of cells had overlapping transcriptional domains between Nkx2.1 and Isl1, a population of Isl1-only derived cells also contributed to the adult cerebral cortex. The data indicate that Isl1-derived cells may originate from both the LGE and the adjacent LGE/MGE boundary regions to generate diverse neuronal progeny. Thus, a small population of neocortical interneurons appear to originate from Isl-1-positive precursors.
Collapse
Affiliation(s)
- Faez Siddiqi
- Children's Hospital of Philadelphia Research Institute, Philadelphia, PA 19104, USA
| | - Alexandria L Trakimas
- Children's Hospital of Philadelphia Research Institute, Philadelphia, PA 19104, USA.,Departments of Neurology and Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Donald J Joseph
- Children's Hospital of Philadelphia Research Institute, Philadelphia, PA 19104, USA.,Division of Child Neurology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | | | - Eric D Marsh
- Children's Hospital of Philadelphia Research Institute, Philadelphia, PA 19104, USA.,Division of Child Neurology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.,Departments of Neurology and Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John H Wolfe
- Children's Hospital of Philadelphia Research Institute, Philadelphia, PA 19104, USA.,Division of Child Neurology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.,Departments of Neurology and Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,W.F. Goodman Center for Comparative Medical Genetics, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
28
|
Caffeine - treat or trigger? Disparate behavioral and long-term dopaminergic changes in control and schizophrenia-like Wisket rats. Physiol Behav 2021; 236:113410. [PMID: 33819453 DOI: 10.1016/j.physbeh.2021.113410] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/12/2021] [Accepted: 03/26/2021] [Indexed: 11/20/2022]
Abstract
The influence of caffeine on behavioral functions in both healthy and schizophrenic subjects is controversial. Here we aimed to reveal the effects of repeated caffeine pre- and post-training treatments on motor and exploratory activities and cognitive functions in a reward-based test (Ambitus) along with a brain region-specific dopamine D2 receptor profile in control and schizophrenia-like WISKET model rats. In the control animals, pre-treatment caused temporary enhancement in motor activity, while permanent improvement in learning function was detected in the WISKET animals. Post-treatment produced significant impairments in both groups. Caffeine caused short-lasting hyperactivity followed by a rebound in the inactive phase determined in undisturbed circumstance. Caffeine treatment substantially enhanced the dopamine D2 receptor mediated G-protein activation in the prefrontal cortex and olfactory bulb of both groups, while it increased in the dorsal striatum and cerebral cortex only in the WISKET animals. Caffeine enhanced the maximal binding capacity in the hippocampus and cerebral cortex of WISKET animals, but it decreased in the prefrontal cortex of the control animals. Regarding the dopamine D2 receptor mRNA expression, caffeine treatment caused significant enhancement in the prefrontal cortex of WISKET animals, while it increased the hippocampal dopamine D2 receptor protein amount in both groups. This study highlights the disparate effects of caffeine pre- versus post-training treatments on behavioral parameters in both control and schizophrenia-like animals and the prolonged changes in the dopaminergic system. It is supposed that the delayed depressive effects of caffeine might be compensated by frequent coffee intake, as observed in schizophrenic patients.
Collapse
|
29
|
Huang Y, Jiang H, Zheng Q, Fok AHK, Li X, Lau CG, Lai CSW. Environmental enrichment or selective activation of parvalbumin-expressing interneurons ameliorates synaptic and behavioral deficits in animal models with schizophrenia-like behaviors during adolescence. Mol Psychiatry 2021; 26:2533-2552. [PMID: 33473150 DOI: 10.1038/s41380-020-01005-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 12/18/2022]
Abstract
Synaptic deficit-induced excitation and inhibition (E/I) imbalance have been implicated in the pathogenesis of schizophrenia. Using in vivo two-photon microscopy, we examined the dynamic plasticity of dendritic spines of pyramidal neurons (PNs) and "en passant" axonal bouton of parvalbumin-expressing interneurons (PVINs) in the frontal association (FrA) cortex in two adolescent mouse models with schizophrenia-like behaviors. Simultaneous imaging of PN dendritic spines and PV axonal boutons showed that repeated exposure to N-methyl-D-aspartate receptor (NMDAR) antagonist MK801 during adolescence disrupted the normal developmental balance of excitatory and inhibitory synaptic structures. This MK801-induced structural E/I imbalance significantly correlated with animal recognition memory deficits and could be ameliorated by environmental enrichment (EE). In addition, selective chemogenetic activation of PVINs in the FrA mimicked the effects of EE on both synaptic plasticity and animal behavior, while selective inhibition of PVIN abolished EE's beneficial effects. Electrophysiological recordings showed that chronic MK801 treatment significantly suppressed the frequency of mEPSC/mIPSC ratio of layer (L) 2/3 PNs and significantly reduced the resting membrane potential of PVINs, the latter was rescued by selective activation of PVINs. Such manipulations of PVINs also showed similar effects in PV-Cre; ErbB4fl/fl animal model with schizophrenia-like behaviors. EE or selective activation of PVINs in the FrA restored behavioral deficits and structural E/I imbalance in adolescent PV-Cre; ErbB4fl/fl mice, while selective inhibition of PVINs abolished EE's beneficial effects. Our findings suggest that the PVIN activity in the FrA plays a crucial role in regulating excitatory and inhibitory synaptic structural dynamics and animal behaviors, which may provide a potential therapeutic target for schizophrenia treatment.
Collapse
Affiliation(s)
- Yuhua Huang
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Hehai Jiang
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon Tong, Hong Kong.,Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Qiyu Zheng
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Albert Hiu Ka Fok
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Xiaoyang Li
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - C Geoffrey Lau
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon Tong, Hong Kong.,Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Cora Sau Wan Lai
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong. .,State Key Laboratory of Cognitive and Brain Research, The University of Hong Kong, Pokfulam, Hong Kong.
| |
Collapse
|
30
|
Qin Z, Zhang L, Zasloff MA, Stewart AFR, Chen HH. Ketamine's schizophrenia-like effects are prevented by targeting PTP1B. Neurobiol Dis 2021; 155:105397. [PMID: 34015491 DOI: 10.1016/j.nbd.2021.105397] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 04/18/2021] [Accepted: 05/14/2021] [Indexed: 11/26/2022] Open
Abstract
Subanesthetic doses of ketamine induce schizophrenia-like behaviors in mice including hyperlocomotion and deficits in working memory and sensorimotor gating. Here, we examined the effect of in vivo ketamine administration on neuronal properties and endocannabinoid (eCB)-dependent modulation of synaptic transmission onto layer 2/3 pyramidal neurons in brain slices of the prefrontal cortex, a region tied to the schizophrenia-like behavioral phenotypes of ketamine. Since deficits in working memory and sensorimotor gating are tied to activation of the tyrosine phosphatase PTP1B in glutamatergic neurons, we asked whether PTP1B contributes to these effects of ketamine. Ketamine increased membrane resistance and excitability of pyramidal neurons. Systemic pharmacological inhibition of PTP1B by Trodusquemine restored these neuronal properties and prevented each of the three main ketamine-induced behavior deficits. Ketamine also reduced mobilization of eCB by pyramidal neurons, while unexpectedly reducing their inhibitory inputs, and these effects of ketamine were blocked or occluded by PTP1B ablation in glutamatergic neurons. While ablation of PTP1B in glutamatergic neurons prevented ketamine-induced deficits in memory and sensorimotor gating, it failed to prevent hyperlocomotion (a psychosis-like phenotype). Taken together, these results suggest that PTP1B in glutamatergic neurons mediates ketamine-induced deficits in eCB mobilization, memory and sensorimotor gating whereas PTP1B in other cell types contributes to hyperlocomotion. Our study suggests that the PTP1B inhibitor Trodusquemine may represent a new class of fast-acting antipsychotic drugs to treat schizophrenia-like symptoms.
Collapse
Affiliation(s)
- Zhaohong Qin
- Ottawa Hospital Research Institute, Ottawa, ON K1H8M5, Canada
| | - Li Zhang
- Ottawa Hospital Research Institute, Ottawa, ON K1H8M5, Canada
| | - Michael A Zasloff
- Georgetown University School of Medicine, MedStar Georgetown Transplant Institute, Washington D.C. 2007, USA
| | - Alexandre F R Stewart
- University of Ottawa Heart Institute, Ottawa, ON K1Y4W7, Canada; Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada.
| | - Hsiao-Huei Chen
- Ottawa Hospital Research Institute, Ottawa, ON K1H8M5, Canada; Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; Brain and Mind Institute, University of Ottawa, Ottawa, ON K1H 8M5, Canada.
| |
Collapse
|
31
|
Liu W, Wang J, Zhang H, Yu C, Liu S, Zhang C, Yu J, Liu Q, Yang B. Determining the effects of blended learning using the community of inquiry on nursing students' learning gains in sudden patient deterioration module. Nurs Open 2021; 8:3635-3644. [PMID: 33973718 PMCID: PMC8510760 DOI: 10.1002/nop2.914] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 04/08/2021] [Accepted: 04/14/2021] [Indexed: 02/06/2023] Open
Abstract
Aims To determine the effectiveness of blended learning using the community of inquiry framework on nursing students' learning gains in a sudden patient deterioration module. Design A quasi‐experimental trial. Methods 233 Chinese nursing students in their fourth semester of a sudden patient deterioration learning module were assigned to control (N = 113) and experimental group (N = 120). Students in experimental group engaged in community of inquiry‐based blended learning in sudden patient deterioration module, including computer‐aided self‐instruction, team‐based topic discussion and simulation training. Control group learned similar contents through face‐to‐face teaching comprising of a presentation with lecture, tutorial and simulation training. Student assessment of learning gains, knowledge and practical ability was quantified after the interventions. Results Compared with control group, students in experimental group had improved student assessment of learning gains (p = .001, Cohen d = 0.69) and practical ability (p < .001, Cohen d = 0.48). Although no significant difference in overall knowledge score, experimental group students did better performance in application and analysis (p = .001, Cohen d = 0.45).
Collapse
Affiliation(s)
- Weichu Liu
- Department of Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jun Wang
- Department of Nursing, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Han Zhang
- School of Nursing, Chongqing Medical University, Chongqing, China
| | - Changfeng Yu
- School of Nursing, Chongqing Medical University, Chongqing, China
| | - Shuai Liu
- School of Nursing, Chongqing Medical University, Chongqing, China
| | - Cen Zhang
- School of Nursing, Chongqing Medical University, Chongqing, China
| | - Jingya Yu
- School of Nursing, Chongqing Medical University, Chongqing, China
| | - Qiao Liu
- Department of Pediatric Dentistry, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Bing Yang
- Department of Nursing, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
32
|
Komatsu H, Watanabe E, Fukuchi M. Psychiatric Neural Networks and Precision Therapeutics by Machine Learning. Biomedicines 2021; 9:403. [PMID: 33917863 PMCID: PMC8068267 DOI: 10.3390/biomedicines9040403] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/28/2021] [Accepted: 04/06/2021] [Indexed: 12/12/2022] Open
Abstract
Learning and environmental adaptation increase the likelihood of survival and improve the quality of life. However, it is often difficult to judge optimal behaviors in real life due to highly complex social dynamics and environment. Consequentially, many different brain regions and neuronal circuits are involved in decision-making. Many neurobiological studies on decision-making show that behaviors are chosen through coordination among multiple neural network systems, each implementing a distinct set of computational algorithms. Although these processes are commonly abnormal in neurological and psychiatric disorders, the underlying causes remain incompletely elucidated. Machine learning approaches with multidimensional data sets have the potential to not only pathologically redefine mental illnesses but also better improve therapeutic outcomes than DSM/ICD diagnoses. Furthermore, measurable endophenotypes could allow for early disease detection, prognosis, and optimal treatment regime for individuals. In this review, decision-making in real life and psychiatric disorders and the applications of machine learning in brain imaging studies on psychiatric disorders are summarized, and considerations for the future clinical translation are outlined. This review also aims to introduce clinicians, scientists, and engineers to the opportunities and challenges in bringing artificial intelligence into psychiatric practice.
Collapse
Affiliation(s)
- Hidetoshi Komatsu
- Medical Affairs, Kyowa Pharmaceutical Industry Co., Ltd., Osaka 530-0005, Japan
- Department of Biological Science, Graduate School of Science, Nagoya University, Nagoya City 464-8602, Japan
| | - Emi Watanabe
- Interactive Group, Accenture Japan Ltd., Tokyo 108-0073, Japan;
| | - Mamoru Fukuchi
- Laboratory of Molecular Neuroscience, Faculty of Pharmacy, Takasaki University of Health and Welfare, Gunma 370-0033, Japan;
| |
Collapse
|
33
|
De la Torre GG, Doval S, López-Sanz D, García-Sedeño M, Ramallo MA, Bernal M, González-Torre S. Neurocognitive Impairment in Severe Mental Illness. Comparative study with Spanish Speaking Patients. Brain Sci 2021; 11:389. [PMID: 33808661 PMCID: PMC8003381 DOI: 10.3390/brainsci11030389] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/12/2021] [Accepted: 03/16/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Serious mental illness (SMI) represents a category of psychiatric disorders characterized by specific difficulties of personal and social functioning, derived from suffering severe and persistent mental health problems. AIMS We wanted to look into differences in cognitive performance among different SMI patients. METHODS Repeatable Battery for the Assessment of Neuropsychological Status (RBANS) screening was applied in one sample of SMI patients (n = 149) and another of healthy comparison participants (n = 35). Within the SMI sample, three different subsamples were formed: one with 97 patients with schizophrenia, a second with 29 patients with mood disorders, and a third with 23 patients with personality disorder. We performed a comparative study within and between groups. RESULTS Analysis of covariance was performed. Significant differences were found for cognitive functioning including attention and memory. CONCLUSIONS RBANS can be recommended for the detection of neurocognitive deficits in psychiatric disorders, especially in Schizophrenia.
Collapse
Affiliation(s)
- Gabriel G. De la Torre
- Neuropsychology and Experimental Psychology Lab, University of Cadiz, 11510 Puerto Real, Spain; (M.G.-S.); (M.A.R.); (S.G.-T.)
| | - Sandra Doval
- Center for Biomedical Technology, Technical University of Madrid, 28040 Madrid, Spain; (S.D.); (D.L.-S.)
- Department of Experimental Psychology, Complutense University of Madrid (UCM), 28223 Madrid, Spain
| | - David López-Sanz
- Center for Biomedical Technology, Technical University of Madrid, 28040 Madrid, Spain; (S.D.); (D.L.-S.)
- Department of Psychobiology and Methodology in Behavioral Sciences, Complutense University of Madrid (UCM), 28223 Madrid, Spain
| | - Manuel García-Sedeño
- Neuropsychology and Experimental Psychology Lab, University of Cadiz, 11510 Puerto Real, Spain; (M.G.-S.); (M.A.R.); (S.G.-T.)
| | - Miguel A. Ramallo
- Neuropsychology and Experimental Psychology Lab, University of Cadiz, 11510 Puerto Real, Spain; (M.G.-S.); (M.A.R.); (S.G.-T.)
| | | | - Sara González-Torre
- Neuropsychology and Experimental Psychology Lab, University of Cadiz, 11510 Puerto Real, Spain; (M.G.-S.); (M.A.R.); (S.G.-T.)
| |
Collapse
|
34
|
Edinoff AN, Wu NW, Maxey BS, Ren AL, Leethy KN, Girma B, Odisho A, Kaye JS, Kaye AJ, Kaye AM, Kaye AD, Mychaskiw G, Viswanath O, Urits I. Brexpiprazole for the Treatment of Schizophrenia and Major Depressive Disorder: A Comprehensive Review of Pharmacological Considerations in Clinical Practice. PSYCHOPHARMACOLOGY BULLETIN 2021; 51:69-95. [PMID: 34092824 PMCID: PMC8146559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Mood and psychotic disorders are a group of illnesses that affect behavior and cognition. Schizophrenia is characterized by positive symptoms, such as delusions and hallucinations, as well as negative symptoms. Major depressive disorder (MDD) is a mood disorder that affects the patient's emotions, energy, and motivation. Brexpiprazole works as a partial agonist at serotonin 5-hydroxytryptamine1A and dopamine D2 receptors and an antagonist at serotonin 5-hydroxytryptamine2A. Schizophrenia and MDD have a wide range of risk factors, both biological and environmental. Third generation antipsychotics, which include brexpiprazole, are the latest group of drugs to reach the market, demonstrating efficacy and tolerability. Patients with acute schizophrenia have responded well to brexpiprazole. In this regard, in patients who have MDD plus anxiety symptoms, brexpiprazole can be effective as an adjunctive therapy and can reduce anxiety symptoms. In summary, brexpiprazole has proved to be an effective alternative to typical or first and second-generation atypical antipsychotics.
Collapse
Affiliation(s)
- Amber N Edinoff
- Edinoff, MD, Wu, MD, Odisho, MD, Louisiana State University Health Science Center Shreveport, Department of Psychiatry and Behavioral Medicine. Maxey, BS, Ren, BS, Leethy, BS, Louisiana State University Shreveport School of Medicine. Girma, MD, Alan D. Kaye, MD, PhD, Mychaskiw, DO, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA. Jessica S. Kaye, Adam M. Kaye, PharmD, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Department of Pharmacy Practice, Stockton, CA. Aaron J. Kaye, MD, Medical University of South Carolina, Department of Anesthesiology, Charleston, SC. Viswanath, MD, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE, Valley Anesthesiology and Pain Consultants-Envision Physician Services, Phoenix, AZ. Urits, MD, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA, Southcoast Health, Southcoast Physicians Group Pain Medicine, Wareham, MA
| | - Natalie W Wu
- Edinoff, MD, Wu, MD, Odisho, MD, Louisiana State University Health Science Center Shreveport, Department of Psychiatry and Behavioral Medicine. Maxey, BS, Ren, BS, Leethy, BS, Louisiana State University Shreveport School of Medicine. Girma, MD, Alan D. Kaye, MD, PhD, Mychaskiw, DO, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA. Jessica S. Kaye, Adam M. Kaye, PharmD, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Department of Pharmacy Practice, Stockton, CA. Aaron J. Kaye, MD, Medical University of South Carolina, Department of Anesthesiology, Charleston, SC. Viswanath, MD, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE, Valley Anesthesiology and Pain Consultants-Envision Physician Services, Phoenix, AZ. Urits, MD, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA, Southcoast Health, Southcoast Physicians Group Pain Medicine, Wareham, MA
| | - Benjamin S Maxey
- Edinoff, MD, Wu, MD, Odisho, MD, Louisiana State University Health Science Center Shreveport, Department of Psychiatry and Behavioral Medicine. Maxey, BS, Ren, BS, Leethy, BS, Louisiana State University Shreveport School of Medicine. Girma, MD, Alan D. Kaye, MD, PhD, Mychaskiw, DO, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA. Jessica S. Kaye, Adam M. Kaye, PharmD, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Department of Pharmacy Practice, Stockton, CA. Aaron J. Kaye, MD, Medical University of South Carolina, Department of Anesthesiology, Charleston, SC. Viswanath, MD, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE, Valley Anesthesiology and Pain Consultants-Envision Physician Services, Phoenix, AZ. Urits, MD, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA, Southcoast Health, Southcoast Physicians Group Pain Medicine, Wareham, MA
| | - Amy L Ren
- Edinoff, MD, Wu, MD, Odisho, MD, Louisiana State University Health Science Center Shreveport, Department of Psychiatry and Behavioral Medicine. Maxey, BS, Ren, BS, Leethy, BS, Louisiana State University Shreveport School of Medicine. Girma, MD, Alan D. Kaye, MD, PhD, Mychaskiw, DO, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA. Jessica S. Kaye, Adam M. Kaye, PharmD, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Department of Pharmacy Practice, Stockton, CA. Aaron J. Kaye, MD, Medical University of South Carolina, Department of Anesthesiology, Charleston, SC. Viswanath, MD, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE, Valley Anesthesiology and Pain Consultants-Envision Physician Services, Phoenix, AZ. Urits, MD, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA, Southcoast Health, Southcoast Physicians Group Pain Medicine, Wareham, MA
| | - Kenna N Leethy
- Edinoff, MD, Wu, MD, Odisho, MD, Louisiana State University Health Science Center Shreveport, Department of Psychiatry and Behavioral Medicine. Maxey, BS, Ren, BS, Leethy, BS, Louisiana State University Shreveport School of Medicine. Girma, MD, Alan D. Kaye, MD, PhD, Mychaskiw, DO, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA. Jessica S. Kaye, Adam M. Kaye, PharmD, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Department of Pharmacy Practice, Stockton, CA. Aaron J. Kaye, MD, Medical University of South Carolina, Department of Anesthesiology, Charleston, SC. Viswanath, MD, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE, Valley Anesthesiology and Pain Consultants-Envision Physician Services, Phoenix, AZ. Urits, MD, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA, Southcoast Health, Southcoast Physicians Group Pain Medicine, Wareham, MA
| | - Brook Girma
- Edinoff, MD, Wu, MD, Odisho, MD, Louisiana State University Health Science Center Shreveport, Department of Psychiatry and Behavioral Medicine. Maxey, BS, Ren, BS, Leethy, BS, Louisiana State University Shreveport School of Medicine. Girma, MD, Alan D. Kaye, MD, PhD, Mychaskiw, DO, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA. Jessica S. Kaye, Adam M. Kaye, PharmD, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Department of Pharmacy Practice, Stockton, CA. Aaron J. Kaye, MD, Medical University of South Carolina, Department of Anesthesiology, Charleston, SC. Viswanath, MD, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE, Valley Anesthesiology and Pain Consultants-Envision Physician Services, Phoenix, AZ. Urits, MD, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA, Southcoast Health, Southcoast Physicians Group Pain Medicine, Wareham, MA
| | - Amira Odisho
- Edinoff, MD, Wu, MD, Odisho, MD, Louisiana State University Health Science Center Shreveport, Department of Psychiatry and Behavioral Medicine. Maxey, BS, Ren, BS, Leethy, BS, Louisiana State University Shreveport School of Medicine. Girma, MD, Alan D. Kaye, MD, PhD, Mychaskiw, DO, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA. Jessica S. Kaye, Adam M. Kaye, PharmD, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Department of Pharmacy Practice, Stockton, CA. Aaron J. Kaye, MD, Medical University of South Carolina, Department of Anesthesiology, Charleston, SC. Viswanath, MD, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE, Valley Anesthesiology and Pain Consultants-Envision Physician Services, Phoenix, AZ. Urits, MD, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA, Southcoast Health, Southcoast Physicians Group Pain Medicine, Wareham, MA
| | - Jessica S Kaye
- Edinoff, MD, Wu, MD, Odisho, MD, Louisiana State University Health Science Center Shreveport, Department of Psychiatry and Behavioral Medicine. Maxey, BS, Ren, BS, Leethy, BS, Louisiana State University Shreveport School of Medicine. Girma, MD, Alan D. Kaye, MD, PhD, Mychaskiw, DO, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA. Jessica S. Kaye, Adam M. Kaye, PharmD, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Department of Pharmacy Practice, Stockton, CA. Aaron J. Kaye, MD, Medical University of South Carolina, Department of Anesthesiology, Charleston, SC. Viswanath, MD, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE, Valley Anesthesiology and Pain Consultants-Envision Physician Services, Phoenix, AZ. Urits, MD, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA, Southcoast Health, Southcoast Physicians Group Pain Medicine, Wareham, MA
| | - Aaron J Kaye
- Edinoff, MD, Wu, MD, Odisho, MD, Louisiana State University Health Science Center Shreveport, Department of Psychiatry and Behavioral Medicine. Maxey, BS, Ren, BS, Leethy, BS, Louisiana State University Shreveport School of Medicine. Girma, MD, Alan D. Kaye, MD, PhD, Mychaskiw, DO, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA. Jessica S. Kaye, Adam M. Kaye, PharmD, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Department of Pharmacy Practice, Stockton, CA. Aaron J. Kaye, MD, Medical University of South Carolina, Department of Anesthesiology, Charleston, SC. Viswanath, MD, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE, Valley Anesthesiology and Pain Consultants-Envision Physician Services, Phoenix, AZ. Urits, MD, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA, Southcoast Health, Southcoast Physicians Group Pain Medicine, Wareham, MA
| | - Adam M Kaye
- Edinoff, MD, Wu, MD, Odisho, MD, Louisiana State University Health Science Center Shreveport, Department of Psychiatry and Behavioral Medicine. Maxey, BS, Ren, BS, Leethy, BS, Louisiana State University Shreveport School of Medicine. Girma, MD, Alan D. Kaye, MD, PhD, Mychaskiw, DO, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA. Jessica S. Kaye, Adam M. Kaye, PharmD, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Department of Pharmacy Practice, Stockton, CA. Aaron J. Kaye, MD, Medical University of South Carolina, Department of Anesthesiology, Charleston, SC. Viswanath, MD, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE, Valley Anesthesiology and Pain Consultants-Envision Physician Services, Phoenix, AZ. Urits, MD, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA, Southcoast Health, Southcoast Physicians Group Pain Medicine, Wareham, MA
| | - Alan D Kaye
- Edinoff, MD, Wu, MD, Odisho, MD, Louisiana State University Health Science Center Shreveport, Department of Psychiatry and Behavioral Medicine. Maxey, BS, Ren, BS, Leethy, BS, Louisiana State University Shreveport School of Medicine. Girma, MD, Alan D. Kaye, MD, PhD, Mychaskiw, DO, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA. Jessica S. Kaye, Adam M. Kaye, PharmD, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Department of Pharmacy Practice, Stockton, CA. Aaron J. Kaye, MD, Medical University of South Carolina, Department of Anesthesiology, Charleston, SC. Viswanath, MD, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE, Valley Anesthesiology and Pain Consultants-Envision Physician Services, Phoenix, AZ. Urits, MD, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA, Southcoast Health, Southcoast Physicians Group Pain Medicine, Wareham, MA
| | - George Mychaskiw
- Edinoff, MD, Wu, MD, Odisho, MD, Louisiana State University Health Science Center Shreveport, Department of Psychiatry and Behavioral Medicine. Maxey, BS, Ren, BS, Leethy, BS, Louisiana State University Shreveport School of Medicine. Girma, MD, Alan D. Kaye, MD, PhD, Mychaskiw, DO, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA. Jessica S. Kaye, Adam M. Kaye, PharmD, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Department of Pharmacy Practice, Stockton, CA. Aaron J. Kaye, MD, Medical University of South Carolina, Department of Anesthesiology, Charleston, SC. Viswanath, MD, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE, Valley Anesthesiology and Pain Consultants-Envision Physician Services, Phoenix, AZ. Urits, MD, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA, Southcoast Health, Southcoast Physicians Group Pain Medicine, Wareham, MA
| | - Omar Viswanath
- Edinoff, MD, Wu, MD, Odisho, MD, Louisiana State University Health Science Center Shreveport, Department of Psychiatry and Behavioral Medicine. Maxey, BS, Ren, BS, Leethy, BS, Louisiana State University Shreveport School of Medicine. Girma, MD, Alan D. Kaye, MD, PhD, Mychaskiw, DO, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA. Jessica S. Kaye, Adam M. Kaye, PharmD, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Department of Pharmacy Practice, Stockton, CA. Aaron J. Kaye, MD, Medical University of South Carolina, Department of Anesthesiology, Charleston, SC. Viswanath, MD, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE, Valley Anesthesiology and Pain Consultants-Envision Physician Services, Phoenix, AZ. Urits, MD, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA, Southcoast Health, Southcoast Physicians Group Pain Medicine, Wareham, MA
| | - Ivan Urits
- Edinoff, MD, Wu, MD, Odisho, MD, Louisiana State University Health Science Center Shreveport, Department of Psychiatry and Behavioral Medicine. Maxey, BS, Ren, BS, Leethy, BS, Louisiana State University Shreveport School of Medicine. Girma, MD, Alan D. Kaye, MD, PhD, Mychaskiw, DO, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA. Jessica S. Kaye, Adam M. Kaye, PharmD, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Department of Pharmacy Practice, Stockton, CA. Aaron J. Kaye, MD, Medical University of South Carolina, Department of Anesthesiology, Charleston, SC. Viswanath, MD, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE, Valley Anesthesiology and Pain Consultants-Envision Physician Services, Phoenix, AZ. Urits, MD, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA, Southcoast Health, Southcoast Physicians Group Pain Medicine, Wareham, MA
| |
Collapse
|
35
|
Chini M, Hanganu-Opatz IL. Prefrontal Cortex Development in Health and Disease: Lessons from Rodents and Humans. Trends Neurosci 2020; 44:227-240. [PMID: 33246578 DOI: 10.1016/j.tins.2020.10.017] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/15/2020] [Accepted: 10/29/2020] [Indexed: 12/22/2022]
Abstract
The role of the prefrontal cortex (PFC) takes center stage among unanswered questions in modern neuroscience. The PFC has a Janus-faced nature: it enables sophisticated cognitive and social abilities that reach their maximum expression in humans, yet it underlies some of the devastating symptoms of psychiatric disorders. Accordingly, appropriate prefrontal development is crucial for many high-order cognitive abilities and dysregulation of this process has been linked to various neuropsychiatric diseases. Reviewing recent advances in the field, with a primary focus on rodents and humans, we highlight why, despite differences across species, a cross-species approach is a fruitful strategy for understanding prefrontal development. We briefly review the developmental contribution of molecules and extensively discuss how electrical activity controls the early maturation and wiring of prefrontal areas, as well as the emergence and refinement of input-output circuitry involved in cognitive processing. Finally, we highlight the mechanisms of developmental dysfunction and their relevance for psychiatric disorders.
Collapse
Affiliation(s)
- Mattia Chini
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Ileana L Hanganu-Opatz
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| |
Collapse
|
36
|
Mewton L, Hodge A, Gates N, Visontay R, Lees B, Teesson M. A randomised double-blind trial of cognitive training for the prevention of psychopathology in at-risk youth. Behav Res Ther 2020; 132:103672. [PMID: 32629291 DOI: 10.1016/j.brat.2020.103672] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 05/21/2020] [Accepted: 06/04/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND The aim of this study was to evaluate the effectiveness of online cognitive training as a means of reducing psychopathology in at-risk youth. METHODS In a double-blind randomised controlled trial, 228 youths (mean age = 18.6, 74.6% female) were randomly allocated to either an intervention group (n = 114; online cognitive training focused on executive functioning) and a control group (n = 114; online cognitive training focused on other cognitive abilities). Participants were assessed online at baseline, post-training, 3-, 6- and 12-month follow-up. The primary outcome of the study was overall psychopathology as measured by the Strengths and Difficulties Questionnaire. Secondary outcomes were executive functioning ability (assessed using the n-back, trail-making and Stroop tasks), day-to-day functioning and risky drinking. RESULTS Mixed model intention-to-treat analyses indicated that psychopathology increased and day-to-day functioning decreased, regardless of intervention group. Those in the intervention group improved more than those in the control group in terms of the n-back task, but this was not statistically significant after adjusting for multiple comparisons. There were no statistically significant effects on risky drinking, or the trail-making and Stroop tasks. CONCLUSION This study failed to provide evidence for the efficacy of cognitive training as a stand-alone intervention for psychopathology.
Collapse
Affiliation(s)
- Louise Mewton
- Centre for Healthy Brain Ageing, University of New South Wales, Sydney, Australia.
| | - Antoinette Hodge
- Child Development Unit, The Children's Hospital, Westmead, Australia
| | - Nicola Gates
- Centre for Healthy Brain Ageing, University of New South Wales, Sydney, Australia
| | - Rachel Visontay
- The Matilda Centre for Research in Mental Health and Substance Use, University of Sydney, Sydney, Australia
| | - Briana Lees
- The Matilda Centre for Research in Mental Health and Substance Use, University of Sydney, Sydney, Australia
| | - Maree Teesson
- The Matilda Centre for Research in Mental Health and Substance Use, University of Sydney, Sydney, Australia
| |
Collapse
|
37
|
Al-Absi AR, Qvist P, Okujeni S, Khan AR, Glerup S, Sanchez C, Nyengaard JR. Layers II/III of Prefrontal Cortex in Df(h22q11)/+ Mouse Model of the 22q11.2 Deletion Display Loss of Parvalbumin Interneurons and Modulation of Neuronal Morphology and Excitability. Mol Neurobiol 2020; 57:4978-4988. [PMID: 32820460 DOI: 10.1007/s12035-020-02067-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 08/09/2020] [Indexed: 11/26/2022]
Abstract
The 22q11.2 deletion has been identified as a risk factor for multiple neurodevelopmental disorders. Behavioral and cognitive impairments are common among carriers of the 22q11.2 deletion. Parvalbumin expressing (PV+) interneurons provide perisomatic inhibition of excitatory neuronal circuits through GABAA receptors, and a deficit of PV+ inhibitory circuits may underlie a multitude of the behavioral and functional deficits in the 22q11.2 deletion syndrome. We investigated putative deficits of PV+ inhibitory circuits and the associated molecular, morphological, and functional alterations in the prefrontal cortex (PFC) of the Df(h22q11)/+ mouse model of the 22q11.2 hemizygous deletion. We detected a significant decrease in the number of PV+ interneurons in layers II/III of PFC in Df(h22q11)/+ mice together with a reduction in the mRNA and protein levels of GABAA (α3), a PV+ putative postsynaptic receptor subunit. Pyramidal neurons from the same layers further experienced morphological reorganizations of spines and dendrites. Accordingly, a decrease in the levels of the postsynaptic density protein 95 (PSD95) and a higher neuronal activity in response to the GABAA antagonist bicuculline were measured in these layers in PFC of Df(h22q11)/+ mice compared with their wild-type littermates. Our study shows that a hemizygotic deletion of the 22q11.2 locus leads to deficit in the GABAergic control of network activity and involves molecular and morphological changes in both the inhibitory and excitatory synapses of parvalbumin interneurons and pyramidal neurons specifically in layers II/III PFC.
Collapse
Affiliation(s)
- Abdel-Rahman Al-Absi
- Centre for Molecular Morphology, Section for Stereology and Microscopy; Centre for Stochastic Geometry and Advanced Bioimaging, Department of Clinical Medicine, Aarhus University, Palle Juul Jensens Boulevard, 99 8200, Aarhus N, Denmark.
| | - Per Qvist
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark
- Centre for Genomics and Personalized Medicine, CGPM, Aarhus University, Aarhus, Denmark
| | - Samora Okujeni
- Laboratory for Biomicrotechnology, Department of Microsystems Engineering IMTEK, University of Freiburg, Freiburg, Germany
| | - Ahmad Raza Khan
- Center of Functionally Integrative Neuroscience (CFIN), Aarhus University, Aarhus, Denmark
- Centre of Biomedical Research (CBMR), SGPGIMS Campus, Lucknow, India
| | - Simon Glerup
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Connie Sanchez
- Translational Neuropsychiatry Unit, Aarhus University, Aarhus, Denmark
| | - Jens R Nyengaard
- Centre for Molecular Morphology, Section for Stereology and Microscopy; Centre for Stochastic Geometry and Advanced Bioimaging, Department of Clinical Medicine, Aarhus University, Palle Juul Jensens Boulevard, 99 8200, Aarhus N, Denmark
| |
Collapse
|
38
|
Yang J, Pu W, Wu G, Chen E, Lee E, Liu Z, Palaniyappan L. Connectomic Underpinnings of Working Memory Deficits in Schizophrenia: Evidence From a replication fMRI study. Schizophr Bull 2020; 46:916-926. [PMID: 32016430 PMCID: PMC7345823 DOI: 10.1093/schbul/sbz137] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Working memory (WM) deficit is a key feature of schizophrenia that relates to a generalized neural inefficiency of extensive brain areas. To date, it remains unknown how these distributed regions are systemically organized at the connectome level and how the disruption of such organization brings about the WM impairment seen in schizophrenia. METHODS We used graph theory to examine the neural efficiency of the functional connectome in different granularity in 155 patients with schizophrenia and 96 healthy controls during a WM task. These analyses were repeated in another independent dataset (81 patients and 54 controls). Linear regression analysis was used to test associations of altered graph properties, clinical symptoms, and WM accuracy in patients. A machine-learning approach was adopted to study the ability of multivariate connectome features from one dataset to discriminate patients from controls in the second dataset. RESULTS Small-worldness of the whole-brain connectome was significantly increased in schizophrenia during the WM task; this increase is related to better (though subpar) WM accuracy in patients with more severe negative symptom burden. There was a shift in the degree distribution to a more homogeneous form in patients. The machine-learning approach classified a new set of patients from controls with 84.3% true-positivity rate for schizophrenia and 71.6% overall accuracy. CONCLUSIONS We demonstrate a putative mechanistic link between connectome topology, hub redistribution, and impaired n-back performance in schizophrenia. The task-dependent modulation of the connectome relates to, but remains inefficient in, improving the performance above par in the presence of severe negative symptoms.
Collapse
Affiliation(s)
- Jie Yang
- Institute of Mental Health, Second Xiangya Hospital, Central South University, Changsha, PR China
| | - Weidan Pu
- Medical Psychological Center, the Second Xiangya Hospital, Central South University, Changsha, P.R. China
- Medical Psychological Institute of Central South University, Changsha, P.R. China
| | - Guowei Wu
- Institute of Mental Health, Second Xiangya Hospital, Central South University, Changsha, PR China
| | - Eric Chen
- Department of Psychiatry, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Edwin Lee
- Department of Psychiatry, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Zhening Liu
- Institute of Mental Health, Second Xiangya Hospital, Central South University, Changsha, PR China
| | - Lena Palaniyappan
- Institute of Mental Health, Second Xiangya Hospital, Central South University, Changsha, PR China
- Department of Psychiatry, University of Western Ontario, London, ON, Canada
- Robarts Research Institute, University of Western Ontario, London, ON, Canada
- Lawson Health Research Institute, London, ON, Canada
| |
Collapse
|
39
|
Robison A, Thakkar K, Diwadkar VA. Cognition and Reward Circuits in Schizophrenia: Synergistic, Not Separate. Biol Psychiatry 2020; 87:204-214. [PMID: 31733788 PMCID: PMC6946864 DOI: 10.1016/j.biopsych.2019.09.021] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 09/05/2019] [Accepted: 09/17/2019] [Indexed: 01/29/2023]
Abstract
Schizophrenia has been studied from the perspective of cognitive or reward-related impairments, yet it cannot be wholly related to one or the other process and their corresponding neural circuits. We posit a comprehensive circuit-based model proposing that dysfunctional interactions between the brain's cognitive and reward circuits underlie schizophrenia. The model is underpinned by how the relationship between glutamatergic and dopaminergic dysfunction in schizophrenia drives interactions between cognition and reward circuits. We argue that this interaction is synergistic: that is, deficits of cognition and reward processing interact, and this interaction is a core feature of schizophrenia. In adopting this position, we undertake a focused review of animal physiology and human clinical data, and in proposing this synergistic model, we highlight dopaminergic afferents from the ventral tegmental area to nucleus accumbens (mesolimbic circuit) and frontal cortex (mesocortical circuit). We then expand on the role of glutamatergic inputs to these dopamine circuits and dopaminergic modulation of critical excitatory pathways with attention given to the role of glutamatergic hippocampal outputs onto nucleus accumbens. Finally, we present evidence for how in schizophrenia, dysfunction in the mesolimbic and mesocortical circuits and their corresponding glutamatergic inputs gives rise to clinical and cognitive phenotypes and is associated with positive and negative symptom dimensions. The synthesis attempted here provides an impetus for a conceptual shift that links cognitive and motivational aspects of schizophrenia and that can lead to treatment approaches that seek to harmonize network interactions between the brain's cognition and reward circuits with ameliorative effects in each behavioral domain.
Collapse
Affiliation(s)
| | - Katharine Thakkar
- Dept. of Psychology, Michigan State University,Division of Psychiatry and Behavioral Medicine, Michigan State University
| | | |
Collapse
|
40
|
Takahashi S, Keeser D, Rauchmann BS, Schneider-Axmann T, Keller-Varady K, Maurus I, Dechent P, Wobrock T, Hasan A, Schmitt A, Ertl-Wagner B, Malchow B, Falkai P. Effect of aerobic exercise combined with cognitive remediation on cortical thickness and prediction of social adaptation in patients with schizophrenia. Schizophr Res 2020; 216:397-407. [PMID: 31806522 DOI: 10.1016/j.schres.2019.11.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 07/29/2019] [Accepted: 11/03/2019] [Indexed: 01/09/2023]
Abstract
Aerobic exercise is a promising intervention for patients with schizophrenia, but structural neuroplastic effects on brain regions relevant to the pathophysiology of the disease remain unclear. This study aimed to elucidate longitudinal changes in cortical thickness after aerobic exercise intervention in schizophrenia patients and the relationship of these changes to clinical correlates. We investigated 21 schizophrenia patients and 23 healthy controls who performed aerobic exercise and 21 schizophrenia patients who played table soccer. The 12-week exercise intervention was combined with computer-assisted cognitive remediation training from week 6 to week 12. Magnetic resonance imaging (MRI) scans were acquired at baseline and weeks 6, 12, and 24. The thickness of the entorhinal, parahippocampal, and lateral and medial prefrontal cortices was assessed with FreeSurfer 6.0. The schizophrenia aerobic exercise group showed a significant increase of cortical thickness in the right entorhinal cortex at week 6, and we found a significant correlation between the cortical thickness of the right lateral prefrontal cortex at baseline and improvement of social adaptation at week 12. In the schizophrenia table soccer and healthy control groups, we found no significant longitudinal change in cortical thickness through the intervention and follow-up period and no correlation of cortical thickness at baseline with clinical measures. Our results suggest that aerobic exercise in schizophrenia modulates the thickness of the entorhinal cortex, a structure adjacent to the hippocampus. Greater cortical thickness of the right lateral prefrontal cortex appears to predict better clinical response to an aerobic exercise intervention in patients with schizophrenia.
Collapse
Affiliation(s)
- Shun Takahashi
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Nussbaumstraße 7, 80336, Munich, Germany; Department of Neuropsychiatry, Wakayama Medical University, 811-1 Kimiidera, 6410012, Wakayama, Japan.
| | - Daniel Keeser
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Nussbaumstraße 7, 80336, Munich, Germany; Department of Radiology, University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany
| | - Boris-Stephan Rauchmann
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Nussbaumstraße 7, 80336, Munich, Germany; Department of Radiology, University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany
| | - Thomas Schneider-Axmann
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Nussbaumstraße 7, 80336, Munich, Germany
| | - Katriona Keller-Varady
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Nussbaumstraße 7, 80336, Munich, Germany
| | - Isabel Maurus
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Nussbaumstraße 7, 80336, Munich, Germany
| | - Peter Dechent
- Institute for Cognitive Neurology, University Medical Center Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
| | - Thomas Wobrock
- Department of Psychiatry and Psychotherapy, Georg-August-University, Von-Siebold-Str. 5, 37075, Göttingen, Germany; Centre of Mental Health, County Hospitals Darmstadt-Dieburg, Krankenhausstraße 7, 64823, Groß-Umstadt, Germany
| | - Alkomiet Hasan
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Nussbaumstraße 7, 80336, Munich, Germany
| | - Andrea Schmitt
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Nussbaumstraße 7, 80336, Munich, Germany; Laboratory of Neuroscience (LIM27), Institute of Psychiatry, University of Sao Paulo, 05403-010, São Paulo, Brazil
| | - Birgit Ertl-Wagner
- Department of Radiology, University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany; Department of Medical Imaging, University of Toronto, McCaul Street 263, Toronto, M5T1W7, Ontario, Canada
| | - Berend Malchow
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Nussbaumstraße 7, 80336, Munich, Germany; Department of Psychiatry and Psychotherapy, University of Jena, Philosophenweg 3, 07743, Jena, Germany
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Nussbaumstraße 7, 80336, Munich, Germany
| |
Collapse
|
41
|
Cholecystokinin-Expressing Interneurons of the Medial Prefrontal Cortex Mediate Working Memory Retrieval. J Neurosci 2020; 40:2314-2331. [PMID: 32005764 DOI: 10.1523/jneurosci.1919-19.2020] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 01/20/2020] [Accepted: 01/23/2020] [Indexed: 12/14/2022] Open
Abstract
Distinct components of working memory are coordinated by different classes of inhibitory interneurons in the PFC, but the role of cholecystokinin (CCK)-positive interneurons remains enigmatic. In humans, this major population of interneurons shows histological abnormalities in schizophrenia, an illness in which deficient working memory is a core defining symptom and the best predictor of long-term functional outcome. Yet, CCK interneurons as a molecularly distinct class have proved intractable to examination by typical molecular methods due to widespread expression of CCK in the pyramidal neuron population. Using an intersectional approach in mice of both sexes, we have succeeded in labeling, interrogating, and manipulating CCK interneurons in the mPFC. Here, we describe the anatomical distribution, electrophysiological properties, and postsynaptic connectivity of CCK interneurons, and evaluate their role in cognition. We found that CCK interneurons comprise a larger proportion of the mPFC interneurons compared with parvalbumin interneurons, targeting a wide range of neuronal subtypes with a distinct connectivity pattern. Phase-specific optogenetic inhibition revealed that CCK, but not parvalbumin, interneurons play a critical role in the retrieval of working memory. These findings shine new light on the relationship between cortical CCK interneurons and cognition and offer a new set of tools to investigate interneuron dysfunction and cognitive impairments associated with schizophrenia.SIGNIFICANCE STATEMENT Cholecystokinin-expressing interneurons outnumber other interneuron populations in key brain areas involved in cognition and memory, including the mPFC. However, they have proved intractable to examination as experimental techniques have lacked the necessary selectivity. To the best of our knowledge, the present study is the first to report detailed properties of cortical cholecystokinin interneurons, revealing their anatomical organization, electrophysiological properties, postsynaptic connectivity, and behavioral function in working memory.
Collapse
|
42
|
Disruption of gamma-delta relationship related to working memory deficits in first-episode psychosis. J Neural Transm (Vienna) 2019; 127:103-115. [PMID: 31858267 DOI: 10.1007/s00702-019-02126-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 12/14/2019] [Indexed: 12/22/2022]
Abstract
Working memory (WM) deficits constitute a core symptom of schizophrenia. Inadequacy of WM maintenance in schizophrenia has been reported to reflect abnormalities in the excitation/inhibition (E/I) balance between pyramidal neurons and parvalbumin basket cells, which may explain alterations of the dynamics of gamma and delta oscillations. To address this issue, we assessed event-related gamma (35-45 Hz) and delta (0.5-4 Hz) oscillatory responses in a visual n-back WM task in patients with first-episode psychosis (FEP) and healthy controls (HC). Periodicity analyses of oscillations were computed to explore the relationship between the psychiatric status and the WM load-related processes reflected by each frequency range. The correspondence between nested delta-gamma oscillations was estimated to assess the strength of the frontal E/I balance. In HC, gamma oscillations were synchronized by the stimulus in a 50-150 ms time range for all tasks, and periodicity of the delta cycle was comparable between the tasks. In addition, synchronization of gamma oscillations in HC occurred at the maximal descending phase of the delta cycle half-period, supporting the coexistence of delta-nested gamma oscillations. Compared with controls, FEP patients showed a lack of gamma synchronization independently of the nature of the task, and the period of delta oscillation increased significantly with the difficulty of the WM task. We thus demonstrated in FEP an inability to encode multiple items in short-term memory associated with abnormalities in the relationship between oscillations related to the difficulty of the WM task. These results argue in favor of a dysfunction of the E/I balance in psychosis.
Collapse
|
43
|
Disrupted GABAergic facilitation of working memory performance in people with schizophrenia. NEUROIMAGE-CLINICAL 2019; 25:102127. [PMID: 31864216 PMCID: PMC6928454 DOI: 10.1016/j.nicl.2019.102127] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/25/2019] [Accepted: 12/13/2019] [Indexed: 11/21/2022]
Abstract
As in a previous study, higher GABA concentrations in the dorsolateral prefrontal cortex (DLPFC) were associated with better working memory (WM) in healthy participants. Despite no overall group difference in DLPFC GABA concentrations, people with schizophrenia showed significantly different inverse associations, with higher DLPFC GABA associated with worse rather than better WM. This opposite pattern of correlations despite a lack of group differences suggests that schizophrenia alters the distribution of different classes of GABAergic interneurons rather than producing a general deficit across the total population of neurons.
Objectives Gamma-Amiobutyric acid (GABA) is a primary inhibitory neurotransmitter that facilitates neural oscillations that coordinate neural activity between brain networks to facilitate cognition. The present magnetic resonance spectroscopy (MRS) study tests the hypothesis that GABAergic facilitation of working memory is disrupted in people with schizophrenia (PSZ). Methods 51 healthy participants and 40 PSZ from the UC Davis Early Psychosis Program performed an item and temporal order working memory (WM) task and underwent resting MRS to measure GABA and glutamate concentrations in dorsolateral prefrontal (DLPFC) and anterior cingulate (ACC) regions of interest. MRS was acquired on a 3 Tesla Siemens scanner and GABA and glutamate concentrations were referenced to creatine. Percent correct on the WM task indexed performance and correlation coefficients examined GABAergic or Glutamatergic facilitation of WM, with Fisher's Z transformation testing for group differences. Results There were no group differences in GABA or glutamate concentrations, but WM correlations were reversed between groups. In patients, higher DLPFC GABA was associated with worse rather than better WM performance. This pattern was not observed for glutamate or in the ACC. Although under-powered, there was no indication of medication effects. Conclusions and Relevance Results cannot be explained by group differences in DLPFC GABA or glutamate concentrations but, instead, indicate that schizophrenia disrupts the GABAergic facilitation of WM seen in healthy individuals. Results appear to parallel post mortem findings in suggesting that schizophrenia alters the distribution of different classes of GABAergic interneurons rather than producing a general deficit across the total population of neurons.
Collapse
|
44
|
Li J, Ryan SK, Deboer E, Cook K, Fitzgerald S, Lachman HM, Wallace DC, Goldberg EM, Anderson SA. Mitochondrial deficits in human iPSC-derived neurons from patients with 22q11.2 deletion syndrome and schizophrenia. Transl Psychiatry 2019; 9:302. [PMID: 31740674 PMCID: PMC6861238 DOI: 10.1038/s41398-019-0643-y] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 08/11/2019] [Indexed: 12/23/2022] Open
Abstract
Schizophrenia (SZ) is a highly heterogeneous disorder in both its symptoms and risk factors. One of the most prevalent genetic risk factors for SZ is the hemizygous microdeletion at chromosome 22q11.2 (22q11DS) that confers a 25-fold increased risk. Six of the genes directly disrupted in 22qDS encode for mitochondrial-localizing proteins. Here, we test the hypothesis that stem cell-derived neurons from subjects with the 22q11DS and SZ have mitochondrial deficits relative to typically developing controls. Human iPSCs from four lines of affected subjects and five lines of controls were differentiated into forebrain-like excitatory neurons. In the patient group, we find significant reductions of ATP levels that appear to be secondary to reduced activity in oxidative phosphorylation complexes I and IV. Protein products of mitochondrial-encoded genes are also reduced. As one of the genes deleted in the 22q11.2 region is MRPL40, a component of the mitochondrial ribosome, we generated a heterozygous mutation of MRPL40 in a healthy control iPSC line. Relative to its isogenic control, this line shows similar deficits in mitochondrial DNA-encoded proteins, ATP level, and complex I and IV activity. These results suggest that in the 22q11DS MRPL40 heterozygosity leads to reduced mitochondria ATP production secondary to altered mitochondrial protein levels. Such defects could have profound effects on neuronal function in vivo.
Collapse
Affiliation(s)
- Jianping Li
- Department of Psychiatry, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sean K Ryan
- Department of Psychiatry, The Children's Hospital of Philadelphia and the University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Erik Deboer
- Mallinckrodt Pharmaceuticals, Bedminster, NJ, USA
| | - Kieona Cook
- University of Pennsylvania, Philadelphia, PA, USA
| | - Shane Fitzgerald
- Department of Psychiatry, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Herbert M Lachman
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Douglas C Wallace
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia and Department of Pediatrics, Division of Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Ethan M Goldberg
- Department of Pediatrics, The Children's Hospital of Philadelphia and the University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Stewart A Anderson
- Department of Psychiatry, Children's Hospital of Philadelphia and the University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
45
|
Nucifora LG, MacDonald ML, Lee BJ, Peters ME, Norris AL, Orsburn BC, Yang K, Gleason K, Margolis RL, Pevsner J, Tamminga CA, Sweet RA, Ross CA, Sawa A, Nucifora FC. Increased Protein Insolubility in Brains From a Subset of Patients With Schizophrenia. Am J Psychiatry 2019; 176:730-743. [PMID: 31055969 DOI: 10.1176/appi.ajp.2019.18070864] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE The mechanisms leading to schizophrenia are likely to be diverse. However, there may be common pathophysiological pathways for subtypes of the disease. The authors tested the hypothesis that increased protein insolubility and ubiquitination underlie the pathophysiology for a subtype of schizophrenia. METHODS Prefrontal cortex and superior temporal gyrus from postmortem brains of individuals with and without schizophrenia were subjected to cold sarkosyl fractionation, separating proteins into soluble and insoluble fractions. Protein insolubility and ubiquitin levels were quantified for each insoluble fraction, with normalization to total homogenate protein. Mass spectrometry analysis was then performed to identify the protein contents of the insoluble fractions. The potential biological relevance of the detected proteins was assessed using Gene Ontology enrichment analysis and Ingenuity Pathway Analysis. RESULTS A subset of the schizophrenia brains showed an increase in protein insolubility and ubiquitination in the insoluble fraction. Mass spectrometry of the insoluble fraction revealed that brains with increased insolubility and ubiquitination exhibited a similar peptide expression by principal component analysis. The proteins that were significantly altered in the insoluble fraction were enriched for pathways relating to axon target recognition as well as nervous system development and function. CONCLUSIONS This study suggests a pathological process related to protein insolubility for a subset of patients with schizophrenia. Determining the molecular mechanism of this subtype of schizophrenia could lead to a better understanding of the pathways underlying the clinical phenotype in some patients with major mental illness as well as to improved nosology and identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Leslie G Nucifora
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Matthew L MacDonald
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Brian J Lee
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Matthew E Peters
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Alexis L Norris
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Benjamin C Orsburn
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Kun Yang
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Kelly Gleason
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Russell L Margolis
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Jonathan Pevsner
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Carol A Tamminga
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Robert A Sweet
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Christopher A Ross
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Akira Sawa
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Frederick C Nucifora
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| |
Collapse
|
46
|
Pérez MÁ, Morales C, Santander O, García F, Gómez I, Peñaloza-Sancho V, Fuentealba P, Dagnino-Subiabre A, Moya PR, Fuenzalida M. Ketamine-Treatment During Late Adolescence Impairs Inhibitory Synaptic Transmission in the Prefrontal Cortex and Working Memory in Adult Rats. Front Cell Neurosci 2019; 13:372. [PMID: 31481877 PMCID: PMC6710447 DOI: 10.3389/fncel.2019.00372] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 07/30/2019] [Indexed: 12/11/2022] Open
Abstract
Schizophrenia (SZ) is associated with changes in the structure and function of several brain areas. Several findings suggest that these impairments are related to a dysfunction in γ-aminobutyric acid (GABA) neurotransmission in brain areas such as the medial prefrontal cortex (mPFC), the hippocampus (HPC) and the primary auditory cortex (A1); however, it is still unclear how the GABAergic system is disrupted in these brain areas. Here, we examined the effect of ketamine (Ket) administration during late adolescence in rats on inhibition in the mPFC-, ventral HPC (vHPC), and A1. We observe that Ket treatment reduced the expression of the calcium-binding protein parvalbumin (PV) and the GABA-producing enzyme glutamic acid decarboxylase 67 (GAD67) as well as decreased inhibitory synaptic efficacy in the mPFC. In addition, Ket-treated rats performed worse in executive tasks that depend on the integrity and proper functioning of the mPFC. Conversely, we do not find such changes in vHPC or A1. Together, our results provide strong experimental support for the hypothesis that during adolescence, the function of the mPFC is more susceptible than that of HPC or A1 to NMDAR hypofunction, showing apparent structure specificity. Thus, the impairment of inhibitory circuitry in mPFC could be a convergent primary site of SZ-like behavior during the adulthood.
Collapse
Affiliation(s)
- Miguel Ángel Pérez
- Laboratorio de Plasticidad Neuronal, Universidad de Valparaíso, Valparaíso, Chile.,Facultad de Ciencias, Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Universidad de Valparaíso, Valparaíso, Chile.,Escuela de Ciencias de la Salud, Carrera de Kinesiología, Universidad Viña del Mar, Viña del Mar, Chile
| | - Camila Morales
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| | - Odra Santander
- Laboratorio de Plasticidad Neuronal, Universidad de Valparaíso, Valparaíso, Chile.,Facultad de Ciencias, Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Universidad de Valparaíso, Valparaíso, Chile.,Programa de Doctorado en Ciencias, Mención Neurociencias, Universidad de Valparaíso, Chile
| | - Francisca García
- Laboratorio de Plasticidad Neuronal, Universidad de Valparaíso, Valparaíso, Chile.,Facultad de Ciencias, Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Universidad de Valparaíso, Valparaíso, Chile.,Programa de Doctorado en Ciencias, Mención Neurociencias, Universidad de Valparaíso, Chile
| | - Isabel Gómez
- Laboratorio de Neurogenética, Universidad de Valparaíso, Valparaíso, Chile
| | - Valentín Peñaloza-Sancho
- Facultad de Ciencias, Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Universidad de Valparaíso, Valparaíso, Chile.,Laboratorio de Neurobiología del Estrés, Universidad de Valparaíso, Valparaíso, Chile
| | - Pablo Fuentealba
- Laboratory of Neural Circuits, Centro de Neurociencia Universidad Católica, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexies Dagnino-Subiabre
- Facultad de Ciencias, Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Universidad de Valparaíso, Valparaíso, Chile.,Laboratorio de Neurobiología del Estrés, Universidad de Valparaíso, Valparaíso, Chile
| | - Pablo R Moya
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile.,Laboratorio de Neurogenética, Universidad de Valparaíso, Valparaíso, Chile
| | - Marco Fuenzalida
- Laboratorio de Plasticidad Neuronal, Universidad de Valparaíso, Valparaíso, Chile.,Facultad de Ciencias, Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
47
|
Riedemann T. Diversity and Function of Somatostatin-Expressing Interneurons in the Cerebral Cortex. Int J Mol Sci 2019; 20:E2952. [PMID: 31212931 PMCID: PMC6627222 DOI: 10.3390/ijms20122952] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 06/08/2019] [Accepted: 06/14/2019] [Indexed: 02/01/2023] Open
Abstract
Inhibitory interneurons make up around 10-20% of the total neuron population in the cerebral cortex. A hallmark of inhibitory interneurons is their remarkable diversity in terms of morphology, synaptic connectivity, electrophysiological and neurochemical properties. It is generally understood that there are three distinct and non-overlapping interneuron classes in the mouse neocortex, namely, parvalbumin-expressing, 5-HT3A receptor-expressing and somatostatin-expressing interneuron classes. Each class is, in turn, composed of a multitude of subclasses, resulting in a growing number of interneuron classes and subclasses. In this review, I will focus on the diversity of somatostatin-expressing interneurons (SOM+ INs) in the cerebral cortex and elucidate their function in cortical circuits. I will then discuss pathological consequences of a malfunctioning of SOM+ INs in neurological disorders such as major depressive disorder, and present future avenues in SOM research and brain pathologies.
Collapse
Affiliation(s)
- Therese Riedemann
- Ludwig-Maximilians-University, Biomedical Center, Physiological Genomics, Großhaderner Str. 9, 82152 Planegg-Martinsried, Germany.
| |
Collapse
|
48
|
Gaillard-Bigot F, Zendjidjian XY, Kheloufi F, Casse-Perrot C, Guilhaumou R, Micallef J, Fakra E, Azorin JM, Blin O. Quantitative System Pharmacology (QSP): An Integrative Framework for paradigm change in the treatment of the first-episode schizophrenia. Encephale 2019; 44:S34-S38. [PMID: 30935485 DOI: 10.1016/s0013-7006(19)30077-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Despite the lack of progress in the curative treatment of mental illness, especially schizophrenia, the accumulation of neuroscience data over the past decade suggests the re-conceptualization of schizophrenia. With the advent of new biomarkers and cognitive tools, new neuroscience technologies such as functional dynamic connectivity and the identification of subtle clinical features; it is now possible to detect early stages at risk or prodromes of a first psychotic episode. Current concepts reconceptualizes schizophrenia as a neurodevelopmental disorder at early onset, with polygenic risk and only symptomatic treatment for positive symptoms at this time. The use of such technologies in the future suggests new diagnostic and therapeutic options. Next steps include new pharmacological perspectives and potential contributions of new technologies such as quantitative system pharmacology brain computational modeling approach.
Collapse
Affiliation(s)
- F Gaillard-Bigot
- Service de pharmacologie clinique et pharmacovigilance, CIC CPCET, assistance publique des hôpitaux de Marseille, Institut de neurosciences des systèmes, Inserm UMR 1106, université d'Aix-Marseille, France
| | - X-Y Zendjidjian
- Pôle psychiatrie centre, hôpital de la Conception, assistance publique des hôpitaux de Marseille, Marseille, France
| | - F Kheloufi
- Service de pharmacologie clinique et pharmacovigilance, CIC CPCET, assistance publique des hôpitaux de Marseille, Institut de neurosciences des systèmes, Inserm UMR 1106, université d'Aix-Marseille, France
| | - C Casse-Perrot
- Service de pharmacologie clinique et pharmacovigilance, CIC CPCET, assistance publique des hôpitaux de Marseille, Institut de neurosciences des systèmes, Inserm UMR 1106, université d'Aix-Marseille, France
| | - R Guilhaumou
- Service de pharmacologie clinique et pharmacovigilance, CIC CPCET, assistance publique des hôpitaux de Marseille, Institut de neurosciences des systèmes, Inserm UMR 1106, université d'Aix-Marseille, France
| | - J Micallef
- Service de pharmacologie clinique et pharmacovigilance, CIC CPCET, assistance publique des hôpitaux de Marseille, Institut de neurosciences des systèmes, Inserm UMR 1106, université d'Aix-Marseille, France
| | - E Fakra
- Department of Psychiatry, University Hospital of Saint-Etienne, Saint-Etienne, France, Inserm U1059, University of Lyon, Saint-Etienne F-42023, France
| | - J-M Azorin
- Department of Psychiatry, Sainte Marguerite University Hospital, Marseille, France
| | - O Blin
- Service de pharmacologie clinique et pharmacovigilance, CIC CPCET, assistance publique des hôpitaux de Marseille, Institut de neurosciences des systèmes, Inserm UMR 1106, université d'Aix-Marseille, France.
| |
Collapse
|
49
|
Cao B, Jin M, Brietzke E, McIntyre RS, Wang D, Rosenblat JD, Ragguett RM, Zhang C, Sun X, Rong C, Wang J. Serum metabolic profiling using small molecular water-soluble metabolites in individuals with schizophrenia: A longitudinal study using a pre-post-treatment design. Psychiatry Clin Neurosci 2019; 73:100-108. [PMID: 30156046 DOI: 10.1111/pcn.12779] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 07/24/2018] [Accepted: 08/21/2018] [Indexed: 12/12/2022]
Abstract
AIM We sought to compare alterations in serum bioenergetic markers within a well-characterized sample of adults with schizophrenia at baseline and after 8 weeks of pharmacological treatment with the hypothesis that treatment would be associated with significant changes in bioenergetic markers given the role of bioenergetic dysfunction in schizophrenia. METHODS We recruited adults with schizophrenia (n = 122) who had not received pharmacological treatment for at least 1 month prior to enrollment, including drug-naïve (i.e., first-episode) participants and treatment non-adherent participants. Pre- and post-treatment serum samples were analyzed using liquid chromatography-tandem mass spectrometry. RESULTS Metabolites with the greatest change, when comparing pre- and post-treatment levels, were identified revealing 14 water-soluble metabolites of interest. The composition of these metabolites was: amino acids (n = 6), carnitines (n = 4), polar lipids (n = 3), and organic acid (n = 1). All amino acids and lysophosphatidylcholines (LysoPC) were increased, while the four carnitines - oleoylcarnitine, L-palmitoylcarnitine, linoleyl carnitine, and L-acetylcarnitine - were decreased post-treatment. Of these metabolite biomarkers, six - oleoylcarnitine, linoleyl carnitine, L-acetylcarnitine, LysoPC(15:0), D-glutamic acid, and L-arginine - were identified as having most consistently and predictably changed after 8 weeks of treatment. CONCLUSION The current study identified several bioenergetic markers that consistently change with pharmacological treatment. These bioenergetic changes may provide further insights into the pathophysiology of schizophrenia along with furthering our understanding of the mechanisms subserving both the effects (e.g., antipsychotic effects) and side-effects (e.g., metabolic syndrome) of antipsychotics.
Collapse
Affiliation(s)
- Bing Cao
- Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, China
| | - Min Jin
- School of Public Health, Baotou Medical College, Baotou, China
| | - Elisa Brietzke
- Mood Disorders Psychopharmacology Unit, Toronto Western Hospital, University Health Network, Toronto, Canada
| | - Roger S McIntyre
- Mood Disorders Psychopharmacology Unit, Toronto Western Hospital, University Health Network, Toronto, Canada.,The Brain and Cognition Discovery Foundation, Toronto, Canada
| | - Dongfang Wang
- Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, China
| | - Joshua D Rosenblat
- Mood Disorders Psychopharmacology Unit, Toronto Western Hospital, University Health Network, Toronto, Canada
| | - Renee-Marie Ragguett
- Mood Disorders Psychopharmacology Unit, Toronto Western Hospital, University Health Network, Toronto, Canada
| | | | - Xiaoyu Sun
- Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, China
| | - Carola Rong
- Mood Disorders Psychopharmacology Unit, Toronto Western Hospital, University Health Network, Toronto, Canada
| | - Jingyu Wang
- Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, China.,Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, China
| |
Collapse
|
50
|
Maćkowiak M, Latusz J, Głowacka U, Bator E, Bilecki W. Adolescent social isolation affects parvalbumin expression in the medial prefrontal cortex in the MAM-E17 model of schizophrenia. Metab Brain Dis 2019; 34:341-352. [PMID: 30519836 DOI: 10.1007/s11011-018-0359-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 11/28/2018] [Indexed: 10/27/2022]
Abstract
Altered parvalbumin (PV) expression is observed in the prefrontal cortex of subjects with schizophrenia. Environmental context, particularly during adolescence, might regulate PV expression. In the present study, we investigated the effect of adolescent social isolation (SI) on PV expression in the medial prefrontal cortex in a neurodevelopmental model (MAM-E17) of schizophrenia. SI exposure occurred from postnatal day 30 to 40, followed by resocialization until late adolescence or early adulthood. PV mRNA and protein levels, as well as the number of PV cells, were analysed at these ages. Moreover, epigenetic regulation of PV expression by histone methylation was examined by measuring the total and PV gene-bound H3K4me3 levels. MAM only decreased levels of the PV mRNA and protein in adulthood. Decreases in total H3K4me3 levels and its level at the PV gene were also observed at this age. In contrast, in late adolescence, SI induced a decrease in the expression of the PV mRNA in the MAM group that was related to the reduction in total and PV gene-bound H3K4me3 levels. However, at this age, SI increased the levels of the PV protein in both the control and MAM groups. In adulthood, SI did not affect PV mRNA or H3K4me3 levels but decreased levels of the PV protein in both groups. Both MAM and SI failed to change the number of PV cells at any age. The results indicate that adolescent SI accelerated epigenetic impairments of PV expression in MAM-E17 rats; however, subsequent resocialization abolished this dysfunction, but failed to prevent alterations in PV protein.
Collapse
Affiliation(s)
- Marzena Maćkowiak
- Institute of Pharmacology, Polish Academy of Sciences, Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Smętna Street 12, 31-343, Kraków, Poland.
| | - Joachim Latusz
- Institute of Pharmacology, Polish Academy of Sciences, Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Smętna Street 12, 31-343, Kraków, Poland
| | - Urszula Głowacka
- Institute of Pharmacology, Polish Academy of Sciences, Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Smętna Street 12, 31-343, Kraków, Poland
| | - Ewelina Bator
- Institute of Pharmacology, Polish Academy of Sciences, Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Smętna Street 12, 31-343, Kraków, Poland
| | - Wiktor Bilecki
- Institute of Pharmacology, Polish Academy of Sciences, Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Smętna Street 12, 31-343, Kraków, Poland
| |
Collapse
|