101
|
Mannekote Thippaiah S, Pradhan B, Voyiaziakis E, Shetty R, Iyengar S, Olson C, Tang YY. Possible Role of Parvalbumin Interneurons in Meditation and Psychiatric Illness. J Neuropsychiatry Clin Neurosci 2022; 34:113-123. [PMID: 35040663 DOI: 10.1176/appi.neuropsych.21050136] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Parvalbumin (PV) interneurons are present in multiple brain regions and produce complex influences on brain functioning. An increasing number of research findings indicate that the function of these interneurons is more complex than solely to inhibit pyramidal neurons in the cortex. They generate feedback and feedforward inhibition of cortical neurons, and they are critically involved in the generation of neuronal network oscillation. These oscillations, generated by various brain regions, are linked to perceptions, thought processes, and cognitive functions, all of which, in turn, influence human emotions and behavior. Both animal and human studies consistently have found that meditation practice results in enhancement in the effects of alpha-, theta-, and gamma-frequency oscillations, which may correspond to positive changes in cognition, emotion, conscious awareness, and, subsequently, behavior. Although the study of meditation has moved into mainstream neuroscience research, the link between PV interneurons and any role they might play in meditative states remains elusive. This article is focused primarily on gamma-frequency oscillation, which is generated by PV interneurons, to develop insight and perspective into the role of PV interneurons in meditation. This article also points to new and emerging directions that address whether this role of PV interneurons in meditation extends to a beneficial, and potentially therapeutic, role in the treatment of common psychiatric disorders, including schizophrenia.
Collapse
Affiliation(s)
- Srinagesh Mannekote Thippaiah
- Department of Psychiatry, Valleywise Behavioral Health Center, School of Medicine, Creighton University, Phoenix (Mannekote Thippaiah, Olson); Division of Neuromodulation and Integrative Psychiatry, Department of Psychiatry and Pediatrics, Cooper Medical School, Rowan University, Camden, N.J. (Pradhan); Department of Psychiatry, Donald and Barbara Zucker School of Medicine, Hofstra/Northwell, Glen Oaks, N.Y. (Voyiaziakis); Department of Neuroscience, College of Biological Sciences, University of Minnesota, Minneapolis (Shetty); American Museum of Natural History, New York (Iyengar); Psychiatry Division, District Medical Group, Phoenix (Olson); and College of Health Solutions, Arizona State University, Tempe (Tang)
| | - Basant Pradhan
- Department of Psychiatry, Valleywise Behavioral Health Center, School of Medicine, Creighton University, Phoenix (Mannekote Thippaiah, Olson); Division of Neuromodulation and Integrative Psychiatry, Department of Psychiatry and Pediatrics, Cooper Medical School, Rowan University, Camden, N.J. (Pradhan); Department of Psychiatry, Donald and Barbara Zucker School of Medicine, Hofstra/Northwell, Glen Oaks, N.Y. (Voyiaziakis); Department of Neuroscience, College of Biological Sciences, University of Minnesota, Minneapolis (Shetty); American Museum of Natural History, New York (Iyengar); Psychiatry Division, District Medical Group, Phoenix (Olson); and College of Health Solutions, Arizona State University, Tempe (Tang)
| | - Emanuel Voyiaziakis
- Department of Psychiatry, Valleywise Behavioral Health Center, School of Medicine, Creighton University, Phoenix (Mannekote Thippaiah, Olson); Division of Neuromodulation and Integrative Psychiatry, Department of Psychiatry and Pediatrics, Cooper Medical School, Rowan University, Camden, N.J. (Pradhan); Department of Psychiatry, Donald and Barbara Zucker School of Medicine, Hofstra/Northwell, Glen Oaks, N.Y. (Voyiaziakis); Department of Neuroscience, College of Biological Sciences, University of Minnesota, Minneapolis (Shetty); American Museum of Natural History, New York (Iyengar); Psychiatry Division, District Medical Group, Phoenix (Olson); and College of Health Solutions, Arizona State University, Tempe (Tang)
| | - Rashika Shetty
- Department of Psychiatry, Valleywise Behavioral Health Center, School of Medicine, Creighton University, Phoenix (Mannekote Thippaiah, Olson); Division of Neuromodulation and Integrative Psychiatry, Department of Psychiatry and Pediatrics, Cooper Medical School, Rowan University, Camden, N.J. (Pradhan); Department of Psychiatry, Donald and Barbara Zucker School of Medicine, Hofstra/Northwell, Glen Oaks, N.Y. (Voyiaziakis); Department of Neuroscience, College of Biological Sciences, University of Minnesota, Minneapolis (Shetty); American Museum of Natural History, New York (Iyengar); Psychiatry Division, District Medical Group, Phoenix (Olson); and College of Health Solutions, Arizona State University, Tempe (Tang)
| | - Sloka Iyengar
- Department of Psychiatry, Valleywise Behavioral Health Center, School of Medicine, Creighton University, Phoenix (Mannekote Thippaiah, Olson); Division of Neuromodulation and Integrative Psychiatry, Department of Psychiatry and Pediatrics, Cooper Medical School, Rowan University, Camden, N.J. (Pradhan); Department of Psychiatry, Donald and Barbara Zucker School of Medicine, Hofstra/Northwell, Glen Oaks, N.Y. (Voyiaziakis); Department of Neuroscience, College of Biological Sciences, University of Minnesota, Minneapolis (Shetty); American Museum of Natural History, New York (Iyengar); Psychiatry Division, District Medical Group, Phoenix (Olson); and College of Health Solutions, Arizona State University, Tempe (Tang)
| | - Carol Olson
- Department of Psychiatry, Valleywise Behavioral Health Center, School of Medicine, Creighton University, Phoenix (Mannekote Thippaiah, Olson); Division of Neuromodulation and Integrative Psychiatry, Department of Psychiatry and Pediatrics, Cooper Medical School, Rowan University, Camden, N.J. (Pradhan); Department of Psychiatry, Donald and Barbara Zucker School of Medicine, Hofstra/Northwell, Glen Oaks, N.Y. (Voyiaziakis); Department of Neuroscience, College of Biological Sciences, University of Minnesota, Minneapolis (Shetty); American Museum of Natural History, New York (Iyengar); Psychiatry Division, District Medical Group, Phoenix (Olson); and College of Health Solutions, Arizona State University, Tempe (Tang)
| | - Yi-Yuan Tang
- Department of Psychiatry, Valleywise Behavioral Health Center, School of Medicine, Creighton University, Phoenix (Mannekote Thippaiah, Olson); Division of Neuromodulation and Integrative Psychiatry, Department of Psychiatry and Pediatrics, Cooper Medical School, Rowan University, Camden, N.J. (Pradhan); Department of Psychiatry, Donald and Barbara Zucker School of Medicine, Hofstra/Northwell, Glen Oaks, N.Y. (Voyiaziakis); Department of Neuroscience, College of Biological Sciences, University of Minnesota, Minneapolis (Shetty); American Museum of Natural History, New York (Iyengar); Psychiatry Division, District Medical Group, Phoenix (Olson); and College of Health Solutions, Arizona State University, Tempe (Tang)
| |
Collapse
|
102
|
Bühning F, Miguel Telega L, Tong Y, Pereira J, Coenen V, Döbrössy M. Electrophysiological and molecular effects of bilateral deep brain stimulation of the medial forebrain bundle in a rodent model of depression. Exp Neurol 2022; 355:114122. [DOI: 10.1016/j.expneurol.2022.114122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 05/10/2022] [Accepted: 05/18/2022] [Indexed: 11/04/2022]
|
103
|
Khushboo, Siddiqi NJ, de Lourdes Pereira M, Sharma B. Neuroanatomical, Biochemical, and Functional Modifications in Brain Induced by Treatment with Antidepressants. Mol Neurobiol 2022; 59:3564-3584. [DOI: 10.1007/s12035-022-02780-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 02/24/2022] [Indexed: 12/13/2022]
|
104
|
Wang B, Zartaloudi E, Linden JF, Bramon E. Neurophysiology in psychosis: The quest for disease biomarkers. Transl Psychiatry 2022; 12:100. [PMID: 35277479 PMCID: PMC8917164 DOI: 10.1038/s41398-022-01860-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 02/14/2022] [Accepted: 02/18/2022] [Indexed: 01/11/2023] Open
Abstract
Psychotic disorders affect 3% of the population at some stage in life, are a leading cause of disability, and impose a great economic burden on society. Major breakthroughs in the genetics of psychosis have not yet been matched by an understanding of its neurobiology. Biomarkers of perception and cognition obtained through non-invasive neurophysiological tools, especially EEG, offer a unique opportunity to gain mechanistic insights. Techniques for measuring neurophysiological markers are inexpensive and ubiquitous, thus having the potential as an accessible tool for patient stratification towards early treatments leading to better outcomes. In this paper, we review the literature on neurophysiological markers for psychosis and their relevant disease mechanisms, mainly covering event-related potentials including P50/N100 sensory gating, mismatch negativity, and the N100 and P300 waveforms. While several neurophysiological deficits are well established in patients with psychosis, more research is needed to study neurophysiological markers in their unaffected relatives and individuals at clinical high risk. We need to harness EEG to investigate markers of disease risk as key steps to elucidate the aetiology of psychosis and facilitate earlier detection and treatment.
Collapse
Affiliation(s)
- Baihan Wang
- Division of Psychiatry, University College London, London, UK.
| | - Eirini Zartaloudi
- Division of Psychiatry, University College London, London, UK.
- Institute of Clinical Trials and Methodology, University College London, London, UK.
| | - Jennifer F Linden
- Ear Institute, University College London, London, UK
- Department of Neuroscience, Physiology & Pharmacology, University College London, London, UK
| | - Elvira Bramon
- Division of Psychiatry, University College London, London, UK
- Institute of Cognitive Neuroscience, University College London, London, UK
| |
Collapse
|
105
|
Haaf M, Curic S, Steinmann S, Rauh J, Leicht G, Mulert C. Glycine attenuates impairments of stimulus-evoked gamma oscillations in the ketamine model of schizophrenia. Neuroimage 2022; 251:119004. [PMID: 35176492 DOI: 10.1016/j.neuroimage.2022.119004] [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: 05/17/2021] [Revised: 01/18/2022] [Accepted: 02/13/2022] [Indexed: 01/17/2023] Open
Abstract
Although a substantial number of studies suggests some clinical benefit concerning negative symptoms in schizophrenia through the modulation of NMDA-receptor function, none of these approaches achieved clinical approval. Given the large body of evidence concerning glutamatergic dysfunction in a subgroup of patients, biomarkers to identify those with a relevant clinical benefit through glutamatergic modulation are urgently needed. A similar reduction of the early auditory evoked gamma-band response (aeGBR) as found in schizophrenia patients can be observed in healthy subjects following the application of an NMDA-receptor antagonist in the ketamine-model, which addresses the excitation / inhibition (E/I) imbalance of the disease. Moreover, this oscillatory change can be related to the emergence of negative symptoms. Accordingly, this study investigated whether glycine-related increases of the aeGBR, through NMDA-receptor co-agonism, accompany an improvement concerning negative symptoms in the ketamine-model. The impact of subanesthetic ketamine doses and the pretreatment with glycine was examined in twenty-four healthy male participants while performing a cognitively demanding aeGBR paradigm with 64-channel electroencephalography. Negative Symptoms were assessed through the PANSS. S-Ketamine alone caused a reduction of the aeGBR amplitude associated with more pronounced negative symptoms compared to placebo. Pretreatment with glycine attenuated both, the ketamine-induced alterations of the aeGBR amplitude and the increased PANSS negative scores in glycine-responders, classified based on relative aeGBR increase. Thus, we propose that the aeGBR represents a possible biomarker for negative symptoms in schizophrenia related to insufficient glutamatergic neurotransmission. This would allow to identify patients with negative symptoms, who might benefit from glutamatergic treatment.
Collapse
Affiliation(s)
- Moritz Haaf
- Department of Psychiatry and Psychotherapy, Psychiatry Neuroimaging Branch (PNB), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stjepan Curic
- Department of Psychiatry and Psychotherapy, Psychiatry Neuroimaging Branch (PNB), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Saskia Steinmann
- Department of Psychiatry and Psychotherapy, Psychiatry Neuroimaging Branch (PNB), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jonas Rauh
- Department of Psychiatry and Psychotherapy, Psychiatry Neuroimaging Branch (PNB), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gregor Leicht
- Department of Psychiatry and Psychotherapy, Psychiatry Neuroimaging Branch (PNB), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christoph Mulert
- Department of Psychiatry and Psychotherapy, Psychiatry Neuroimaging Branch (PNB), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Center of Psychiatry, Justus-Liebig University, Giessen, Germany.
| |
Collapse
|
106
|
Montgomery DP, Hayden DJ, Chaloner FA, Cooke SF, Bear MF. Stimulus-Selective Response Plasticity in Primary Visual Cortex: Progress and Puzzles. Front Neural Circuits 2022; 15:815554. [PMID: 35173586 PMCID: PMC8841555 DOI: 10.3389/fncir.2021.815554] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/29/2021] [Indexed: 11/23/2022] Open
Abstract
Stimulus-selective response plasticity (SRP) is a robust and lasting modification of primary visual cortex (V1) that occurs in response to exposure to novel visual stimuli. It is readily observed as a pronounced increase in the magnitude of visual evoked potentials (VEPs) recorded in response to phase-reversing grating stimuli in neocortical layer 4. The expression of SRP at the individual neuron level is equally robust, but the qualities vary depending on the neuronal type and how activity is measured. This form of plasticity is highly selective for stimulus features such as stimulus orientation, spatial frequency, and contrast. Several key insights into the significance and underlying mechanisms of SRP have recently been made. First, it occurs concomitantly and shares core mechanisms with behavioral habituation, indicating that SRP reflects the formation of long-term familiarity that can support recognition of innocuous stimuli. Second, SRP does not manifest within a recording session but only emerges after an off-line period of several hours that includes sleep. Third, SRP requires not only canonical molecular mechanisms of Hebbian synaptic plasticity within V1, but also the opposing engagement of two key subclasses of cortical inhibitory neuron: the parvalbumin- and somatostatin-expressing GABAergic interneurons. Fourth, pronounced shifts in the power of cortical oscillations from high frequency (gamma) to low frequency (alpha/beta) oscillations provide respective readouts of the engagement of these inhibitory neuronal subtypes following familiarization. In this article we will discuss the implications of these findings and the outstanding questions that remain to gain a deeper understanding of this striking form of experience-dependent plasticity.
Collapse
Affiliation(s)
- Daniel P. Montgomery
- Department of Brain and Cognitive Sciences, The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Dustin J. Hayden
- Department of Brain and Cognitive Sciences, The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Francesca A. Chaloner
- MRC Centre for Neurodevelopmental Disorders (CNDD), King’s College London, London, United Kingdom
- Department of Basic and Clinical Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Samuel F. Cooke
- MRC Centre for Neurodevelopmental Disorders (CNDD), King’s College London, London, United Kingdom
- Department of Basic and Clinical Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Mark F. Bear
- Department of Brain and Cognitive Sciences, The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, United States
| |
Collapse
|
107
|
Ulloa JL. The Control of Movements via Motor Gamma Oscillations. Front Hum Neurosci 2022; 15:787157. [PMID: 35111006 PMCID: PMC8802912 DOI: 10.3389/fnhum.2021.787157] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/22/2021] [Indexed: 01/07/2023] Open
Abstract
The ability to perform movements is vital for our daily life. Our actions are embedded in a complex environment where we need to deal efficiently in the face of unforeseen events. Neural oscillations play an important role in basic sensorimotor processes related to the execution and preparation of movements. In this review, I will describe the state of the art regarding the role of motor gamma oscillations in the control of movements. Experimental evidence from electrophysiological studies has shown that motor gamma oscillations accomplish a range of functions in motor control beyond merely signaling the execution of movements. However, these additional aspects associated with motor gamma oscillation remain to be fully clarified. Future work on different spatial, temporal and spectral scales is required to further understand the implications of gamma oscillations in motor control.
Collapse
Affiliation(s)
- José Luis Ulloa
- Programa de Investigación Asociativa (PIA) en Ciencias Cognitivas, Centro de Investigación en Ciencias Cognitivas (CICC), Facultad de Psicología, Universidad de Talca, Talca, Chile
| |
Collapse
|
108
|
Okamoto H, Onitsuka T, Kuga H, Oribe N, Nakayama N, Fukushima S, Nakao T, Ueno T. Decreased BOLD signals elicited by 40-Hz auditory stimulation of the right primary auditory cortex in bipolar disorder: An fMRI study. Front Psychiatry 2022; 13:833896. [PMID: 36186861 PMCID: PMC9519862 DOI: 10.3389/fpsyt.2022.833896] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 08/15/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND A number studies have been conducted on abnormalities in the cortical circuitry of gamma oscillations, including deficit in auditory steady-state response (ASSR) to gamma-frequency (≧ 30-Hz) stimulation, in patients with bipolar disorder (BD). In the current study, we investigated neural responses during click stimulation by blood oxygen level-dependent (BOLD) signals. We focused on Broadman 41 and 42, the main sources of ASSR. MATERIALS AND METHODS We acquired BOLD responses elicited by click trains of 80-, 40-, 30- and 20-Hz frequencies from 25 patients with BD to 27 healthy controls (HC) with normal hearing between 22 and 59 years of age assessed via a standard general linear-model-based analysis. We extracted contrast values by identifying the primary auditory cortex and Brodmann areas 41 and 42 as regions of interest (ROI)s. RESULTS BD group showed significantly decreased ASSR-BOLD signals in response to 40-Hz stimuli compared to the HC group in the right Brodmann areas 41 and 42. We found significant negative correlations between the BOLD change in the right Brodmann areas 41 and 42 and Structured Interview Guide for the Hamilton Depression Rating Scale (SIGH-D) scores, also the BOLD change in the right Brodmann areas 41 and 42 and the Positive and Negative Syndrome Scale (PANSS)-Negative scores. CONCLUSION The observed decrease in BOLD signal patterns in the right primary auditory cortex during 40-Hz ASSR may be a potential biomarker option for bipolar disorder.
Collapse
Affiliation(s)
- Hiroshi Okamoto
- Division of Clinical Research, National Hospital Organization, Hizen Psychiatric Medical Center, Saga, Japan.,Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Toshiaki Onitsuka
- Department of Neuroimaging Psychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hironori Kuga
- Division of Clinical Research, National Hospital Organization, Hizen Psychiatric Medical Center, Saga, Japan.,National Center for Cognitive Behavioral Therapy and Research, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Naoya Oribe
- Division of Clinical Research, National Hospital Organization, Hizen Psychiatric Medical Center, Saga, Japan.,Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Naho Nakayama
- Division of Clinical Research, National Hospital Organization, Hizen Psychiatric Medical Center, Saga, Japan
| | - Shou Fukushima
- Division of Clinical Research, National Hospital Organization, Hizen Psychiatric Medical Center, Saga, Japan.,Medical Corporation Kouseikai, Michinoo Hospital, Nagasaki, Japan
| | - Tomohiro Nakao
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takefumi Ueno
- Division of Clinical Research, National Hospital Organization, Hizen Psychiatric Medical Center, Saga, Japan.,Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
109
|
Rais M, Lovelace JW, Shuai XS, Woodard W, Bishay S, Estrada L, Sharma AR, Nguy A, Kulinich A, Pirbhoy PS, Palacios AR, Nelson DL, Razak KA, Ethell IM. Functional consequences of postnatal interventions in a mouse model of Fragile X syndrome. Neurobiol Dis 2022; 162:105577. [PMID: 34871737 DOI: 10.1016/j.nbd.2021.105577] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 10/22/2021] [Accepted: 12/02/2021] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Fragile X syndrome (FXS) is a leading genetic cause of autism and intellectual disability with cortical hyperexcitability and sensory hypersensitivity attributed to loss and hypofunction of inhibitory parvalbumin-expressing (PV) cells. Our studies provide novel insights into the role of excitatory neurons in abnormal development of PV cells during a postnatal period of inhibitory circuit refinement. METHODS To achieve Fragile X mental retardation gene (Fmr1) deletion and re-expression in excitatory neurons during the postnatal day (P)14-P21 period, we generated CreCaMKIIa/Fmr1Flox/y (cOFF) and CreCaMKIIa/Fmr1FloxNeo/y (cON) mice, respectively. Cortical phenotypes were evaluated in adult mice using biochemical, cellular, clinically relevant electroencephalogram (EEG) and behavioral tests. RESULTS We found that similar to global Fmr1 KO mice, the density of PV-expressing cells, their activation, and sound-evoked gamma synchronization were impaired in cOFF mice, but the phenotypes were improved in cON mice. cOFF mice also showed enhanced cortical gelatinase activity and baseline EEG gamma power, which were reduced in cON mice. In addition, TrkB phosphorylation and PV levels were lower in cOFF mice, which also showed increased locomotor activity and anxiety-like behaviors. Remarkably, when FMRP levels were restored in only excitatory neurons during the P14-P21 period, TrkB phosphorylation and mouse behaviors were also improved. CONCLUSIONS These results indicate that postnatal deletion or re-expression of FMRP in excitatory neurons is sufficient to elicit or ameliorate structural and functional cortical deficits, and abnormal behaviors in mice, informing future studies about appropriate treatment windows and providing fundamental insights into the cellular mechanisms of cortical circuit dysfunction in FXS.
Collapse
Affiliation(s)
- Maham Rais
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Jonathan W Lovelace
- Department of Psychology, University of California Riverside, Riverside, CA 92521, USA
| | - Xinghao S Shuai
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Walker Woodard
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Steven Bishay
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Leo Estrada
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Ashwin R Sharma
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Austin Nguy
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Anna Kulinich
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Patricia S Pirbhoy
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Arnold R Palacios
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | | | - Khaleel A Razak
- Department of Psychology, University of California Riverside, Riverside, CA 92521, USA; Neuroscience Graduate Program, University of California Riverside, Riverside, CA 92521, USA
| | - Iryna M Ethell
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA; Neuroscience Graduate Program, University of California Riverside, Riverside, CA 92521, USA.
| |
Collapse
|
110
|
Pathway-specific contribution of parvalbumin interneuron NMDARs to synaptic currents and thalamocortical feedforward inhibition. Mol Psychiatry 2022; 27:5124-5134. [PMID: 36075962 PMCID: PMC9763122 DOI: 10.1038/s41380-022-01747-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 08/08/2022] [Accepted: 08/11/2022] [Indexed: 01/19/2023]
Abstract
Prefrontal cortex (PFC) is a site of information convergence important for behaviors relevant to psychiatric disorders. Despite the importance of inhibitory GABAergic parvalbumin-expressing (PV+) interneurons to PFC circuit function and decades of interest in N-methyl-D-aspartate receptors (NMDARs) in these neurons, examples of defined circuit functions that depend on PV+ interneuron NMDARs have been elusive. Indeed, it remains controversial whether all PV+ interneurons contain functional NMDARs in adult PFC, which has major consequences for hypotheses of the pathogenesis of psychiatric disorders. Using a combination of fluorescent in situ hybridization, pathway-specific optogenetics, cell-type-specific gene ablation, and electrophysiological recordings from PV+ interneurons, here we resolve this controversy. We found that nearly 100% of PV+ interneurons in adult medial PFC (mPFC) express transcripts encoding GluN1 and GluN2B, and they have functional NMDARs. By optogenetically stimulating corticocortical and thalamocortical inputs to mPFC, we show that synaptic NMDAR contribution to PV+ interneuron EPSCs is pathway-specific, which likely explains earlier reports of PV+ interneurons without synaptic NMDAR currents. Lastly, we report a major contribution of NMDARs in PV+ interneurons to thalamus-mediated feedforward inhibition in adult mPFC circuits, suggesting molecular and circuit-based mechanisms for cognitive impairment under conditions of reduced NMDAR function. These findings represent an important conceptual advance that has major implications for hypotheses of the pathogenesis of psychiatric disorders.
Collapse
|
111
|
Farashahi S, Soltani A. Computational mechanisms of distributed value representations and mixed learning strategies. Nat Commun 2021; 12:7191. [PMID: 34893597 PMCID: PMC8664930 DOI: 10.1038/s41467-021-27413-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 11/16/2021] [Indexed: 11/25/2022] Open
Abstract
Learning appropriate representations of the reward environment is challenging in the real world where there are many options, each with multiple attributes or features. Despite existence of alternative solutions for this challenge, neural mechanisms underlying emergence and adoption of value representations and learning strategies remain unknown. To address this, we measure learning and choice during a multi-dimensional probabilistic learning task in humans and trained recurrent neural networks (RNNs) to capture our experimental observations. We find that human participants estimate stimulus-outcome associations by learning and combining estimates of reward probabilities associated with the informative feature followed by those of informative conjunctions. Through analyzing representations, connectivity, and lesioning of the RNNs, we demonstrate this mixed learning strategy relies on a distributed neural code and opponency between excitatory and inhibitory neurons through value-dependent disinhibition. Together, our results suggest computational and neural mechanisms underlying emergence of complex learning strategies in naturalistic settings.
Collapse
Affiliation(s)
- Shiva Farashahi
- Department of Psychological and Brain Sciences, Dartmouth College, Hanover, NH, USA.
- Center for Computational Neuroscience, Flatiron Institute, Simons Foundation, New York, NY, USA.
| | - Alireza Soltani
- Department of Psychological and Brain Sciences, Dartmouth College, Hanover, NH, USA.
| |
Collapse
|
112
|
Park M, Hoang GM, Nguyen T, Lee E, Jung HJ, Choe Y, Lee MH, Hwang JY, Kim JG, Kim T. Effects of transcranial ultrasound stimulation pulsed at 40 Hz on Aβ plaques and brain rhythms in 5×FAD mice. Transl Neurodegener 2021; 10:48. [PMID: 34872618 PMCID: PMC8650290 DOI: 10.1186/s40035-021-00274-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 11/25/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common cause of dementia, and is characterized by amyloid-β (Aβ) plaques and tauopathy. Reducing Aβ has been considered a major AD treatment strategy in pharmacological and non-pharmacological approaches. Impairment of gamma oscillations, which play an important role in perception and cognitive function, has been shown in mouse AD models and human patients. Recently, the therapeutic effect of gamma entrainment in AD mouse models has been reported. Given that ultrasound is an emerging neuromodulation modality, we investigated the effect of ultrasound stimulation pulsed at gamma frequency (40 Hz) in an AD mouse model. METHODS We implanted electroencephalogram (EEG) electrodes and a piezo-ceramic disc ultrasound transducer on the skull surface of 6-month-old 5×FAD and wild-type control mice (n = 12 and 6, respectively). Six 5×FAD mice were treated with two-hour ultrasound stimulation at 40 Hz daily for two weeks, and the other six mice received sham treatment. Soluble and insoluble Aβ levels in the brain were measured by enzyme-linked immunosorbent assay. Spontaneous EEG gamma power was computed by wavelet analysis, and the brain connectivity was examined with phase-locking value and cross-frequency phase-amplitude coupling. RESULTS We found that the total Aβ42 levels, especially insoluble Aβ42, in the treatment group decreased in pre- and infra-limbic cortex (PIL) compared to that of the sham treatment group. A reduction in the number of Aβ plaques was also observed in the hippocampus. There was no increase in microbleeding in the transcranial ultrasound stimulation (tUS) group. In addition, the length and number of microglial processes decreased in PIL and hippocampus. Encelphalographic spontaneous gamma power was increased, and cross-frequency coupling was normalized, implying functional improvement after tUS stimulation. CONCLUSION These results suggest that the transcranial ultrasound-based gamma-band entrainment technique can be an effective therapy for AD by reducing the Aβ load and improving brain connectivity.
Collapse
Affiliation(s)
- Mincheol Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Gia Minh Hoang
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Thien Nguyen
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Eunkyung Lee
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Hyun Jin Jung
- Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Youngshik Choe
- Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Moon Hwan Lee
- Department of Information and Communication Engineering, Daegu Gyeongbuk Institute of Science and Technology, Daegu, 42988, Republic of Korea
| | - Jae Youn Hwang
- Department of Information and Communication Engineering, Daegu Gyeongbuk Institute of Science and Technology, Daegu, 42988, Republic of Korea
| | - Jae Gwan Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea.
| | - Tae Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea.
| |
Collapse
|
113
|
Delgado-Sallent C, Nebot P, Gener T, Fath AB, Timplalexi M, Puig MV. Atypical, but Not Typical, Antipsychotic Drugs Reduce Hypersynchronized Prefrontal-Hippocampal Circuits during Psychosis-Like States in Mice: Contribution of 5-HT2A and 5-HT1A Receptors. Cereb Cortex 2021; 32:3472-3487. [PMID: 34875009 DOI: 10.1093/cercor/bhab427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 10/30/2021] [Accepted: 11/02/2021] [Indexed: 11/12/2022] Open
Abstract
Neural synchrony and functional connectivity are disrupted in schizophrenia. We investigated changes in prefrontal-hippocampal neural dynamics during psychosis-like states induced by the NMDAR antagonist phencyclidine and subsequent rescue by two atypical antipsychotic drugs (AAPDs), risperidone and clozapine, and the classical APD haloperidol. The psychotomimetic effects of phencyclidine were associated with prefrontal hypersynchronization, hippocampal desynchronization, and disrupted circuit connectivity. Phencyclidine boosted prefrontal oscillatory power at atypical bands within delta, gamma, and high frequency ranges, while irregular cross-frequency and spike-LFP coupling emerged. In the hippocampus, phencyclidine enhanced delta rhythms but suppressed theta oscillations, theta-gamma coupling, and theta-beta spike-LFP coupling. Baseline interregional theta-gamma coupling, theta phase coherence, and hippocampus-to-cortex theta signals were redirected to delta frequencies. Risperidone and clozapine, but not haloperidol, reduced phencyclidine-induced prefrontal and cortical-hippocampal hypersynchrony. None of the substances restored hippocampal and circuit desynchronization. These results suggest that AAPDs, but not typical APDs, target prefrontal-hippocampal pathways to elicit antipsychotic action. We investigated whether the affinity of AAPDs for serotonin receptors could explain their distinct effects. Serotonin 5-HT2AR antagonism by M100907 and 5-HT1AR agonism by 8-OH-DPAT reduced prefrontal hypersynchronization. Our results point to fundamentally different neural mechanisms underlying the action of atypical versus typical APDs with selective contribution of serotonin receptors.
Collapse
Affiliation(s)
- Cristina Delgado-Sallent
- Hospital del Mar Medical Research Institute, Barcelona Biomedical Research Park, 08003 Barcelona, Spain
| | - Pau Nebot
- Hospital del Mar Medical Research Institute, Barcelona Biomedical Research Park, 08003 Barcelona, Spain
| | - Thomas Gener
- Hospital del Mar Medical Research Institute, Barcelona Biomedical Research Park, 08003 Barcelona, Spain
| | - Amanda B Fath
- Hospital del Mar Medical Research Institute, Barcelona Biomedical Research Park, 08003 Barcelona, Spain.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Melina Timplalexi
- Hospital del Mar Medical Research Institute, Barcelona Biomedical Research Park, 08003 Barcelona, Spain
| | - M Victoria Puig
- Hospital del Mar Medical Research Institute, Barcelona Biomedical Research Park, 08003 Barcelona, Spain
| |
Collapse
|
114
|
Reversing frontal disinhibition rescues behavioural deficits in models of CACNA1A-associated neurodevelopment disorders. Mol Psychiatry 2021; 26:7225-7246. [PMID: 34127816 DOI: 10.1038/s41380-021-01175-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/27/2021] [Accepted: 05/12/2021] [Indexed: 12/20/2022]
Abstract
CACNA1A deletions cause epilepsy, ataxia, and a range of neurocognitive deficits, including inattention, impulsivity, intellectual deficiency and autism. To investigate the underlying mechanisms, we generated mice carrying a targeted Cacna1a deletion restricted to parvalbumin-expressing (PV) neurons (PVCre;Cacna1ac/+) or to cortical pyramidal cells (PC) (Emx1Cre;Cacna1ac/+). GABA release from PV-expressing GABAergic interneurons (PV-INs) is reduced in PVCre;Cacna1ac/+ mutants, resulting in impulsivity, cognitive rigidity and inattention. By contrast, the deletion of Cacna1a in PCs does not impact cortical excitability or behaviour in Emx1Cre;Cacna1ac/+ mutants. A targeted Cacna1a deletion in the orbitofrontal cortex (OFC) results in reversal learning deficits while a medial prefrontal cortex (mPFC) deletion impairs selective attention. These deficits can be rescued by the selective chemogenetic activation of cortical PV-INs in the OFC or mPFC of PVCre;Cacna1ac/+ mutants. Thus, Cacna1a haploinsufficiency disrupts perisomatic inhibition in frontal cortical circuits, leading to a range of potentially reversible neurocognitive deficits.
Collapse
|
115
|
Hirano Y, Uhlhaas PJ. Current findings and perspectives on aberrant neural oscillations in schizophrenia. Psychiatry Clin Neurosci 2021; 75:358-368. [PMID: 34558155 DOI: 10.1111/pcn.13300] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/20/2021] [Accepted: 09/09/2021] [Indexed: 12/11/2022]
Abstract
There is now consistent evidence that neural oscillation at low- and high-frequencies constitute an important aspect of the pathophysiology of schizophrenia. Specifically, impaired rhythmic activity may underlie the deficit to generate coherent cognition and behavior, leading to the characteristic symptoms of psychosis and cognitive deficits. Importantly, the generating mechanisms of neural oscillations are relatively well-understood and thus enable the targeted search for the underlying circuit impairments and novel treatment targets. In the following review, we will summarize and assess the evidence for aberrant rhythmic activity in schizophrenia through evaluating studies that have utilized Electro/Magnetoencephalography to examine neural oscillations during sensory and cognitive tasks as well as during resting-state measurements. These data will be linked to current evidence from post-mortem, neuroimaging, genetics, and animal models that have implicated deficits in GABAergic interneurons and glutamatergic neurotransmission in oscillatory deficits in schizophrenia. Finally, we will highlight methodological and analytical challenges as well as provide recommendations for future research.
Collapse
Affiliation(s)
- Yoji Hirano
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Institute of Industrial Science, The University of Tokyo, Tokyo, Japan
| | - Peter J Uhlhaas
- Department of Child and Adolescent Psychiatry, Charité - Universitätsmedizin, Berlin, Germany
- Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, UK
| |
Collapse
|
116
|
Stein H, Barbosa J, Compte A. Towards biologically constrained attractor models of schizophrenia. Curr Opin Neurobiol 2021; 70:171-181. [PMID: 34839146 DOI: 10.1016/j.conb.2021.10.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 10/19/2021] [Accepted: 10/27/2021] [Indexed: 12/31/2022]
Abstract
Alterations in neuromodulation or synaptic transmission in biophysical attractor network models, as proposed by the dominant dopaminergic and glutamatergic theories of schizophrenia, successfully mimic working memory (WM) deficits in people with schizophrenia (PSZ). Yet, multiple, often opposing alterations in memory circuits can lead to the same behavioral patterns in these network models. Here, we critically revise the computational and experimental literature that links NMDAR hypofunction to WM precision loss in PSZ. We show in network simulations that currently available experimental evidence cannot set apart competing biophysical accounts. Critical points to resolve are the effects of increases vs. decreases in E/I ratio (e.g. through NMDAR blockade) on firing rate tuning and shared noise modulations and possible concomitant deficits in short-term plasticity. We argue that these concerted experimental and computational efforts will lead to a better understanding of the neurobiology underlying cognitive deficits in PSZ.
Collapse
Affiliation(s)
- Heike Stein
- Laboratoire de Neurosciences Cognitives et Computationnelles, Département d'Études Cognitives, École Normale Supérieure, INSERM U960, PSL University, Paris, France
| | - Joao Barbosa
- Laboratoire de Neurosciences Cognitives et Computationnelles, Département d'Études Cognitives, École Normale Supérieure, INSERM U960, PSL University, Paris, France
| | - Albert Compte
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| |
Collapse
|
117
|
Genetic risk for schizophrenia is associated with altered visually-induced gamma band activity: evidence from a population sample stratified polygenic risk. Transl Psychiatry 2021; 11:592. [PMID: 34785639 PMCID: PMC8595678 DOI: 10.1038/s41398-021-01678-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 09/29/2021] [Accepted: 10/05/2021] [Indexed: 12/12/2022] Open
Abstract
Gamma oscillations (30-90 Hz) have been proposed as a signature of cortical visual information processing, particularly the balance between excitation and inhibition, and as a biomarker of neuropsychiatric diseases. Magnetoencephalography (MEG) provides highly reliable visual-induced gamma oscillation estimates, both at sensor and source level. Recent studies have reported a deficit of visual gamma activity in schizophrenia patients, in medication naive subjects, and high-risk clinical participants, but the genetic contribution to such a deficit has remained unresolved. Here, for the first time, we use a genetic risk score approach to assess the relationship between genetic risk for schizophrenia and visual gamma activity in a population-based sample drawn from a birth cohort. We compared visual gamma activity in a group (N = 104) with a high genetic risk profile score for schizophrenia (SCZ-PRS) to a group with low SCZ-PRS (N = 99). Source-reconstructed V1 activity was extracted using beamformer analysis applied to MEG recordings using individual MRI scans. No group differences were found in the induced gamma peak amplitude or peak frequency. However, a non-parametric statistical contrast of the response spectrum revealed more robust group differences in the amplitude of high-beta/gamma power across the frequency range, suggesting that overall spectral shape carries important biological information beyond the individual frequency peak. Our findings show that changes in gamma band activity correlate with liability to schizophrenia and suggest that the index changes to synaptic function and neuronal firing patterns that are of pathophysiological relevance rather than consequences of the disorder.
Collapse
|
118
|
Carreño-Muñoz MI, Chattopadhyaya B, Agbogba K, Côté V, Wang S, Lévesque M, Avoli M, Michaud JL, Lippé S, Di Cristo G. Sensory processing dysregulations as reliable translational biomarkers in SYNGAP1 haploinsufficiency. Brain 2021; 145:754-769. [PMID: 34791091 DOI: 10.1093/brain/awab329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 08/02/2021] [Accepted: 08/05/2021] [Indexed: 11/13/2022] Open
Abstract
Amongst the numerous genes associated with intellectual disability, SYNGAP1 stands out for its frequency and penetrance of loss-of-function variants found in patients, as well as the wide range of co-morbid disorders associated with its mutation. Most studies exploring the pathophysiological alterations caused by Syngap1 haploinsufficiency in mouse models have focused on cognitive problems and epilepsy, however whether and to what extent sensory perception and processing are altered by Syngap1 haploinsufficiency is less clear. By performing EEG recordings in awake mice, we identified specific alterations in multiple aspects of auditory and visual processing, including increased baseline gamma oscillation power, increased theta/gamma phase amplitude coupling following stimulus presentation and abnormal neural entrainment in response to different sensory modality-specific frequencies. We also report lack of habituation to repetitive auditory stimuli and abnormal deviant sound detection. Interestingly, we found that most of these alterations are present in human patients as well, thus making them strong candidates as translational biomarkers of sensory-processing alterations associated with SYNGAP1/Syngap1 haploinsufficiency.
Collapse
Affiliation(s)
- Maria Isabel Carreño-Muñoz
- Centre de Recherche, CHU Sainte-Justine (CHUSJ), Montreal, Quebec, Canada.,Department of Neurosciences, Université de Montréal, Montreal, Quebec, Canada
| | | | - Kristian Agbogba
- Centre de Recherche, CHU Sainte-Justine (CHUSJ), Montreal, Quebec, Canada
| | - Valérie Côté
- Centre de Recherche, CHU Sainte-Justine (CHUSJ), Montreal, Quebec, Canada.,Department of Psychology, Université de Montréal, Montreal, Quebec, Canada
| | - Siyan Wang
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
| | - Maxime Lévesque
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
| | - Massimo Avoli
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
| | - Jacques L Michaud
- Centre de Recherche, CHU Sainte-Justine (CHUSJ), Montreal, Quebec, Canada.,Department of Neurosciences, Université de Montréal, Montreal, Quebec, Canada.,Department of Pediatrics, Université de Montréal, Montreal, Quebec, Canada
| | - Sarah Lippé
- Centre de Recherche, CHU Sainte-Justine (CHUSJ), Montreal, Quebec, Canada.,Department of Psychology, Université de Montréal, Montreal, Quebec, Canada
| | - Graziella Di Cristo
- Centre de Recherche, CHU Sainte-Justine (CHUSJ), Montreal, Quebec, Canada.,Department of Neurosciences, Université de Montréal, Montreal, Quebec, Canada.,Department of Pediatrics, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
119
|
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: 36] [Impact Index Per Article: 9.0] [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
|
120
|
Metzner C, Steuber V. The beta component of gamma-band auditory steady-state responses in patients with schizophrenia. Sci Rep 2021; 11:20387. [PMID: 34650135 PMCID: PMC8516862 DOI: 10.1038/s41598-021-99793-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 09/24/2021] [Indexed: 01/18/2023] Open
Abstract
The mechanisms underlying circuit dysfunctions in schizophrenia (SCZ) remain poorly understood. Auditory steady-state responses (ASSRs), especially in the gamma and beta band, have been suggested as a potential biomarker for SCZ. While the reduction of 40 Hz power for 40 Hz drive has been well established and replicated in SCZ patients, studies are inconclusive when it comes to an increase in 20 Hz power during 40 Hz drive. There might be several factors explaining the inconsistencies, including differences in the sensitivity of the recording modality (EEG vs MEG), differences in stimuli (click-trains vs amplitude-modulated tones) and large differences in the amplitude of the stimuli. Here, we used a computational model of ASSR deficits in SCZ and explored the effect of three SCZ-associated microcircuit alterations: reduced GABA activity, increased GABA decay times and NMDA receptor hypofunction. We investigated the effect of input strength on gamma (40 Hz) and beta (20 Hz) band power during gamma ASSR stimulation and saw that the pronounced increase in beta power during gamma stimulation seen experimentally could only be reproduced in the model when GABA decay times were increased and only for a specific range of input strengths. More specifically, when the input was in this specific range, the rhythmic drive at 40 Hz produced a strong 40 Hz rhythm in the control network; however, in the 'SCZ-like' network, the prolonged inhibition led to a so-called 'beat-skipping', where the network would only strongly respond to every other input. This mechanism was responsible for the emergence of the pronounced 20 Hz beta peak in the power spectrum. The other two microcircuit alterations were not able to produce a substantial 20 Hz component but they further narrowed the input strength range for which the network produced a beta component when combined with increased GABAergic decay times. Our finding that the beta component only existed for a specific range of input strengths might explain the seemingly inconsistent reporting in experimental studies and suggests that future ASSR studies should systematically explore different amplitudes of their stimuli. Furthermore, we provide a mechanistic link between a microcircuit alteration and an electrophysiological marker in schizophrenia and argue that more complex ASSR stimuli are needed to disentangle the nonlinear interactions of microcircuit alterations. The computational modelling approach put forward here is ideally suited to facilitate the development of such stimuli in a theory-based fashion.
Collapse
Affiliation(s)
- Christoph Metzner
- Neural Information Processing Group, Institute of Software Engineering and Theoretical Computer Science, Technische Universität Berlin, Berlin, Germany.
- School of Physics, Engineering and Computer Science, University of Hertfordshire, Hatfield, UK.
| | - Volker Steuber
- School of Physics, Engineering and Computer Science, University of Hertfordshire, Hatfield, UK
| |
Collapse
|
121
|
Mahadevan V, Mitra A, Zhang Y, Yuan X, Peltekian A, Chittajallu R, Esnault C, Maric D, Rhodes C, Pelkey KA, Dale R, Petros TJ, McBain CJ. NMDARs Drive the Expression of Neuropsychiatric Disorder Risk Genes Within GABAergic Interneuron Subtypes in the Juvenile Brain. Front Mol Neurosci 2021; 14:712609. [PMID: 34630033 PMCID: PMC8500094 DOI: 10.3389/fnmol.2021.712609] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 07/19/2021] [Indexed: 11/13/2022] Open
Abstract
Medial ganglionic eminence (MGE)-derived parvalbumin (PV)+, somatostatin (SST)+and Neurogliaform (NGFC)-type cortical and hippocampal interneurons, have distinct molecular, anatomical, and physiological properties. However, the molecular mechanisms regulating their maturation remain poorly understood. Here, via single-cell transcriptomics, we show that the obligate NMDA-type glutamate receptor (NMDAR) subunit gene Grin1 mediates transcriptional regulation of gene expression in specific subtypes of MGE-derived interneurons, leading to altered subtype abundances. Notably, MGE-specific early developmental Grin1 loss results in a broad downregulation of diverse transcriptional, synaptogenic and membrane excitability regulatory programs in the juvenile brain. These widespread gene expression abnormalities mirror aberrations that are typically associated with neurodevelopmental disorders. Our study hence provides a road map for the systematic examination of NMDAR signaling in interneuron subtypes, revealing potential MGE-specific genetic targets that could instruct future therapies of psychiatric disorders.
Collapse
Affiliation(s)
- Vivek Mahadevan
- Section on Cellular and Synaptic Physiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Bethesda, MD, United States
| | - Apratim Mitra
- Bioinformatics and Scientific Programming Core, NICHD, Bethesda, MD, United States
| | - Yajun Zhang
- Unit on Cellular and Molecular Neurodevelopment, NICHD, Bethesda, MD, United States
| | - Xiaoqing Yuan
- Section on Cellular and Synaptic Physiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Bethesda, MD, United States
| | - Areg Peltekian
- Section on Cellular and Synaptic Physiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Bethesda, MD, United States
| | - Ramesh Chittajallu
- Section on Cellular and Synaptic Physiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Bethesda, MD, United States
| | - Caroline Esnault
- Bioinformatics and Scientific Programming Core, NICHD, Bethesda, MD, United States
| | - Dragan Maric
- Flow and Imaging Cytometry Core Facility, National Institute of Neurological Disorders and Stroke (NINDS), Bethesda, MD, United States
| | - Christopher Rhodes
- Unit on Cellular and Molecular Neurodevelopment, NICHD, Bethesda, MD, United States
| | - Kenneth A Pelkey
- Section on Cellular and Synaptic Physiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Bethesda, MD, United States
| | - Ryan Dale
- Bioinformatics and Scientific Programming Core, NICHD, Bethesda, MD, United States
| | - Timothy J Petros
- Unit on Cellular and Molecular Neurodevelopment, NICHD, Bethesda, MD, United States
| | - Chris J McBain
- Section on Cellular and Synaptic Physiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Bethesda, MD, United States
| |
Collapse
|
122
|
Speers LJ, Bilkey DK. Disorganization of Oscillatory Activity in Animal Models of Schizophrenia. Front Neural Circuits 2021; 15:741767. [PMID: 34675780 PMCID: PMC8523827 DOI: 10.3389/fncir.2021.741767] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/16/2021] [Indexed: 01/02/2023] Open
Abstract
Schizophrenia is a chronic, debilitating disorder with diverse symptomatology, including disorganized cognition and behavior. Despite considerable research effort, we have only a limited understanding of the underlying brain dysfunction. In this article, we review the potential role of oscillatory circuits in the disorder with a particular focus on the hippocampus, a region that encodes sequential information across time and space, as well as the frontal cortex. Several mechanistic explanations of schizophrenia propose that a loss of oscillatory synchrony between and within these brain regions may underlie some of the symptoms of the disorder. We describe how these oscillations are affected in several animal models of schizophrenia, including models of genetic risk, maternal immune activation (MIA) models, and models of NMDA receptor hypofunction. We then critically discuss the evidence for disorganized oscillatory activity in these models, with a focus on gamma, sharp wave ripple, and theta activity, including the role of cross-frequency coupling as a synchronizing mechanism. Finally, we focus on phase precession, which is an oscillatory phenomenon whereby individual hippocampal place cells systematically advance their firing phase against the background theta oscillation. Phase precession is important because it allows sequential experience to be compressed into a single 120 ms theta cycle (known as a 'theta sequence'). This time window is appropriate for the induction of synaptic plasticity. We describe how disruption of phase precession could disorganize sequential processing, and thereby disrupt the ordered storage of information. A similar dysfunction in schizophrenia may contribute to cognitive symptoms, including deficits in episodic memory, working memory, and future planning.
Collapse
Affiliation(s)
| | - David K. Bilkey
- Department of Psychology, Otago University, Dunedin, New Zealand
| |
Collapse
|
123
|
Van Derveer AB, Bastos G, Ferrell AD, Gallimore CG, Greene ML, Holmes JT, Kubricka V, Ross JM, Hamm JP. A Role for Somatostatin-Positive Interneurons in Neuro-Oscillatory and Information Processing Deficits in Schizophrenia. Schizophr Bull 2021; 47:1385-1398. [PMID: 33370434 PMCID: PMC8379548 DOI: 10.1093/schbul/sbaa184] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Alterations in neocortical GABAergic interneurons (INs) have been affiliated with neuropsychiatric diseases, including schizophrenia (SZ). Significant progress has been made linking the function of a specific subtype of GABAergic cells, parvalbumin (PV) positive INs, to altered gamma-band oscillations, which, in turn, underlie perceptual and feedforward information processing in cortical circuits. Here, we review a smaller but growing volume of literature focusing on a separate subtype of neocortical GABAergic INs, somatostatin (SST) positive INs. Despite sharing similar neurodevelopmental origins, SSTs exhibit distinct morphology and physiology from PVs. Like PVs, SSTs are altered in postmortem brain samples from multiple neocortical regions in SZ, although basic and translational research into consequences of SST dysfunction has been relatively sparse. We highlight a growing body of work in rodents, which now indicates that SSTs may also underlie specific aspects of cortical circuit function, namely low-frequency oscillations, disinhibition, and mediation of cortico-cortical feedback. SSTs may thereby support the coordination of local cortical information processing with more global spatial, temporal, and behavioral context, including predictive coding and working memory. These functions are notably deficient in some cases of SZ, as well as other neuropsychiatric disorders, emphasizing the importance of focusing on SSTs in future translational studies. Finally, we highlight the challenges that remain, including subtypes within the SST class.
Collapse
Affiliation(s)
- Alice B Van Derveer
- Neuroscience Institute, Georgia State University, Petit Science Center, Atlanta, GA
| | - Georgia Bastos
- Neuroscience Institute, Georgia State University, Petit Science Center, Atlanta, GA
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Petit Science Center, Atlanta, GA
| | - Antanovia D Ferrell
- Neuroscience Institute, Georgia State University, Petit Science Center, Atlanta, GA
| | - Connor G Gallimore
- Neuroscience Institute, Georgia State University, Petit Science Center, Atlanta, GA
| | - Michelle L Greene
- Neuroscience Institute, Georgia State University, Petit Science Center, Atlanta, GA
| | - Jacob T Holmes
- Neuroscience Institute, Georgia State University, Petit Science Center, Atlanta, GA
| | - Vivien Kubricka
- Neuroscience Institute, Georgia State University, Petit Science Center, Atlanta, GA
| | - Jordan M Ross
- Neuroscience Institute, Georgia State University, Petit Science Center, Atlanta, GA
- Center for Behavioral Neuroscience, Georgia State University, Petit Science Center, Atlanta, GA
| | - Jordan P Hamm
- Neuroscience Institute, Georgia State University, Petit Science Center, Atlanta, GA
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Petit Science Center, Atlanta, GA
- Center for Behavioral Neuroscience, Georgia State University, Petit Science Center, Atlanta, GA
| |
Collapse
|
124
|
Perineuronal Nets and Metal Cation Concentrations in the Microenvironments of Fast-Spiking, Parvalbumin-Expressing GABAergic Interneurons: Relevance to Neurodevelopment and Neurodevelopmental Disorders. Biomolecules 2021; 11:biom11081235. [PMID: 34439901 PMCID: PMC8391699 DOI: 10.3390/biom11081235] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/11/2021] [Accepted: 08/14/2021] [Indexed: 12/31/2022] Open
Abstract
Because of their abilities to catalyze generation of toxic free radical species, free concentrations of the redox reactive metals iron and copper are highly regulated. Importantly, desired neurobiological effects of these redox reactive metal cations occur within very narrow ranges of their local concentrations. For example, synaptic release of free copper acts locally to modulate NMDA receptor-mediated neurotransmission. Moreover, within the developing brain, iron is critical to hippocampal maturation and the differentiation of parvalbumin-expressing neurons, whose soma and dendrites are surrounded by perineuronal nets (PNNs). The PNNs are a specialized component of brain extracellular matrix, whose polyanionic character supports the fast-spiking electrophysiological properties of these parvalbumin-expressing GABAergic interneurons. In addition to binding cations and creation of the Donnan equilibrium that support the fast-spiking properties of this subset of interneurons, the complex architecture of PNNs also binds metal cations, which may serve a protective function against oxidative damage, especially of these fast-spiking neurons. Data suggest that pathological disturbance of the population of fast-spiking, parvalbumin-expressing GABAergic inhibitory interneurons occur in at least some clinical presentations, which leads to disruption of the synchronous oscillatory output of assemblies of pyramidal neurons. Increased expression of the GluN2A NMDA receptor subunit on parvalbumin-expressing interneurons is linked to functional maturation of both these neurons and the perineuronal nets that surround them. Disruption of GluN2A expression shows increased susceptibility to oxidative stress, reflected in redox dysregulation and delayed maturation of PNNs. This may be especially relevant to neurodevelopmental disorders, including autism spectrum disorder. Conceivably, binding of metal redox reactive cations by the perineuronal net helps to maintain safe local concentrations, and also serves as a reservoir buffering against second-to-second fluctuations in their concentrations outside of a narrow physiological range.
Collapse
|
125
|
Miller DS, Wright KM. Neuronal Dystroglycan regulates postnatal development of CCK/cannabinoid receptor-1 interneurons. Neural Dev 2021; 16:4. [PMID: 34362433 PMCID: PMC8349015 DOI: 10.1186/s13064-021-00153-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 05/20/2021] [Indexed: 12/02/2022] Open
Abstract
Background The development of functional neural circuits requires the precise formation of synaptic connections between diverse neuronal populations. The molecular pathways that allow GABAergic interneuron subtypes in the mammalian brain to initially recognize their postsynaptic partners remain largely unknown. The transmembrane glycoprotein Dystroglycan is localized to inhibitory synapses in pyramidal neurons, where it is required for the proper function of CCK+ interneurons. However, the precise temporal requirement for Dystroglycan during inhibitory synapse development has not been examined. Methods In this study, we use NEXCre or Camk2aCreERT2 to conditionally delete Dystroglycan from newly-born or adult pyramidal neurons, respectively. We then analyze forebrain development from postnatal day 3 through adulthood, with a particular focus on CCK+ interneurons. Results In the absence of postsynaptic Dystroglycan in developing pyramidal neurons, presynaptic CCK+ interneurons fail to elaborate their axons and largely disappear from the cortex, hippocampus, amygdala, and olfactory bulb during the first two postnatal weeks. Other interneuron subtypes are unaffected, indicating that CCK+ interneurons are unique in their requirement for postsynaptic Dystroglycan. Dystroglycan does not appear to be required in adult pyramidal neurons to maintain CCK+ interneurons. Bax deletion did not rescue CCK+ interneurons in Dystroglycan mutants during development, suggesting that they are not eliminated by canonical apoptosis. Rather, we observed increased innervation of the striatum, suggesting that the few remaining CCK+ interneurons re-directed their axons to neighboring areas where Dystroglycan expression remained intact. Conclusion Together these findings show that Dystroglycan functions as part of a synaptic partner recognition complex that is required early for CCK+ interneuron development in the forebrain. Supplementary Information The online version contains supplementary material available at 10.1186/s13064-021-00153-1.
Collapse
Affiliation(s)
- Daniel S Miller
- Neuroscience Graduate Program, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Kevin M Wright
- Vollum Institute, Oregon Health & Science University, VIB 3435A, 3181 SW Sam Jackson Park Road, L474, Portland, OR, 97239-3098, USA.
| |
Collapse
|
126
|
Kadriu B, Musazzi L, Johnston JN, Kalynchuk LE, Caruncho HJ, Popoli M, Zarate CA. Positive AMPA receptor modulation in the treatment of neuropsychiatric disorders: A long and winding road. Drug Discov Today 2021; 26:2816-2838. [PMID: 34358693 DOI: 10.1016/j.drudis.2021.07.027] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/12/2021] [Accepted: 07/28/2021] [Indexed: 12/13/2022]
Abstract
Glutamatergic transmission is widely implicated in neuropsychiatric disorders, and the discovery that ketamine elicits rapid-acting antidepressant effects by modulating α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor (AMPAR) signaling has spurred a resurgence of interest in the field. This review explores agents in various stages of development for neuropsychiatric disorders that positively modulate AMPARs, both directly and indirectly. Despite promising preclinical research, few direct and indirect AMPAR positive modulators have progressed past early clinical development. Challenges such as low potency have created barriers to effective implementation. Nevertheless, the functional complexity of AMPARs sets them apart from other drug targets and allows for specificity in drug discovery. Additional effective treatments for neuropsychiatric disorders that work through positive AMPAR modulation may eventually be developed.
Collapse
Affiliation(s)
- Bashkim Kadriu
- Experimental Therapeutics & Pathophysiology Branch, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| | - Laura Musazzi
- School of Medicine and Surgery, University of Milano-Bicocca, Italy
| | - Jenessa N Johnston
- Experimental Therapeutics & Pathophysiology Branch, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA; Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Lisa E Kalynchuk
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Hector J Caruncho
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Maurizio Popoli
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Italy
| | - Carlos A Zarate
- Experimental Therapeutics & Pathophysiology Branch, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
127
|
Hayden DJ, Montgomery DP, Cooke SF, Bear MF. Visual Recognition Is Heralded by Shifts in Local Field Potential Oscillations and Inhibitory Networks in Primary Visual Cortex. J Neurosci 2021; 41:6257-6272. [PMID: 34103358 PMCID: PMC8287992 DOI: 10.1523/jneurosci.0391-21.2021] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 11/22/2022] Open
Abstract
Learning to recognize and filter familiar, irrelevant sensory stimuli eases the computational burden on the cerebral cortex. Inhibition is a candidate mechanism in this filtration process, and oscillations in the cortical local field potential (LFP) serve as markers of the engagement of different inhibitory neurons. We show here that LFP oscillatory activity in visual cortex is profoundly altered as male and female mice learn to recognize an oriented grating stimulus-low-frequency (∼15 Hz peak) power sharply increases, whereas high-frequency (∼65 Hz peak) power decreases. These changes report recognition of the familiar pattern as they disappear when the stimulus is rotated to a novel orientation. Two-photon imaging of neuronal activity reveals that parvalbumin-expressing inhibitory neurons disengage with familiar stimuli and reactivate to novelty, whereas somatostatin-expressing inhibitory neurons show opposing activity patterns. We propose a model in which the balance of two interacting interneuron circuits shifts as novel stimuli become familiar.SIGNIFICANCE STATEMENT Habituation, familiarity, and novelty detection are fundamental cognitive processes that enable organisms to adaptively filter meaningless stimuli and focus attention on potentially important elements of their environment. We have shown that this process can be studied fruitfully in the mouse primary visual cortex by using simple grating stimuli for which novelty and familiarity are defined by orientation and by measuring stimulus-evoked and continuous local field potentials. Altered event-related and spontaneous potentials, and deficient habituation, are well-documented features of several neurodevelopmental psychiatric disorders. The paradigm described here will be valuable to interrogate the origins of these signals and the meaning of their disruption more deeply.
Collapse
Affiliation(s)
- Dustin J Hayden
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Daniel P Montgomery
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Samuel F Cooke
- Medical Research Council Centre for Neurodevelopmental Disorders, Department of Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London SE5 9RT, England
| | - Mark F Bear
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| |
Collapse
|
128
|
McQuail JA, Beas BS, Kelly KB, Hernandez CM, Bizon JL, Frazier CJ. Attenuated NMDAR signaling on fast-spiking interneurons in prefrontal cortex contributes to age-related decline of cognitive flexibility. Neuropharmacology 2021; 197:108720. [PMID: 34273386 DOI: 10.1016/j.neuropharm.2021.108720] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 07/08/2021] [Accepted: 07/13/2021] [Indexed: 02/01/2023]
Abstract
Ionotropic glutamate receptors of the NMDA and AMPA subtypes transduce excitatory signaling on neurons in the prefrontal cortex (PFC) in support of cognitive flexibility. Cognitive flexibility is reliably observed to decline at advanced ages, coinciding with changes in PFC glutamate receptor expression and neuronal physiology. However, the relationship between age-related impairment of cognitive flexibility and changes to excitatory signaling on distinct classes of PFC neurons is not known. In this study, one cohort of young adult (4 months) and aged (20 months) male F344 rats were characterized for cognitive flexibility on an operant set-shifting task. Expression of the essential NMDAR subunit, NR1, was correlated with individual differences in set-shifting abilities such that lower NR1 in the aged PFC was associated with worse set-shifting. In contrast, lower expression of two AMPAR subunits, GluR1 and GluR2, was not associated with set-shift abilities in aging. As NMDARs are expressed by both pyramidal cells and fast-spiking interneurons (FSI) in PFC, whole-cell patch clamp recordings were performed in a second cohort of age-matched rats to compare age-associated changes on these neuronal subtypes. Evoked excitatory postsynaptic currents were generated using a bipolar stimulator while AMPAR vs. NMDAR-mediated components were isolated using pharmacological tools. The results revealed a clear increase in AMPA/NMDA ratio in FSIs that was not present in pyramidal neurons. Together, these data indicate that loss of NMDARs on interneurons in PFC contributes to age-related impairment of cognitive flexibility.
Collapse
Affiliation(s)
- Joseph A McQuail
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, 29208, USA.
| | - B Sofia Beas
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, 32610, USA; Unit on the Neurobiology of Affective Memory, National Institute of Mental Health, Bethesda, MD, 20892, USA
| | - Kyle B Kelly
- Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, FL, 32610, USA
| | - Caesar M Hernandez
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, 32610, USA; Department of Cellular, Development, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Jennifer L Bizon
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, 32610, USA
| | - Charles J Frazier
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, 32610, USA; Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, FL, 32610, USA.
| |
Collapse
|
129
|
McNally JM, Aguilar DD, Katsuki F, Radzik LK, Schiffino FL, Uygun DS, McKenna JT, Strecker RE, Deisseroth K, Spencer KM, Brown RE. Optogenetic manipulation of an ascending arousal system tunes cortical broadband gamma power and reveals functional deficits relevant to schizophrenia. Mol Psychiatry 2021; 26:3461-3475. [PMID: 32690865 PMCID: PMC7855059 DOI: 10.1038/s41380-020-0840-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 06/30/2020] [Accepted: 07/07/2020] [Indexed: 01/06/2023]
Abstract
Increases in broadband cortical electroencephalogram (EEG) power in the gamma band (30-80 Hz) range have been observed in schizophrenia patients and in mouse models of schizophrenia. They are also seen in humans and animals treated with the psychotomimetic agent ketamine. However, the mechanisms which can result in increased broadband gamma power and the pathophysiological implications for cognition and behavior are poorly understood. Here we report that tonic optogenetic manipulation of an ascending arousal system bidirectionally tunes cortical broadband gamma power, allowing on-demand tests of the effect on cortical processing and behavior. Constant, low wattage optogenetic stimulation of basal forebrain (BF) neurons containing the calcium-binding protein parvalbumin (PV) increased broadband gamma frequency power, increased locomotor activity, and impaired novel object recognition. Concomitantly, task-associated gamma band oscillations induced by trains of auditory stimuli, or exposure to novel objects, were impaired, reminiscent of findings in schizophrenia patients. Conversely, tonic optogenetic inhibition of BF-PV neurons partially rescued the elevated broadband gamma power elicited by subanesthetic doses of ketamine. These results support the idea that increased cortical broadband gamma activity leads to impairments in cognition and behavior, and identify BF-PV activity as a modulator of this activity. As such, BF-PV neurons may represent a novel target for pharmacotherapy in disorders such as schizophrenia which involve aberrant increases in cortical broadband gamma activity.
Collapse
Affiliation(s)
- James M McNally
- Department of Psychiatry, VA Boston Healthcare System and Harvard Medical School, West Roxbury, Boston, MA, USA.
| | - David D Aguilar
- Department of Psychiatry, VA Boston Healthcare System and Harvard Medical School, West Roxbury, Boston, MA, USA
| | - Fumi Katsuki
- Department of Psychiatry, VA Boston Healthcare System and Harvard Medical School, West Roxbury, Boston, MA, USA
| | - Leana K Radzik
- Department of Neuroscience, Stonehill College, Easton, MA, USA
| | - Felipe L Schiffino
- Department of Psychiatry, VA Boston Healthcare System and Harvard Medical School, West Roxbury, Boston, MA, USA
| | - David S Uygun
- Department of Psychiatry, VA Boston Healthcare System and Harvard Medical School, West Roxbury, Boston, MA, USA
| | - James T McKenna
- Department of Psychiatry, VA Boston Healthcare System and Harvard Medical School, West Roxbury, Boston, MA, USA
| | - Robert E Strecker
- Department of Psychiatry, VA Boston Healthcare System and Harvard Medical School, West Roxbury, Boston, MA, USA
| | - Karl Deisseroth
- Psychiatry and Behavioral Sciences/Bioengineering, Stanford University, Stanford, CA, USA
| | - Kevin M Spencer
- Department of Psychiatry, VA Boston Healthcare System and Harvard Medical School, Jamaica Plain, Boston, MA, USA
| | - Ritchie E Brown
- Department of Psychiatry, VA Boston Healthcare System and Harvard Medical School, West Roxbury, Boston, MA, USA
| |
Collapse
|
130
|
Tan LL, Oswald MJ, Kuner R. Neurobiology of brain oscillations in acute and chronic pain. Trends Neurosci 2021; 44:629-642. [PMID: 34176645 DOI: 10.1016/j.tins.2021.05.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 03/19/2021] [Accepted: 05/07/2021] [Indexed: 01/08/2023]
Abstract
Pain is a complex perceptual phenomenon. Coordinated activity among local and distant brain networks is a central element of the neural underpinnings of pain. Brain oscillatory rhythms across diverse frequency ranges provide a functional substrate for coordinating activity across local neuronal ensembles and anatomically distant brain areas in pain networks. This review addresses parallels between insights from human and rodent analyses of oscillatory rhythms in acute and chronic pain and discusses recent rodent-based studies that have shed light on mechanistic underpinnings of brain oscillatory dynamics in pain-related behaviors. We highlight the potential for therapeutic modulation of oscillatory rhythms, and identify outstanding questions and challenges to be addressed in future research.
Collapse
Affiliation(s)
- Linette Liqi Tan
- Institute of Pharmacology, Heidelberg University, Im Neuenheimer Feld 366, D-69120 Heidelberg, Germany.
| | - Manfred Josef Oswald
- Institute of Pharmacology, Heidelberg University, Im Neuenheimer Feld 366, D-69120 Heidelberg, Germany
| | - Rohini Kuner
- Institute of Pharmacology, Heidelberg University, Im Neuenheimer Feld 366, D-69120 Heidelberg, Germany.
| |
Collapse
|
131
|
Patrono E, Svoboda J, Stuchlík A. Schizophrenia, the gut microbiota, and new opportunities from optogenetic manipulations of the gut-brain axis. Behav Brain Funct 2021; 17:7. [PMID: 34158061 PMCID: PMC8218443 DOI: 10.1186/s12993-021-00180-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 06/01/2021] [Indexed: 12/18/2022] Open
Abstract
Schizophrenia research arose in the twentieth century and is currently rapidly developing, focusing on many parallel research pathways and evaluating various concepts of disease etiology. Today, we have relatively good knowledge about the generation of positive and negative symptoms in patients with schizophrenia. However, the neural basis and pathophysiology of schizophrenia, especially cognitive symptoms, are still poorly understood. Finding new methods to uncover the physiological basis of the mental inabilities related to schizophrenia is an urgent task for modern neuroscience because of the lack of specific therapies for cognitive deficits in the disease. Researchers have begun investigating functional crosstalk between NMDARs and GABAergic neurons associated with schizophrenia at different resolutions. In another direction, the gut microbiota is getting increasing interest from neuroscientists. Recent findings have highlighted the role of a gut-brain axis, with the gut microbiota playing a crucial role in several psychopathologies, including schizophrenia and autism. There have also been investigations into potential therapies aimed at normalizing altered microbiota signaling to the enteric nervous system (ENS) and the central nervous system (CNS). Probiotics diets and fecal microbiota transplantation (FMT) are currently the most common therapies. Interestingly, in rodent models of binge feeding, optogenetic applications have been shown to affect gut colony sensitivity, thus increasing colonic transit. Here, we review recent findings on the gut microbiota–schizophrenia relationship using in vivo optogenetics. Moreover, we evaluate if manipulating actors in either the brain or the gut might improve potential treatment research. Such research and techniques will increase our knowledge of how the gut microbiota can manipulate GABA production, and therefore accompany changes in CNS GABAergic activity.
Collapse
Affiliation(s)
- Enrico Patrono
- Institute of Physiology of the Czech Academy of Sciences, Videnska, 1830, Prague, 142 20, Czech Republic.
| | - Jan Svoboda
- Institute of Physiology of the Czech Academy of Sciences, Videnska, 1830, Prague, 142 20, Czech Republic
| | - Aleš Stuchlík
- Institute of Physiology of the Czech Academy of Sciences, Videnska, 1830, Prague, 142 20, Czech Republic.
| |
Collapse
|
132
|
Plataki ME, Diskos K, Sougklakos C, Velissariou M, Georgilis A, Stavroulaki V, Sidiropoulou K. Effect of Neonatal Treatment With the NMDA Receptor Antagonist, MK-801, During Different Temporal Windows of Postnatal Period in Adult Prefrontal Cortical and Hippocampal Function. Front Behav Neurosci 2021; 15:689193. [PMID: 34177484 PMCID: PMC8230549 DOI: 10.3389/fnbeh.2021.689193] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/10/2021] [Indexed: 11/13/2022] Open
Abstract
The neonatal MK-801 model of schizophrenia has been developed based on the neurodevelopmental and NMDA receptor hypofunction hypotheses of schizophrenia. This animal model is generated with the use of the NMDA receptor antagonist, MK-801, during different temporal windows of postnatal life of rodents leading to behavioral defects in adulthood. However, no studies have examined the role of specific postnatal time periods in the neonatal MK-801 (nMK-801) rodent model and the resulting behavioral and neurobiological effects. Thus, the goal of this study is to systematically investigate the role of NMDA hypofunction, during specific temporal windows in postnatal life on different cognitive and social behavioral paradigms, as well as various neurobiological effects during adulthood. Both female and male mice were injected intraperitoneally (i.p.) with MK-801 during postnatal days 7-14 (p7-14) or 11-15 (p11-15). Control mice were injected with saline during the respective time period. In adulthood, mice were tested in various cognitive and social behavioral tasks. Mice nMK-801-treated on p7-14 show impaired performance in the novel object, object-to-place, and temporal order object recognition (TOR) tasks, the sociability test, and contextual fear extinction. Mice nMK-801-treated on p11-15 only affects performance in the TOR task, the social memory test, and contextual fear extinction. No differences were identified in the expression of NMDA receptor subunits, the synapsin or PSD-95 proteins, either in the prefrontal cortex (PFC) or the hippocampus (HPC), brain regions significantly affected in schizophrenia. The number of parvalbumin (PV)-expressing cells is significantly reduced in the PFC, but not in the HPC, of nMK-801-treated mice on p7-14 compared to their controls. No differences in PV-expressing cells (PFC or HPC) were identified in nMK-801-treated mice on p11-15. We further examined PFC function by recording spontaneous activity in a solution that allows up state generation. We find that the frequency of up states is significantly reduced in both nMK-801-treated mice on p7-14 and p11-15 compared to saline-treated mice. Furthermore, we find adaptations in the gamma and high gamma activity in nMK-801-treated mice. In conclusion, our results show that MK-801 treatment during specific postnatal temporal windows has differential effects on cognitive and social behaviors, as well as on underlying neurobiological substrates.
Collapse
Affiliation(s)
- Maria E Plataki
- Department of Biology, University of Crete, Heraklion, Greece.,Institute of Molecular Biology and Biotechnology-Foundation for Research and Technology Hellas, Heraklion, Greece
| | - Konstantinos Diskos
- Department of Biology, University of Crete, Heraklion, Greece.,Institute of Molecular Biology and Biotechnology-Foundation for Research and Technology Hellas, Heraklion, Greece
| | | | | | | | | | - Kyriaki Sidiropoulou
- Department of Biology, University of Crete, Heraklion, Greece.,Institute of Molecular Biology and Biotechnology-Foundation for Research and Technology Hellas, Heraklion, Greece
| |
Collapse
|
133
|
Lovelace JW, Rais M, Palacios AR, Shuai XS, Bishay S, Popa O, Pirbhoy PS, Binder DK, Nelson DL, Ethell IM, Razak KA. Deletion of Fmr1 from Forebrain Excitatory Neurons Triggers Abnormal Cellular, EEG, and Behavioral Phenotypes in the Auditory Cortex of a Mouse Model of Fragile X Syndrome. Cereb Cortex 2021; 30:969-988. [PMID: 31364704 DOI: 10.1093/cercor/bhz141] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 05/08/2019] [Accepted: 05/29/2019] [Indexed: 12/13/2022] Open
Abstract
Fragile X syndrome (FXS) is a leading genetic cause of autism with symptoms that include sensory processing deficits. In both humans with FXS and a mouse model [Fmr1 knockout (KO) mouse], electroencephalographic (EEG) recordings show enhanced resting state gamma power and reduced sound-evoked gamma synchrony. We previously showed that elevated levels of matrix metalloproteinase-9 (MMP-9) may contribute to these phenotypes by affecting perineuronal nets (PNNs) around parvalbumin (PV) interneurons in the auditory cortex of Fmr1 KO mice. However, how different cell types within local cortical circuits contribute to these deficits is not known. Here, we examined whether Fmr1 deletion in forebrain excitatory neurons affects neural oscillations, MMP-9 activity, and PV/PNN expression in the auditory cortex. We found that cortical MMP-9 gelatinase activity, mTOR/Akt phosphorylation, and resting EEG gamma power were enhanced in CreNex1/Fmr1Flox/y conditional KO (cKO) mice, whereas the density of PV/PNN cells was reduced. The CreNex1/Fmr1Flox/y cKO mice also show increased locomotor activity, but not the anxiety-like behaviors. These results indicate that fragile X mental retardation protein changes in excitatory neurons in the cortex are sufficient to elicit cellular, electrophysiological, and behavioral phenotypes in Fmr1 KO mice. More broadly, these results indicate that local cortical circuit abnormalities contribute to sensory processing deficits in autism spectrum disorders.
Collapse
Affiliation(s)
| | - Maham Rais
- Division of Biomedical Sciences, School of Medicine
| | | | | | | | - Otilia Popa
- Division of Biomedical Sciences, School of Medicine
| | | | - Devin K Binder
- Division of Biomedical Sciences, School of Medicine.,Graduate Neuroscience Program, University of California Riverside, Riverside, CA 92521,USA
| | - David L Nelson
- Molecular and Human Genetics, Baylor College of Medicine , Houston, TX 77030, USA
| | - Iryna M Ethell
- Division of Biomedical Sciences, School of Medicine.,Graduate Neuroscience Program, University of California Riverside, Riverside, CA 92521,USA
| | - Khaleel A Razak
- Department of Psychology.,Graduate Neuroscience Program, University of California Riverside, Riverside, CA 92521,USA
| |
Collapse
|
134
|
Jami SA, Cameron S, Wong JM, Daly ER, McAllister AK, Gray JA. Increased excitation-inhibition balance and loss of GABAergic synapses in the serine racemase knockout model of NMDA receptor hypofunction. J Neurophysiol 2021; 126:11-27. [PMID: 34038186 DOI: 10.1152/jn.00661.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
There is substantial evidence that both N-methyl-D-aspartate receptor (NMDAR) hypofunction and dysfunction of GABAergic neurotransmission contribute to schizophrenia, though the relationship between these pathophysiological processes remains largely unknown. Although models using cell-type-specific genetic deletion of NMDARs have been informative, they display overly pronounced phenotypes extending beyond those of schizophrenia. Here, we used the serine racemase knockout (SRKO) mice, a model of reduced NMDAR activity rather than complete receptor elimination, to examine the link between NMDAR hypofunction and decreased GABAergic inhibition. The SRKO mice, in which there is a >90% reduction in the NMDAR coagonist d-serine, exhibit many of the neurochemical and behavioral abnormalities observed in schizophrenia. We found a significant reduction in inhibitory synapses onto CA1 pyramidal neurons in the SRKO mice. This reduction increases the excitation/inhibition balance resulting in enhanced synaptically driven neuronal excitability without changes in intrinsic excitability. Consistently, significant reductions in inhibitory synapse density in CA1 were observed by immunohistochemistry. We further show, using a single-neuron genetic deletion approach, that the loss of GABAergic synapses onto pyramidal neurons observed in the SRKO mice is driven in a cell-autonomous manner following the deletion of SR in individual CA1 pyramidal cells. These results support a model whereby NMDAR hypofunction in pyramidal cells disrupts GABAergic synapses leading to disrupted feedback inhibition and impaired neuronal synchrony.NEW & NOTEWORTHY Recently, disruption of excitation/inhibition (E/I) balance has become an area of considerable interest for psychiatric research. Here, we report a reduction in inhibition in the serine racemase knockout mouse model of schizophrenia that increases E/I balance and enhances synaptically driven neuronal excitability. This reduced inhibition was driven cell-autonomously in pyramidal cells lacking serine racemase, suggesting a novel mechanism for how chronic NMDA receptor hypofunction can disrupt information processing in schizophrenia.
Collapse
Affiliation(s)
- Shekib A Jami
- Center for Neuroscience, University of California, Davis, California
| | - Scott Cameron
- Center for Neuroscience, University of California, Davis, California
| | - Jonathan M Wong
- Center for Neuroscience, University of California, Davis, California
| | - Emily R Daly
- Center for Neuroscience, University of California, Davis, California
| | - A Kimberley McAllister
- Center for Neuroscience, University of California, Davis, California.,Department of Neurobiology, Physiology, and Behavior, University of California, Davis, California.,Department of Neurology, University of California, Davis, California
| | - John A Gray
- Center for Neuroscience, University of California, Davis, California.,Department of Neurology, University of California, Davis, California
| |
Collapse
|
135
|
Imbriglio T, Verhaeghe R, Antenucci N, Maccari S, Battaglia G, Nicoletti F, Cannella M. Developmental up-regulation of NMDA receptors in the prefrontal cortex and hippocampus of mGlu5 receptor knock-out mice. Mol Brain 2021; 14:77. [PMID: 33962661 PMCID: PMC8106212 DOI: 10.1186/s13041-021-00784-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 04/21/2021] [Indexed: 12/02/2022] Open
Abstract
mGlu5 metabotropic glutamate receptors are highly expressed and functional in the early postnatal life, and are known to positively modulate NMDA receptor function. Here, we examined the expression of NMDA receptor subunits and interneuron-related genes in the prefrontal cortex and hippocampus of mGlu5-/- mice and wild-type littermates at three developmental time points (PND9, - 21, and - 75). We were surprised to find that expression of all NMDA receptor subunits was greatly enhanced in mGlu5-/- mice at PND21. In contrast, at PND9, expression of the GluN2B subunit was enhanced, whereas expression of GluN2A and GluN2D subunits was reduced in both regions. These modifications were transient and disappeared in the adult life (PND75). Changes in the transcripts of interneuron-related genes (encoding parvalbumin, somatostatin, vasoactive intestinal peptide, reelin, and the two isoforms of glutamate decarboxylase) were also observed in mGlu5-/- mice across postnatal development. For example, the transcript encoding parvalbumin was up-regulated in the prefrontal cortex of mGlu5-/- mice at PND9 and PND21, whereas it was significantly reduced at PND75. These findings suggest that in mGlu5-/- mice a transient overexpression of NMDA receptor subunits may compensate for the lack of the NMDA receptor partner, mGlu5. Interestingly, in mGlu5-/- mice the behavioral response to the NMDA channel blocker, MK-801, was significantly increased at PND21, and largely reduced at PND75. The impact of adaptive changes in the expression of NMDA receptor subunits should be taken into account when mGlu5-/- mice are used for developmental studies.
Collapse
Affiliation(s)
| | | | - Nico Antenucci
- Department of Physiology and Pharmacology "V. Erspamer", University Sapienza of Rome, Piazzale Aldo Moro, 5, 00185, Rome, Italy
| | - Stefania Maccari
- Department of Physiology and Pharmacology "V. Erspamer", University Sapienza of Rome, Piazzale Aldo Moro, 5, 00185, Rome, Italy
- CNRS, UMR 8576, UGSF, Unité de Glycobiologie Structurale et Fonctionnelle, University of Lille, Lille, France
| | - Giuseppe Battaglia
- IRCCS Neuromed, Pozzilli, IS, Italy
- Department of Physiology and Pharmacology "V. Erspamer", University Sapienza of Rome, Piazzale Aldo Moro, 5, 00185, Rome, Italy
| | - Ferdinando Nicoletti
- IRCCS Neuromed, Pozzilli, IS, Italy.
- Department of Physiology and Pharmacology "V. Erspamer", University Sapienza of Rome, Piazzale Aldo Moro, 5, 00185, Rome, Italy.
| | | |
Collapse
|
136
|
Koshiyama D, Miyakoshi M, Joshi YB, Nakanishi M, Tanaka-Koshiyama K, Sprock J, Light GA. Source decomposition of the frontocentral auditory steady-state gamma band response in schizophrenia patients and healthy subjects. Psychiatry Clin Neurosci 2021; 75:172-179. [PMID: 33470494 DOI: 10.1111/pcn.13201] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 01/12/2021] [Accepted: 01/16/2021] [Indexed: 12/27/2022]
Abstract
AIM Gamma-band auditory steady-state response (ASSR) is a neurophysiologic index that is increasingly used as a translational biomarker in the development of treatments of neuropsychiatric disorders. While gamma-band ASSR is generated by distributed networks of highly interactive temporal and frontal cortical sources, the majority of human gamma-band ASSR studies using electroencephalography (EEG) highlight activity from only a single frontocentral scalp site, Fz, where responses tend to be largest and reductions in schizophrenia patients are most evident. However, no previous study has characterized the relative source contributions to Fz, which is a necessary step to improve the concordance of preclinical and clinical EEG studies. METHODS A novel method to back-project the contributions of independent cortical source components was applied to assess the independent sources and their proportional contributions to Fz as well as source-resolved responses in 432 schizophrenia patients and 294 healthy subjects. RESULTS Independent contributions of gamma-band ASSR to Fz were detected from orbitofrontal, bilateral superior/middle/inferior temporal, bilateral middle frontal, and posterior cingulate gyri in both groups. In contrast to expectations, the groups showed comparable source contribution weight to gamma-band ASSR at Fz. While gamma-band ASSR reductions at Fz were present in schizophrenia patients consistent with previous studies, no group differences in individual source-level responses to Fz were detected. CONCLUSION Small differences in multiple independent sources summate to produce scalp-level differences at Fz. The identification of independent source contributions to a single scalp sensor represents a promising methodology for measuring dissociable and homologous biomarker targets in future translational studies.
Collapse
Affiliation(s)
- Daisuke Koshiyama
- Department of Psychiatry, University of California San Diego, La Jolla, USA
| | - Makoto Miyakoshi
- Swartz Center for Neural Computation, University of California San Diego, La Jolla, USA
| | - Yash B Joshi
- Department of Psychiatry, University of California San Diego, La Jolla, USA.,VISN-22 Mental Illness, Research, Education and Clinical Center (MIRECC), VA San Diego Healthcare System, San Diego, USA
| | - Masaki Nakanishi
- Swartz Center for Neural Computation, University of California San Diego, La Jolla, USA
| | | | - Joyce Sprock
- Department of Psychiatry, University of California San Diego, La Jolla, USA.,VISN-22 Mental Illness, Research, Education and Clinical Center (MIRECC), VA San Diego Healthcare System, San Diego, USA
| | - Gregory A Light
- Department of Psychiatry, University of California San Diego, La Jolla, USA.,VISN-22 Mental Illness, Research, Education and Clinical Center (MIRECC), VA San Diego Healthcare System, San Diego, USA
| |
Collapse
|
137
|
Kilonzo K, van der Veen B, Teutsch J, Schulz S, Kapanaiah SKT, Liss B, Kätzel D. Delayed-matching-to-position working memory in mice relies on NMDA-receptors in prefrontal pyramidal cells. Sci Rep 2021; 11:8788. [PMID: 33888809 PMCID: PMC8062680 DOI: 10.1038/s41598-021-88200-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 03/30/2021] [Indexed: 02/02/2023] Open
Abstract
A hypofunction of N-methyl-D-aspartate glutamate receptors (NMDARs) has been implicated in the pathogenesis of schizophrenia by clinical and rodent studies. However, to what extent NMDAR-hypofunction in distinct cell-types across the brain causes different symptoms of this disease is largely unknown. One pharmaco-resistant core symptom of schizophrenia is impaired working memory (WM). NMDARs have been suggested to mediate sustained firing in excitatory neurons of the prefrontal cortex (PFC) that might underlie WM storage. However, if NMDAR-hypofunction in prefrontal excitatory neurons may indeed entail WM impairments is unknown. We here investigated this question in mice, in which NMDARs were genetically-ablated in PFC excitatory cells. This cell type-selective NMDAR-hypofunction caused a specific deficit in a delayed-matching-to-position (DMTP) 5-choice-based operant WM task. In contrast, T-maze rewarded alternation and several psychological functions including attention, spatial short-term habituation, novelty-processing, motivation, sociability, impulsivity, and hedonic valuation remained unimpaired at the level of GluN1-hypofunction caused by our manipulation. Our data suggest that a hypofunction of NMDARs in prefrontal excitatory neurons may indeed cause WM impairments, but are possibly not accounting for most other deficits in schizophrenia.
Collapse
Affiliation(s)
- Kasyoka Kilonzo
- Institute of Applied Physiology, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | - Bastiaan van der Veen
- Institute of Applied Physiology, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | - Jasper Teutsch
- Institute of Applied Physiology, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany
- Newcastle University, Newcastle upon Tyne, UK
| | - Stefanie Schulz
- Institute of Applied Physiology, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | - Sampath K T Kapanaiah
- Institute of Applied Physiology, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | - Birgit Liss
- Institute of Applied Physiology, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany
- Linacre College and New College, University of Oxford, Oxford, UK
| | - Dennis Kätzel
- Institute of Applied Physiology, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany.
| |
Collapse
|
138
|
Leung LS, Chu L, Prado MAM, Prado VF. Forebrain Acetylcholine Modulates Isoflurane and Ketamine Anesthesia in Adult Mice. Anesthesiology 2021; 134:588-606. [PMID: 33635947 DOI: 10.1097/aln.0000000000003713] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Cholinergic drugs are known to modulate general anesthesia, but anesthesia responses in acetylcholine-deficient mice have not been studied. It was hypothesized that mice with genetic deficiency of forebrain acetylcholine show increased anesthetic sensitivity to isoflurane and ketamine and decreased gamma-frequency brain activity. METHODS Male adult mice with heterozygous knockdown of vesicular acetylcholine transporter in the brain or homozygous knockout of the transporter in the basal forebrain were compared with wild-type mice. Hippocampal and frontal cortical electrographic activity and righting reflex were studied in response to isoflurane and ketamine doses. RESULTS The loss-of-righting-reflex dose for isoflurane was lower in knockout (mean ± SD, 0.76 ± 0.08%, n = 18, P = 0.005) but not knockdown (0.78 ± 0.07%, n = 24, P = 0.021), as compared to wild-type mice (0.83 ± 0.07%, n = 23), using a significance criterion of P = 0.017 for three planned comparisons. Loss-of-righting-reflex dose for ketamine was lower in knockout (144 ± 39 mg/kg, n = 14, P = 0.006) but not knockdown (162 ± 32 mg/kg, n = 20, P = 0.602) as compared to wild-type mice (168 ± 24 mg/kg, n = 21). Hippocampal high-gamma (63 to 100 Hz) power after isoflurane was significantly lower in knockout and knockdown mice compared to wild-type mice (isoflurane-dose and mouse-group interaction effect, F[8,56] = 2.87, P = 0.010; n = 5 to 6 mice per group). Hippocampal high-gamma power after ketamine was significantly lower in both knockout and knockdown mice when compared to wild-type mice (interaction effect F[2,13] = 6.06, P = 0.014). The change in frontal cortical gamma power with isoflurane or ketamine was not statistically different among knockout, knockdown, and wild-type mice. CONCLUSIONS These findings suggest that forebrain cholinergic neurons modulate behavioral sensitivity and hippocampal gamma activity during isoflurane and ketamine anesthesia. EDITOR’S PERSPECTIVE
Collapse
|
139
|
Koshiyama D, Miyakoshi M, Joshi YB, Molina JL, Tanaka-Koshiyama K, Sprock J, Braff DL, Swerdlow NR, Light GA. Neural network dynamics underlying gamma synchronization deficits in schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2021; 107:110224. [PMID: 33340619 PMCID: PMC8631608 DOI: 10.1016/j.pnpbp.2020.110224] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/06/2020] [Accepted: 12/09/2020] [Indexed: 01/09/2023]
Abstract
Gamma-band (40-Hz) activity is critical for cortico-cortical transmission and the integration of information across neural networks during sensory and cognitive processing. Patients with schizophrenia show selective reductions in the capacity to support synchronized gamma-band oscillations in response to auditory stimulation presented 40-Hz. Despite widespread application of this 40-Hz auditory steady-state response (ASSR) as a translational electroencephalographic biomarker for therapeutic development for neuropsychiatric disorders, the spatiotemporal dynamics underlying the ASSR have not been fully characterized. In this study, a novel Granger causality analysis was applied to assess the propagation of gamma oscillations in response to 40-Hz steady-state stimulation across cortical sources in schizophrenia patients (n = 426) and healthy comparison subjects (n = 293). Both groups showed multiple ASSR source interactions that were broadly distributed across brain regions. Schizophrenia patients showed distinct, hierarchically sequenced connectivity abnormalities. During the response onset interval, patients exhibited abnormal increased connectivity from the inferior frontal gyrus to the superior temporal gyrus, followed by decreased connectivity from the superior temporal to the middle cingulate gyrus. In the later portion of the ASSR response (300-500 ms), patients showed significantly increased connectivity from the superior temporal to the middle frontal gyrus followed by decreased connectivity from the left superior frontal gyrus to the right superior and middle frontal gyri. These findings highlight both the orchestration of distributed multiple sources in response to simple gamma-frequency stimulation in healthy subjects as well as the patterns of deficits in the generation and maintenance of gamma-band oscillations across the temporo-frontal sources in schizophrenia patients.
Collapse
Affiliation(s)
- Daisuke Koshiyama
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093-0804, USA
| | - Makoto Miyakoshi
- Swartz Center for Neural Computation, University of California San Diego, La Jolla, CA 92093-0559, USA.
| | - Yash B. Joshi
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093-0804, USA,VISN-22 Mental Illness, Research, Education and Clinical Center (MIRECC), VA San Diego Healthcare System, San Diego, CA 92161, USA
| | - Juan L. Molina
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093-0804, USA,VISN-22 Mental Illness, Research, Education and Clinical Center (MIRECC), VA San Diego Healthcare System, San Diego, CA 92161, USA
| | | | - Joyce Sprock
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093-0804, USA,VISN-22 Mental Illness, Research, Education and Clinical Center (MIRECC), VA San Diego Healthcare System, San Diego, CA 92161, USA
| | - David L. Braff
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093-0804, USA,VISN-22 Mental Illness, Research, Education and Clinical Center (MIRECC), VA San Diego Healthcare System, San Diego, CA 92161, USA
| | - Neal R. Swerdlow
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093-0804, USA
| | - Gregory A. Light
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093-0804, USA,VISN-22 Mental Illness, Research, Education and Clinical Center (MIRECC), VA San Diego Healthcare System, San Diego, CA 92161, USA
| |
Collapse
|
140
|
Yang J, Yang X, Tang K. Interneuron development and dysfunction. FEBS J 2021; 289:2318-2336. [PMID: 33844440 DOI: 10.1111/febs.15872] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 04/09/2021] [Indexed: 12/17/2022]
Abstract
Understanding excitation and inhibition balance in the brain begins with the tale of two basic types of neurons, glutamatergic projection neurons and GABAergic interneurons. The diversity of cortical interneurons is contributed by multiple origins in the ventral forebrain, various tangential migration routes, and complicated regulations of intrinsic factors, extrinsic signals, and activities. Abnormalities of interneuron development lead to dysfunction of interneurons and inhibitory circuits, which are highly associated with neurodevelopmental disorders including schizophrenia, autism spectrum disorders, and intellectual disability. In this review, we mainly discuss recent findings on the development of cortical interneuron and on neurodevelopmental disorders related to interneuron dysfunction.
Collapse
Affiliation(s)
- Jiaxin Yang
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, China
| | - Xiong Yang
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, China
| | - Ke Tang
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, China
| |
Collapse
|
141
|
Guyon N, Zacharias LR, van Lunteren JA, Immenschuh J, Fuzik J, Märtin A, Xuan Y, Zilberter M, Kim H, Meletis K, Lopes-Aguiar C, Carlén M. Adult trkB Signaling in Parvalbumin Interneurons is Essential to Prefrontal Network Dynamics. J Neurosci 2021; 41:3120-3141. [PMID: 33593856 PMCID: PMC8026352 DOI: 10.1523/jneurosci.1848-20.2021] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 01/16/2021] [Accepted: 01/25/2021] [Indexed: 01/29/2023] Open
Abstract
Inhibitory interneurons expressing parvalbumin (PV) are central to cortical network dynamics, generation of γ oscillations, and cognition. Dysfunction of PV interneurons disrupts cortical information processing and cognitive behavior. Brain-derived neurotrophic factor (BDNF)/tyrosine receptor kinase B (trkB) signaling regulates the maturation of cortical PV interneurons but is also implicated in their adult multidimensional functions. Using a novel viral strategy for cell-type-specific and spatially restricted expression of a dominant-negative trkB (trkB.DN), we show that BDNF/trkB signaling is essential to the integrity and maintenance of prefrontal PV interneurons in adult male and female mice. Reduced BDNF/trkB signaling in PV interneurons in the medial prefrontal cortex (mPFC) resulted in deficient PV inhibition and increased baseline local field potential (LFP) activity in a broad frequency band. The altered network activity was particularly pronounced during increased activation of the prefrontal network and was associated with changed dynamics of local excitatory neurons, as well as decreased modulation of the LFP, abnormalities that appeared to generalize across stimuli and brain states. In addition, our findings link reduced BDNF/trkB signaling in prefrontal PV interneurons to increased aggression. Together our investigations demonstrate that BDNF/trkB signaling in PV interneurons in the adult mPFC is essential to local network dynamics and cognitive behavior. Our data provide direct support for the suggested association between decreased trkB signaling, deficient PV inhibition, and altered prefrontal circuitry.SIGNIFICANCE STATEMENT Brain-derived neurotrophic factor (BDNF)/tyrosine receptor kinase B (trkB) signaling promotes the maturation of inhibitory parvalbumin (PV) interneurons, neurons central to local cortical dynamics, γ rhythms, and cognition. Here, we used a novel viral approach for reduced BDNF/trkB signaling in PV interneurons in the medial prefrontal cortex (mPFC) to establish the role of BDNF/trkB signaling in adult prefrontal network activities. Reduced BDNF/trkB signaling caused pronounced morphologic alterations, reduced PV inhibition, and deficient prefrontal network dynamics. The altered network activity appeared to manifest across stimuli and brain states and was associated with aberrant local field potential (LFP) activities and increased aggression. The results demonstrate that adult BDNF/trkB signaling is essential to PV inhibition and prefrontal circuit function and directly links BDNF/trkB signaling to network integrity in the adult brain.
Collapse
Affiliation(s)
- Nicolas Guyon
- Department of Neuroscience, Karolinska Institutet, Stockholm 17177, Sweden
| | - Leonardo Rakauskas Zacharias
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, Universidade de São Paulo, Ribeirão Preto 14049-900, Brazil
| | | | - Jana Immenschuh
- Department of Neuroscience, Karolinska Institutet, Stockholm 17177, Sweden
| | - Janos Fuzik
- Department of Neuroscience, Karolinska Institutet, Stockholm 17177, Sweden
| | - Antje Märtin
- Department of Neuroscience, Karolinska Institutet, Stockholm 17177, Sweden
| | - Yang Xuan
- Department of Neuroscience, Karolinska Institutet, Stockholm 17177, Sweden
| | - Misha Zilberter
- Department of Neuroscience, Karolinska Institutet, Stockholm 17177, Sweden
| | - Hoseok Kim
- Department of Neuroscience, Karolinska Institutet, Stockholm 17177, Sweden
| | | | - Cleiton Lopes-Aguiar
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Marie Carlén
- Department of Neuroscience, Karolinska Institutet, Stockholm 17177, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge 14183, Sweden
| |
Collapse
|
142
|
Pafundo DE, Pretell Annan CA, Fulginiti NM, Belforte JE. Early NMDA Receptor Ablation in Interneurons Causes an Activity-Dependent E/I Imbalance in vivo in Prefrontal Cortex Pyramidal Neurons of a Mouse Model Useful for the Study of Schizophrenia. Schizophr Bull 2021; 47:1300-1309. [PMID: 33822178 PMCID: PMC8379555 DOI: 10.1093/schbul/sbab030] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Altered Excitatory/Inhibitory (E/I) balance of cortical synaptic inputs has been proposed as a central pathophysiological factor for psychiatric neurodevelopmental disorders, including schizophrenia (SZ). However, direct measurement of E/I synaptic balance have not been assessed in vivo for any validated SZ animal model. Using a mouse model useful for the study of SZ we show that a selective ablation of NMDA receptors (NMDAr) in cortical and hippocampal interneurons during early postnatal development results in an E/I imbalance in vivo, with synaptic inputs to pyramidal neurons shifted towards excitation in the adult mutant medial prefrontal cortex (mPFC). Remarkably, this imbalance depends on the cortical state, only emerging when theta and gamma oscillations are predominant in the network. Additional brain slice recordings and subsequent 3D morphological reconstruction showed that E/I imbalance emerges after adolescence concomitantly with significant dendritic retraction and dendritic spine re-localization in pyramidal neurons. Therefore, early postnatal ablation of NMDAr in cortical and hippocampal interneurons developmentally impacts on E/I imbalance in vivo in an activity-dependent manner.
Collapse
Affiliation(s)
- Diego E Pafundo
- Grupo de Neurociencia de Sistemas, Instituto de Fisiología y Biofísica “Bernardo Houssay” (IFIBIO-Houssay), Universidad de Buenos Aires y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad de Buenos Aires, Argentina,To whom correspondence should be addressed; 2155 Paraguay 7 floor, Ciudad de Buenos Aires, 1121 Argentina; tel: +54-11-5285-3309, fax: +54-11-5950-9500 ext 2142, e-mail:
| | - Carlos A Pretell Annan
- Grupo de Neurociencia de Sistemas, Instituto de Fisiología y Biofísica “Bernardo Houssay” (IFIBIO-Houssay), Universidad de Buenos Aires y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad de Buenos Aires, Argentina
| | - Nicolas M Fulginiti
- Grupo de Neurociencia de Sistemas, Instituto de Fisiología y Biofísica “Bernardo Houssay” (IFIBIO-Houssay), Universidad de Buenos Aires y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad de Buenos Aires, Argentina
| | - Juan E Belforte
- Grupo de Neurociencia de Sistemas, Instituto de Fisiología y Biofísica “Bernardo Houssay” (IFIBIO-Houssay), Universidad de Buenos Aires y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad de Buenos Aires, Argentina
| |
Collapse
|
143
|
Kohtala S. Ketamine-50 years in use: from anesthesia to rapid antidepressant effects and neurobiological mechanisms. Pharmacol Rep 2021; 73:323-345. [PMID: 33609274 PMCID: PMC7994242 DOI: 10.1007/s43440-021-00232-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 02/03/2021] [Accepted: 02/06/2021] [Indexed: 12/11/2022]
Abstract
Over the past 50 years, ketamine has solidified its position in both human and veterinary medicine as an important anesthetic with many uses. More recently, ketamine has been studied and used for several new indications, ranging from chronic pain to drug addiction and post-traumatic stress disorder. The discovery of the rapid-acting antidepressant effects of ketamine has resulted in a surge of interest towards understanding the precise mechanisms driving its effects. Indeed, ketamine may have had the largest impact for advancements in the research and treatment of psychiatric disorders in the past few decades. While intense research efforts have been aimed towards uncovering the molecular targets underlying ketamine's effects in treating depression, the underlying neurobiological mechanisms remain elusive. These efforts are made more difficult by ketamine's complex dose-dependent effects on molecular mechanisms, multiple pharmacologically active metabolites, and a mechanism of action associated with the facilitation of synaptic plasticity. This review aims to provide a brief overview of the different uses of ketamine, with an emphasis on examining ketamine's rapid antidepressant effects spanning molecular, cellular, and network levels. Another focus of the review is to offer a perspective on studies related to the different doses of ketamine used in antidepressant research. Finally, the review discusses some of the latest hypotheses concerning ketamine's action.
Collapse
Affiliation(s)
- Samuel Kohtala
- Laboratory of Neurotherapeutics, Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P. O. Box 56, 00014, Helsinki, Finland.
- SleepWell Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- Feil Family Brain and Mind Research Institute, Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
144
|
Huang MX, Huang CW, Harrington DL, Nichols S, Robb-Swan A, Angeles-Quinto A, Le L, Rimmele C, Drake A, Song T, Huang JW, Clifford R, Ji Z, Cheng CK, Lerman I, Yurgil KA, Lee RR, Baker DG. Marked Increases in Resting-State MEG Gamma-Band Activity in Combat-Related Mild Traumatic Brain Injury. Cereb Cortex 2021; 30:283-295. [PMID: 31041986 DOI: 10.1093/cercor/bhz087] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 03/29/2019] [Accepted: 04/01/2019] [Indexed: 01/08/2023] Open
Abstract
Combat-related mild traumatic brain injury (mTBI) is a leading cause of sustained impairments in military service members and veterans. Recent animal studies show that GABA-ergic parvalbumin-positive interneurons are susceptible to brain injury, with damage causing abnormal increases in spontaneous gamma-band (30-80 Hz) activity. We investigated spontaneous gamma activity in individuals with mTBI using high-resolution resting-state magnetoencephalography source imaging. Participants included 25 symptomatic individuals with chronic combat-related blast mTBI and 35 healthy controls with similar combat experiences. Compared with controls, gamma activity was markedly elevated in mTBI participants throughout frontal, parietal, temporal, and occipital cortices, whereas gamma activity was reduced in ventromedial prefrontal cortex. Across groups, greater gamma activity correlated with poorer performances on tests of executive functioning and visuospatial processing. Many neurocognitive associations, however, were partly driven by the higher incidence of mTBI participants with both higher gamma activity and poorer cognition, suggesting that expansive upregulation of gamma has negative repercussions for cognition particularly in mTBI. This is the first human study to demonstrate abnormal resting-state gamma activity in mTBI. These novel findings suggest the possibility that abnormal gamma activities may be a proxy for GABA-ergic interneuron dysfunction and a promising neuroimaging marker of insidious mild head injuries.
Collapse
Affiliation(s)
- Ming-Xiong Huang
- Radiology, Research, and Psychiatry Services, VA San Diego Healthcare System, San Diego, CA, USA.,Department of Radiology, University of California, San Diego, CA, USA
| | - Charles W Huang
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Deborah L Harrington
- Radiology, Research, and Psychiatry Services, VA San Diego Healthcare System, San Diego, CA, USA.,Department of Radiology, University of California, San Diego, CA, USA
| | - Sharon Nichols
- Department of Neuroscience, University of California, San Diego, CA, USA
| | - Ashley Robb-Swan
- Radiology, Research, and Psychiatry Services, VA San Diego Healthcare System, San Diego, CA, USA.,Department of Radiology, University of California, San Diego, CA, USA
| | - Annemarie Angeles-Quinto
- Radiology, Research, and Psychiatry Services, VA San Diego Healthcare System, San Diego, CA, USA.,Department of Radiology, University of California, San Diego, CA, USA
| | - Lu Le
- ASPIRE Center, VASDHS Residential Rehabilitation Treatment Program, San Diego, CA, USA
| | - Carl Rimmele
- ASPIRE Center, VASDHS Residential Rehabilitation Treatment Program, San Diego, CA, USA
| | - Angela Drake
- Cedar Sinai Medical Group Chronic Pain Program, Beverly Hills, CA, USA
| | - Tao Song
- Department of Radiology, University of California, San Diego, CA, USA
| | - Jeffrey W Huang
- Department of Computer Science, Columbia University, New York, NY, USA
| | - Royce Clifford
- Radiology, Research, and Psychiatry Services, VA San Diego Healthcare System, San Diego, CA, USA.,Department of Psychiatry, University of California, San Diego, CA, USA.,VA Center of Excellence for Stress and Mental Health, San Diego, CA, USA
| | - Zhengwei Ji
- Department of Radiology, University of California, San Diego, CA, USA
| | - Chung-Kuan Cheng
- Department of Computer Science and Engineering, University of California, San Diego, CA, USA
| | - Imanuel Lerman
- Radiology, Research, and Psychiatry Services, VA San Diego Healthcare System, San Diego, CA, USA
| | - Kate A Yurgil
- Radiology, Research, and Psychiatry Services, VA San Diego Healthcare System, San Diego, CA, USA.,VA Center of Excellence for Stress and Mental Health, San Diego, CA, USA.,Department of Psychological Sciences, Loyola University, New Orleans, LA, USA
| | - Roland R Lee
- Radiology, Research, and Psychiatry Services, VA San Diego Healthcare System, San Diego, CA, USA.,Department of Radiology, University of California, San Diego, CA, USA
| | - Dewleen G Baker
- Radiology, Research, and Psychiatry Services, VA San Diego Healthcare System, San Diego, CA, USA.,Department of Psychiatry, University of California, San Diego, CA, USA.,VA Center of Excellence for Stress and Mental Health, San Diego, CA, USA
| |
Collapse
|
145
|
Yang Z, Xiao X, Chen R, Xu X, Kong W, Zhang T. Disc1 gene down-regulation impaired synaptic plasticity and recognition memory via disrupting neural activity in mice. Brain Res Bull 2021; 171:84-90. [PMID: 33745948 DOI: 10.1016/j.brainresbull.2021.03.011] [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: 10/02/2020] [Revised: 02/25/2021] [Accepted: 03/15/2021] [Indexed: 10/21/2022]
Abstract
The gene of Disrupted-in-schizophrenia 1 (Disc1) is closely related to mental diseases with cognitive deficits, but there are few studies on the changes in neural oscillations and recognition memory. Neural oscillations plays a key role in the nervous system in a dynamic form, which is closely related to advanced cognitive activities such as information processing and memory consolidation. Hence, we aimed to investigate if Disc1 knockdown disrupted the normal pattern of neural activities in the mouse hippocampus network, and determined if quantitative neural oscillation approach could be a potential diagnostic tool for mental disorders. In the study, we reported that Disc1 gene, downregulated by short-hairpin RNA (shRNA), not only induced anxiety-like behavior and sociability impairment but also damaged both synaptic plasticity and recognition memory in mice. Moreover, Disc1 knockdown mice exhibited evidently abnormal power spectral distributions, reduced phase synchronizations, and decreased phase-amplitude coupling strength compared to that of normal animals. In addition, transcriptome analyses showed that there were clearly transcriptional changes in Disc1 knockdown mice. Altogether, our findings suggest that the abnormal pattern of neural activities in the hippocampus network disrupts information processing and finally leads to the impairments of synaptic plasticity and recognition in Disc1 knockdown mice, which are possibly associated with the obstruction of neurotransmitter transmission. Importantly, the data imply that the analysis of neural oscillation pattern provides a potential diagnosis approach for mental disorders.
Collapse
Affiliation(s)
- Ze Yang
- College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, 300071, Tianjin, PR China
| | - Xi Xiao
- College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, 300071, Tianjin, PR China; Tianjin International Joint Research Center for Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, 300072, Tianjin, PR China
| | - Runwen Chen
- College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, 300071, Tianjin, PR China
| | - Xinxin Xu
- College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, 300071, Tianjin, PR China
| | - Wanzeng Kong
- Key Laboratory of Brain Machine Collaborative Intelligence of Zhejiang Province, Hangzhou Dianzi University, 310018, Hangzhou, PR China.
| | - Tao Zhang
- College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, 300071, Tianjin, PR China.
| |
Collapse
|
146
|
Ye T, Bartlett MJ, Sherman SJ, Falk T, Cowen SL. Spectral signatures of L-DOPA-induced dyskinesia depend on L-DOPA dose and are suppressed by ketamine. Exp Neurol 2021; 340:113670. [PMID: 33662379 DOI: 10.1016/j.expneurol.2021.113670] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 02/11/2021] [Accepted: 02/25/2021] [Indexed: 01/22/2023]
Abstract
L-DOPA-induced dyskinesias (LID) are debilitating motor symptoms of dopamine-replacement therapy for Parkinson's disease (PD) that emerge after years of L-DOPA treatment. While there is an abundance of research into the cellular and synaptic origins of LID, less is known about how LID impacts systems-level circuits and neural synchrony, how synchrony is affected by the dose and duration of L-DOPA exposure, or how potential novel treatments for LID, such as sub-anesthetic ketamine, alter this activity. Sub-anesthetic ketamine treatments have recently been shown to reduce LID, and ketamine is known to affect neural synchrony. To investigate these questions, we measured movement and local-field potential (LFP) activity from the motor cortex (M1) and the striatum of preclinical rodent models of PD and LID. In the first experiment, we investigated the effect of the LID priming procedures and L-DOPA dose on neural signatures of LID. Two common priming procedures were compared: a high-dose procedure that exposed unilateral 6-hydroxydopamine-lesioned rats to 12 mg/kg L-DOPA for 7 days, and a low-dose procedure that exposed rats to 7 mg/kg L-DOPA for 21 days. Consistent with reports from other groups, 12 mg/kg L-DOPA triggered LID and 80-Hz oscillations; however, these 80-Hz oscillations were not observed after 7 mg/kg administration despite clear evidence of LID, indicating that 80-Hz oscillations are not an exclusive signature of LID. We also found that weeks-long low-dose priming resulted in the emergence of non-oscillatory broadband gamma activity (> 30 Hz) in the striatum and theta-to-high-gamma cross-frequency coupling (CFC) in M1. In a second set of experiments, we investigated how ketamine exposure affects spectral signatures of low-dose L-DOPA priming. During each neural recording session, ketamine was delivered through 5 injections (20 mg/kg, i.p.) administered every 2 h. We found that ketamine exposure suppressed striatal broadband gamma associated with LID but enhanced M1 broadband activity. We also found that M1 theta-to-high-gamma CFC associated with the LID on-state was suppressed by ketamine. These results suggest that ketamine's therapeutic effects are region specific. Our findings also have clinical implications, as we are the first to report novel oscillatory signatures of the common low-dose LID priming procedure that more closely models dopamine replacement therapy in individuals with PD. We also identify neural correlates of the anti-dyskinetic activity of sub-anesthetic ketamine treatment.
Collapse
Affiliation(s)
- Tony Ye
- Department of Neurology, University of Arizona College of Medicine, Tucson, AZ, United States of America.
| | - Mitchell J Bartlett
- Department of Neurology, University of Arizona College of Medicine, Tucson, AZ, United States of America; Department of Pharmacology, University of Arizona College of Medicine, Tucson, AZ, United States of America.
| | - Scott J Sherman
- Department of Neurology, University of Arizona College of Medicine, Tucson, AZ, United States of America.
| | - Torsten Falk
- Department of Neurology, University of Arizona College of Medicine, Tucson, AZ, United States of America; Department of Pharmacology, University of Arizona College of Medicine, Tucson, AZ, United States of America.
| | - Stephen L Cowen
- Department of Psychology, University of Arizona, Tucson, AZ, United States of America.
| |
Collapse
|
147
|
A randomized cross-over trial to define neurophysiological correlates of AV-101 N-methyl-D-aspartate receptor blockade in healthy veterans. Neuropsychopharmacology 2021; 46:820-827. [PMID: 33318635 PMCID: PMC8027791 DOI: 10.1038/s41386-020-00917-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 10/22/2020] [Accepted: 11/13/2020] [Indexed: 12/03/2022]
Abstract
The kynurenine pathway (KP) is a strategic metabolic system that combines regulation of neuronal excitability via glutamate receptor function and neuroinflammation via other KP metabolites. This pathway has great promise in treatment of depression and suicidality. The KP modulator AV-101 (4-chlorokynurenine, 4-Cl-KYN), an oral prodrug of 7-chlorokynurenic acid (7-Cl-KYNA), an N-methyl-D-aspartate receptor (NMDAR) glycine site antagonist, and of 4-chloro-3-hydroxyanthranilic acid (4-Cl-3-HAA), a suppressor of NMDAR agonist quinolinic acid (QUIN), is a promising potential antidepressant that targets glutamate functioning via the KP. However, a recent placebo-controlled clinical trial of AV-101 in depression found negative results. This raises the question of whether AV-101 can penetrate the brain and engage the NMDAR and KP effectively. To address this problem, ten healthy US military veterans (mean age = 32.6 years ± 6.11; 1 female) completed a phase-1 randomized, double-blind, placebo-controlled, crossover study to examine dose-related effects of AV-101 (720 and 1440 mg) on NMDAR engagement measured by γ-frequency band auditory steady-state response (40 Hz ASSR) and resting EEG. Linear mixed models revealed that 1440 mg AV-101, but not 720 mg, increased 40 Hz ASSR and 40 Hz ASSR γ-inter-trial phase coherence relative to placebo. AV-101 also increased 4-Cl-KYN, 7-Cl-KYNA, 4-Cl-3-HAA, 3-HAA, and KYNA in a dose-dependent manner, without affecting KYN and QUIN. AV-101 was safe and well tolerated. These results corroborate brain target engagement of 1440 mg AV-101 in humans, consistent with blockade of interneuronal NMDAR blockade. Future studies should test higher doses of AV-101 in depression. Suicidal behavior, which has been associated with high QUIN and low KYNA, is also a potential target for AV-101.
Collapse
|
148
|
Network Asynchrony Underlying Increased Broadband Gamma Power. J Neurosci 2021; 41:2944-2963. [PMID: 33593859 DOI: 10.1523/jneurosci.2250-20.2021] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 01/21/2021] [Accepted: 01/30/2021] [Indexed: 12/16/2022] Open
Abstract
Synchronous activity of cortical inhibitory interneurons expressing parvalbumin (PV) underlies expression of cortical γ rhythms. Paradoxically, deficient PV inhibition is associated with increased broadband γ power in the local field potential. Increased baseline broadband γ is also a prominent characteristic in schizophrenia and a hallmark of network alterations induced by NMDAR antagonists, such as ketamine. Whether enhanced broadband γ is a true rhythm, and if so, whether rhythmic PV inhibition is involved or not, is debated. Asynchronous and increased firing activities are thought to contribute to broadband power increases spanning the γ band. Using male and female mice lacking NMDAR activity specifically in PV neurons to model deficient PV inhibition, we here show that neuronal activity with decreased synchronicity is associated with increased prefrontal broadband γ power. Specifically, reduced spike time precision and spectral leakage of spiking activity because of higher firing rates (spike "contamination") affect the broadband γ band. Desynchronization was evident at multiple time scales, with reduced spike entrainment to the local field potential, reduced cross-frequency coupling, and fragmentation of brain states. Local application of S(+)-ketamine in (control) mice with intact NMDAR activity in PV neurons triggered network desynchronization and enhanced broadband γ power. However, our investigations suggest that disparate mechanisms underlie increased broadband γ power caused by genetic alteration of PV interneurons and ketamine-induced power increases in broadband γ. Our study confirms that enhanced broadband γ power can arise from asynchronous activities and demonstrates that long-term deficiency of PV inhibition can be a contributor.SIGNIFICANCE STATEMENT Brain oscillations are fundamental to the coordination of neuronal activity across neurons and structures. γ oscillations (30-80 Hz) have received particular attention through their association with perceptual and cognitive processes. Synchronous activity of inhibitory parvalbumin (PV) interneurons generates cortical γ oscillation, but, paradoxically, PV neuron deficiency is associated with increases in γ oscillations. We here reconcile this conundrum and show how deficient PV inhibition can lead to increased and asynchronous excitatory firing, contaminating the local field potential and manifesting as increased γ power. Thus, increased γ power does not always reflect a genuine rhythm. Further, we show that ketamine-induced γ increases are caused by separate network mechanisms.
Collapse
|
149
|
Optogenetic Approaches to Understand the Neural Circuit Mechanism of Social Deficits Seen in Autism Spectrum Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021. [PMID: 33398839 DOI: 10.1007/978-981-15-8763-4_36] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Individuals with neurodevelopmental disorders, such as autism spectrum disorders (ASDs), are diagnosed based on nonquantitative objective parameters such as behavioral phenotypes. It is still unclear how any neural mechanism affects such behavioral phenotypes in these patients. In human genetics, a large number of genetic abnormalities including single nucleotide variation (SNV) and copy number variation (CNV) have been found in individuals with ASDs. It is thought that influence of such variations converges on dysfunction of neural circuit resulting in common behavioral phenotypes of ASDs such as deficits in social communication and interaction. Recent studies suggest that an excitatory/inhibitory (E/I) imbalanced state, which induces disruption of neural circuit activities, is one of the pathophysiological abnormalities in ASD brains. To assess the causal relationship between brain abnormalities and behavioral deficits, we can take advantage of optogenetics with animal models of ASDs that recapitulate human genetic mutations. Here, we review optogenetics studies being utilized to dissect neural circuit mechanisms associated with social deficits in model mice of ASD. Optogenetic manipulation of disrupted neural activities would help us understand how neural circuits affect behavioral deficits observed in ASDs.
Collapse
|
150
|
Neklyudova AK, Portnova GV, Rebreikina AB, Voinova VY, Vorsanova SG, Iourov IY, Sysoeva OV. 40-Hz Auditory Steady-State Response (ASSR) as a Biomarker of Genetic Defects in the SHANK3 Gene: A Case Report of 15-Year-Old Girl with a Rare Partial SHANK3 Duplication. Int J Mol Sci 2021; 22:1898. [PMID: 33673024 PMCID: PMC7917917 DOI: 10.3390/ijms22041898] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 01/26/2021] [Accepted: 02/09/2021] [Indexed: 12/02/2022] Open
Abstract
SHANK3 encodes a scaffold protein involved in postsynaptic receptor density in glutamatergic synapses, including those in the parvalbumin (PV)+ inhibitory neurons-the key players in the generation of sensory gamma oscillations, such as 40-Hz auditory steady-state response (ASSR). However, 40-Hz ASSR was not studied in relation to SHANK3 functioning. Here, we present a 15-year-old girl (SH01) with previously unreported duplication of the first seven exons of the SHANK3 gene (22q13.33). SH01's electroencephalogram (EEG) during 40-Hz click trains of 500 ms duration binaurally presented with inter-trial intervals of 500-800 ms were compared with those from typically developing children (n = 32). SH01 was diagnosed with mild mental retardation and learning disabilities (F70.88), dysgraphia, dyslexia, and smaller vocabulary than typically developing (TD) peers. Her clinical phenotype resembled the phenotype of previously described patients with 22q13.33 microduplications (≈30 reported so far). SH01 had mild autistic symptoms but below the threshold for ASD diagnosis and microcephaly. No seizures or MRI abnormalities were reported. While SH01 had relatively preserved auditory event-related potential (ERP) with slightly attenuated P1, her 40-Hz ASSR was totally absent significantly deviating from TD's ASSR. The absence of 40-Hz ASSR in patients with microduplication, which affected the SHANK3 gene, indicates deficient temporal resolution of the auditory system, which might underlie language problems and represent a neurophysiological biomarker of SHANK3 abnormalities.
Collapse
Affiliation(s)
- Anastasia K. Neklyudova
- Laboratory of Human Higher Nervous Activity, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Science, 117485 Moscow, Russia; (A.K.N.); (G.V.P.); (A.B.R.)
| | - Galina V. Portnova
- Laboratory of Human Higher Nervous Activity, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Science, 117485 Moscow, Russia; (A.K.N.); (G.V.P.); (A.B.R.)
| | - Anna B. Rebreikina
- Laboratory of Human Higher Nervous Activity, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Science, 117485 Moscow, Russia; (A.K.N.); (G.V.P.); (A.B.R.)
| | - Victoria Yu Voinova
- Veltischev Research and Clinical Institute for Pediatrics of the Pirogov, Russian National Research Medical University, Ministry of Health of Russian Federation, 125412 Moscow, Russia; (V.Y.V.); (S.G.V.); (I.Y.I.)
- Mental Health Research Center, 117152 Moscow, Russia
| | - Svetlana G. Vorsanova
- Veltischev Research and Clinical Institute for Pediatrics of the Pirogov, Russian National Research Medical University, Ministry of Health of Russian Federation, 125412 Moscow, Russia; (V.Y.V.); (S.G.V.); (I.Y.I.)
- Mental Health Research Center, 117152 Moscow, Russia
| | - Ivan Y. Iourov
- Veltischev Research and Clinical Institute for Pediatrics of the Pirogov, Russian National Research Medical University, Ministry of Health of Russian Federation, 125412 Moscow, Russia; (V.Y.V.); (S.G.V.); (I.Y.I.)
- Mental Health Research Center, 117152 Moscow, Russia
| | - Olga V. Sysoeva
- Laboratory of Human Higher Nervous Activity, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Science, 117485 Moscow, Russia; (A.K.N.); (G.V.P.); (A.B.R.)
| |
Collapse
|