1
|
Nguyen QA, Rubchinsky LL. Synaptic effects on the intermittent synchronization of gamma rhythms. Cogn Neurodyn 2024; 18:3821-3837. [PMID: 39712142 PMCID: PMC11655999 DOI: 10.1007/s11571-024-10150-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 05/29/2024] [Accepted: 06/27/2024] [Indexed: 12/24/2024] Open
Abstract
Synchronization of neural activity in the gamma frequency band is associated with various cognitive phenomena. Abnormalities of gamma synchronization may underlie symptoms of several neurological and psychiatric disorders such as schizophrenia and autism spectrum disorder. Properties of neural oscillations in the gamma band depend critically on the synaptic properties of the underlying circuits. This study explores how synaptic properties in pyramidal-interneuronal circuits affect not only the average synchronization strength but also the fine temporal patterning of neural synchrony. If two signals show only moderate synchrony strength, it may be possible to consider these dynamics as alternating between synchronized and desynchronized states. We use a model of connected circuits that produces pyramidal-interneuronal gamma oscillations to explore the temporal patterning of synchronized and desynchronized intervals. Changes in synaptic strength may alter the temporal patterning of synchronized dynamics (even if the average synchrony strength is not changed). Larger values of local synaptic connections promote longer desynchronization durations, while larger values of long-range synaptic connections promote shorter desynchronization durations. Furthermore, we show that circuits with different temporal patterning of synchronization may have different sensitivity to synaptic input. Thus, the alterations of synaptic strength may mediate physiological properties of neural circuits not only through change in the average synchrony level of gamma oscillations, but also through change in how synchrony is patterned in time over very short time scales.
Collapse
Affiliation(s)
- Quynh-Anh Nguyen
- Department of Mathematical Sciences, Indiana University Indianapolis, Indianapolis, IN 46202 USA
- Present Address: Department of Mathematical Sciences, University of Indianapolis, Indianapolis, IN 46227 USA
| | - Leonid L. Rubchinsky
- Department of Mathematical Sciences, Indiana University Indianapolis, Indianapolis, IN 46202 USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| |
Collapse
|
2
|
Gautam D, Shields A, Krepps E, Ummear Raza M, Sivarao DV. Click train elicited local gamma synchrony: differing performance and pharmacological responsivity of primary auditory and prefrontal cortices. Brain Res 2024; 1841:149091. [PMID: 38897535 DOI: 10.1016/j.brainres.2024.149091] [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: 02/22/2024] [Revised: 06/05/2024] [Accepted: 06/16/2024] [Indexed: 06/21/2024]
Abstract
Auditory neural networks in the brain naturally entrain to rhythmic stimuli. Such synchronization is an accessible index of local network performance as captured by EEG. Across species, click trains delivered ∼ 40 Hz show strong entrainment with primary auditory cortex (Actx) being a principal source. Imaging studies have revealed additional cortical sources, but it is unclear if they are functionally distinct. Since auditory processing evolves hierarchically, we hypothesized that local synchrony would differ between between primary and association cortices. In female SD rats (N = 12), we recorded 40 Hz click train-elicited gamma oscillations using epidural electrodes situated at two distinct sites; one above the prefrontal cortex (PFC) and another above the Actx, after dosing with saline (1 ml/kg, sc) or the NMDA antagonist, MK801 (0.025, 0.05 or 0.1 mpk), in a blocked crossover design. Post-saline, both regions showed a strong 40 Hz auditory steady state response (ASSR). The latencies for the N1 response were ∼ 16 ms (Actx) and ∼ 34 ms (PFC). Narrow band (38-42 Hz) gamma oscillations appeared rapidly (<40 ms from stim onset at Actx but in a more delayed fashion (∼200 ms) at PFC. MK801 augmented gamma synchrony at Actx while dose-dependently disrupting at the PFC. Event-related gamma (but not beta) coherence, an index of long-distance connectivity, was disrupted by MK801. In conclusion, local network gamma synchrony in a higher order association cortex performs differently from that of the primary auditory cortex. We discuss these findings in the context of evolving sound processing across the cortical hierarchy.
Collapse
Affiliation(s)
- Deepshila Gautam
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN, United States
| | - Abby Shields
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN, United States
| | - Emily Krepps
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN, United States
| | - Muhammad Ummear Raza
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN, United States
| | - Digavalli V Sivarao
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN, United States.
| |
Collapse
|
3
|
Khelfaoui H, Ibaceta-Gonzalez C, Angulo MC. Functional myelin in cognition and neurodevelopmental disorders. Cell Mol Life Sci 2024; 81:181. [PMID: 38615095 PMCID: PMC11016012 DOI: 10.1007/s00018-024-05222-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 03/18/2024] [Accepted: 03/30/2024] [Indexed: 04/15/2024]
Abstract
In vertebrates, oligodendrocytes (OLs) are glial cells of the central nervous system (CNS) responsible for the formation of the myelin sheath that surrounds the axons of neurons. The myelin sheath plays a crucial role in the transmission of neuronal information by promoting the rapid saltatory conduction of action potentials and providing neurons with structural and metabolic support. Saltatory conduction, first described in the peripheral nervous system (PNS), is now generally recognized as a universal evolutionary innovation to respond quickly to the environment: myelin helps us think and act fast. Nevertheless, the role of myelin in the central nervous system, especially in the brain, may not be primarily focused on accelerating conduction speed but rather on ensuring precision. Its principal function could be to coordinate various neuronal networks, promoting their synchronization through oscillations (or rhythms) relevant for specific information processing tasks. Interestingly, myelin has been directly involved in different types of cognitive processes relying on brain oscillations, and myelin plasticity is currently considered to be part of the fundamental mechanisms for memory formation and maintenance. However, despite ample evidence showing the involvement of myelin in cognition and neurodevelopmental disorders characterized by cognitive impairments, the link between myelin, brain oscillations, cognition and disease is not yet fully understood. In this review, we aim to highlight what is known and what remains to be explored to understand the role of myelin in high order brain processes.
Collapse
Affiliation(s)
- Hasni Khelfaoui
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, 75014, Paris, France
| | - Cristobal Ibaceta-Gonzalez
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, 75014, Paris, France
| | - Maria Cecilia Angulo
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, 75014, Paris, France.
- GHU-PARIS Psychiatrie Et Neurosciences, Hôpital Sainte Anne, 75014, Paris, France.
| |
Collapse
|
4
|
Ummear Raza M, Gautam D, Rorie D, Sivarao DV. Differential Effects of Clozapine and Haloperidol on the 40 Hz Auditory Steady State Response-mediated Phase Resetting in the Prefrontal Cortex of the Female Sprague Dawley Rat. Schizophr Bull 2023; 49:581-591. [PMID: 36691888 PMCID: PMC10154723 DOI: 10.1093/schbul/sbac203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
BACKGROUND Neural synchrony at gamma frequency (~40 Hz) is important for information processing and is disrupted in schizophrenia. From a drug development perspective, molecules that can improve local gamma synchrony are promising candidates for therapeutic development. HYPOTHESIS Given their differentiated clinical profile, clozapine, and haloperidol may have distinct effects on local gamma synchrony engendered by 40 Hz click trains, the so-called auditory steady-state response (ASSR). STUDY DESIGN Clozapine and haloperidol at doses known to mimic clinically relevant D2 receptor occupancy were evaluated using the ASSR in separate cohorts of female SD rats. RESULTS Clozapine (2.5-10 mg/kg, sc) robustly increased intertrial phase coherence (ITC), across all doses. Evoked response increased but less consistently. Background gamma activity, unrelated to the stimulus, showed a reduction at all doses. Closer scrutiny of the data indicated that clozapine accelerated gamma phase resetting. Thus, clozapine augmented auditory information processing in the gamma frequency range by reducing the background gamma, accelerating the gamma phase resetting and improving phase precision and signal power. Modest improvements in ITC were seen with Haloperidol (0.08 and 0.24 mg/kg, sc) without accelerating phase resetting. Evoked power was unaffected while background gamma was reduced at high doses only, which also caused catalepsy. CONCLUSIONS Using click-train evoked gamma synchrony as an index of local neural network function, we provide a plausible neurophysiological basis for the superior and differentiated profile of clozapine. These observations may provide a neurophysiological template for identifying new drug candidates with a therapeutic potential for treatment-resistant schizophrenia.
Collapse
Affiliation(s)
- Muhammad Ummear Raza
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN
| | - Deepshila Gautam
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN
| | - Dakota Rorie
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN
| | - Digavalli V Sivarao
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN
| |
Collapse
|
5
|
Jalewa J, Todd J, Michie PT, Hodgson DM, Harms L. The effect of schizophrenia risk factors on mismatch responses in a rat model. Psychophysiology 2023; 60:e14175. [PMID: 36087044 PMCID: PMC10909418 DOI: 10.1111/psyp.14175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 06/28/2022] [Accepted: 08/04/2022] [Indexed: 01/06/2023]
Abstract
Reduced mismatch negativity (MMN), a robust finding in schizophrenia, has prompted interest in MMN as a preclinical biomarker of schizophrenia. The rat brain can generate human-like mismatch responses (MMRs) which therefore enables the exploration of the neurobiology of reduced MMRs. Given epidemiological evidence that two developmental factors, maternal infection and adolescent cannabis use, increase the risk of schizophrenia, we determined the effect of these two developmental risk factors on rat MMR amplitude in different auditory contexts. MMRs were assessed in awake adult male and female Wistar rats that were offspring of pregnant dams treated with either a viral infection mimetic (poly I:C) inducing maternal immune activation (MIA) or saline control. In adolescence, subgroups of the prenatal treatment groups were exposed to either a synthetic cannabinoid (adolescent cannabinoid exposure: ACE) or vehicle. The context under which MMRs were obtained was manipulated by employing two different oddball paradigms, one that manipulated the physical difference between rare and common auditory stimuli, and another that manipulated the probability of the rare stimulus. The design of the multiple stimulus sequences across the two paradigms also allowed an investigation of context on MMRs to two identical stimulus sequences. Male offspring exposed to each of the risk factors for schizophrenia (MIA, ACE or both) showed a reduction in MMR, which was evident only in the probability paradigm, with no effects seen in the physical difference. Our findings highlight the importance of contextual factors induced by paradigm manipulations and sex for modeling schizophrenia-like MMN impairments in rats.
Collapse
Affiliation(s)
- Jaishree Jalewa
- School of Psychological Sciences, College of Engineering, Science and EnvironmentUniversity of NewcastleCallaghanNew South WalesAustralia
| | - Juanita Todd
- School of Psychological Sciences, College of Engineering, Science and EnvironmentUniversity of NewcastleCallaghanNew South WalesAustralia
- Hunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| | - Patricia T. Michie
- School of Psychological Sciences, College of Engineering, Science and EnvironmentUniversity of NewcastleCallaghanNew South WalesAustralia
- Hunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| | - Deborah M. Hodgson
- School of Psychological Sciences, College of Engineering, Science and EnvironmentUniversity of NewcastleCallaghanNew South WalesAustralia
- Hunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| | - Lauren Harms
- Hunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
- School of Biomedical Science and Pharmacy, College of Health, Medicine and WellbeingUniversity of NewcastleCallaghanNew South WalesAustralia
| |
Collapse
|
6
|
Mancini V, Rochas V, Seeber M, Roehri N, Rihs TA, Ferat V, Schneider M, Uhlhaas PJ, Eliez S, Michel CM. Aberrant Developmental Patterns of Gamma-Band Response and Long-Range Communication Disruption in Youths With 22q11.2 Deletion Syndrome. Am J Psychiatry 2022; 179:204-215. [PMID: 35236117 DOI: 10.1176/appi.ajp.2021.21020190] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Brain oscillations play a pivotal role in synchronizing responses of local and global ensembles of neurons. Patients with schizophrenia exhibit impairments in oscillatory response, which are thought to stem from abnormal maturation during critical developmental stages. Studying individuals at genetic risk for psychosis, such as 22q11.2 deletion carriers, from childhood to adulthood may provide insights into developmental abnormalities. METHODS The authors acquired 106 consecutive T1-weighted MR images and 40-Hz auditory steady-state responses (ASSRs) with high-density (256 channel) EEG in a group of 58 22q11.2 deletion carriers and 48 healthy control subjects. ASSRs were analyzed with 1) time-frequency analysis using Morlet wavelet decomposition, 2) intertrial phase coherence (ITPC), and 3) theta-gamma phase-amplitude coupling estimated in the source space between brain regions activated by the ASSRs. Additionally, volumetric analyses were performed with FreeSurfer. Subanalyses were conducted in deletion carriers who endorsed psychotic symptoms and in subgroups with different age bins. RESULTS Deletion carriers had decreased theta and late-latency 40-Hz ASSRs and phase synchronization compared with control subjects. Deletion carriers with psychotic symptoms displayed a further reduction of gamma-band response, decreased ITPC, and decreased top-down modulation of gamma-band response in the auditory cortex. Reduced gamma-band response was correlated with the atrophy of auditory cortex in individuals with psychotic symptoms. In addition, a linear increase of theta and gamma power from childhood to adulthood was found in control subjects but not in deletion carriers. CONCLUSIONS The results suggest that while all deletion carriers exhibit decreased gamma-band response, more severe local and long-range communication abnormalities are associated with the emergence of psychotic symptoms and gray matter loss. Additionally, the lack of age-related changes in deletion carriers indexes a potential developmental impairment in circuits underlying the maturation of neural oscillations during adolescence. The progressive disruption of gamma-band response in 22q11.2 deletion syndrome supports a developmental perspective toward understanding and treating psychotic disorders.
Collapse
Affiliation(s)
- Valentina Mancini
- Developmental Imaging and Psychopathology Laboratory (Mancini, Schneider, Eliez) and Department of Genetic Medicine and Development (Eliez), University of Geneva School of Medicine, Geneva; Functional Brain Mapping Laboratory, Department of Basic Neurosciences, University of Geneva, Geneva (Rochas, Seeber, Roehri, Rihs, Ferat, Michel); Clinical Psychology Unit for Intellectual and Developmental Disabilities, Faculty of Psychology and Educational Sciences, University of Geneva, Geneva (Schneider); Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, Scotland (Uhlhaas); Department of Child and Adolescent Psychiatry, Psychosomatic Medicine and Psychotherapy, Charité Universitätsmedizin, Berlin (Uhlhaas); Center for Biomedical Imaging (CIBM) of Lausanne and Geneva (Michel)
| | - Vincent Rochas
- Developmental Imaging and Psychopathology Laboratory (Mancini, Schneider, Eliez) and Department of Genetic Medicine and Development (Eliez), University of Geneva School of Medicine, Geneva; Functional Brain Mapping Laboratory, Department of Basic Neurosciences, University of Geneva, Geneva (Rochas, Seeber, Roehri, Rihs, Ferat, Michel); Clinical Psychology Unit for Intellectual and Developmental Disabilities, Faculty of Psychology and Educational Sciences, University of Geneva, Geneva (Schneider); Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, Scotland (Uhlhaas); Department of Child and Adolescent Psychiatry, Psychosomatic Medicine and Psychotherapy, Charité Universitätsmedizin, Berlin (Uhlhaas); Center for Biomedical Imaging (CIBM) of Lausanne and Geneva (Michel)
| | - Martin Seeber
- Developmental Imaging and Psychopathology Laboratory (Mancini, Schneider, Eliez) and Department of Genetic Medicine and Development (Eliez), University of Geneva School of Medicine, Geneva; Functional Brain Mapping Laboratory, Department of Basic Neurosciences, University of Geneva, Geneva (Rochas, Seeber, Roehri, Rihs, Ferat, Michel); Clinical Psychology Unit for Intellectual and Developmental Disabilities, Faculty of Psychology and Educational Sciences, University of Geneva, Geneva (Schneider); Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, Scotland (Uhlhaas); Department of Child and Adolescent Psychiatry, Psychosomatic Medicine and Psychotherapy, Charité Universitätsmedizin, Berlin (Uhlhaas); Center for Biomedical Imaging (CIBM) of Lausanne and Geneva (Michel)
| | - Nicolas Roehri
- Developmental Imaging and Psychopathology Laboratory (Mancini, Schneider, Eliez) and Department of Genetic Medicine and Development (Eliez), University of Geneva School of Medicine, Geneva; Functional Brain Mapping Laboratory, Department of Basic Neurosciences, University of Geneva, Geneva (Rochas, Seeber, Roehri, Rihs, Ferat, Michel); Clinical Psychology Unit for Intellectual and Developmental Disabilities, Faculty of Psychology and Educational Sciences, University of Geneva, Geneva (Schneider); Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, Scotland (Uhlhaas); Department of Child and Adolescent Psychiatry, Psychosomatic Medicine and Psychotherapy, Charité Universitätsmedizin, Berlin (Uhlhaas); Center for Biomedical Imaging (CIBM) of Lausanne and Geneva (Michel)
| | - Tonia A Rihs
- Developmental Imaging and Psychopathology Laboratory (Mancini, Schneider, Eliez) and Department of Genetic Medicine and Development (Eliez), University of Geneva School of Medicine, Geneva; Functional Brain Mapping Laboratory, Department of Basic Neurosciences, University of Geneva, Geneva (Rochas, Seeber, Roehri, Rihs, Ferat, Michel); Clinical Psychology Unit for Intellectual and Developmental Disabilities, Faculty of Psychology and Educational Sciences, University of Geneva, Geneva (Schneider); Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, Scotland (Uhlhaas); Department of Child and Adolescent Psychiatry, Psychosomatic Medicine and Psychotherapy, Charité Universitätsmedizin, Berlin (Uhlhaas); Center for Biomedical Imaging (CIBM) of Lausanne and Geneva (Michel)
| | - Victor Ferat
- Developmental Imaging and Psychopathology Laboratory (Mancini, Schneider, Eliez) and Department of Genetic Medicine and Development (Eliez), University of Geneva School of Medicine, Geneva; Functional Brain Mapping Laboratory, Department of Basic Neurosciences, University of Geneva, Geneva (Rochas, Seeber, Roehri, Rihs, Ferat, Michel); Clinical Psychology Unit for Intellectual and Developmental Disabilities, Faculty of Psychology and Educational Sciences, University of Geneva, Geneva (Schneider); Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, Scotland (Uhlhaas); Department of Child and Adolescent Psychiatry, Psychosomatic Medicine and Psychotherapy, Charité Universitätsmedizin, Berlin (Uhlhaas); Center for Biomedical Imaging (CIBM) of Lausanne and Geneva (Michel)
| | - Maude Schneider
- Developmental Imaging and Psychopathology Laboratory (Mancini, Schneider, Eliez) and Department of Genetic Medicine and Development (Eliez), University of Geneva School of Medicine, Geneva; Functional Brain Mapping Laboratory, Department of Basic Neurosciences, University of Geneva, Geneva (Rochas, Seeber, Roehri, Rihs, Ferat, Michel); Clinical Psychology Unit for Intellectual and Developmental Disabilities, Faculty of Psychology and Educational Sciences, University of Geneva, Geneva (Schneider); Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, Scotland (Uhlhaas); Department of Child and Adolescent Psychiatry, Psychosomatic Medicine and Psychotherapy, Charité Universitätsmedizin, Berlin (Uhlhaas); Center for Biomedical Imaging (CIBM) of Lausanne and Geneva (Michel)
| | - Peter J Uhlhaas
- Developmental Imaging and Psychopathology Laboratory (Mancini, Schneider, Eliez) and Department of Genetic Medicine and Development (Eliez), University of Geneva School of Medicine, Geneva; Functional Brain Mapping Laboratory, Department of Basic Neurosciences, University of Geneva, Geneva (Rochas, Seeber, Roehri, Rihs, Ferat, Michel); Clinical Psychology Unit for Intellectual and Developmental Disabilities, Faculty of Psychology and Educational Sciences, University of Geneva, Geneva (Schneider); Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, Scotland (Uhlhaas); Department of Child and Adolescent Psychiatry, Psychosomatic Medicine and Psychotherapy, Charité Universitätsmedizin, Berlin (Uhlhaas); Center for Biomedical Imaging (CIBM) of Lausanne and Geneva (Michel)
| | - Stephan Eliez
- Developmental Imaging and Psychopathology Laboratory (Mancini, Schneider, Eliez) and Department of Genetic Medicine and Development (Eliez), University of Geneva School of Medicine, Geneva; Functional Brain Mapping Laboratory, Department of Basic Neurosciences, University of Geneva, Geneva (Rochas, Seeber, Roehri, Rihs, Ferat, Michel); Clinical Psychology Unit for Intellectual and Developmental Disabilities, Faculty of Psychology and Educational Sciences, University of Geneva, Geneva (Schneider); Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, Scotland (Uhlhaas); Department of Child and Adolescent Psychiatry, Psychosomatic Medicine and Psychotherapy, Charité Universitätsmedizin, Berlin (Uhlhaas); Center for Biomedical Imaging (CIBM) of Lausanne and Geneva (Michel)
| | - Christoph M Michel
- Developmental Imaging and Psychopathology Laboratory (Mancini, Schneider, Eliez) and Department of Genetic Medicine and Development (Eliez), University of Geneva School of Medicine, Geneva; Functional Brain Mapping Laboratory, Department of Basic Neurosciences, University of Geneva, Geneva (Rochas, Seeber, Roehri, Rihs, Ferat, Michel); Clinical Psychology Unit for Intellectual and Developmental Disabilities, Faculty of Psychology and Educational Sciences, University of Geneva, Geneva (Schneider); Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, Scotland (Uhlhaas); Department of Child and Adolescent Psychiatry, Psychosomatic Medicine and Psychotherapy, Charité Universitätsmedizin, Berlin (Uhlhaas); Center for Biomedical Imaging (CIBM) of Lausanne and Geneva (Michel)
| |
Collapse
|
7
|
Complex Organ Construction from Human Pluripotent Stem Cells for Biological Research and Disease Modeling with New Emerging Techniques. Int J Mol Sci 2021; 22:ijms221910184. [PMID: 34638524 PMCID: PMC8508560 DOI: 10.3390/ijms221910184] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/17/2021] [Accepted: 09/20/2021] [Indexed: 12/13/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) are grouped into two cell types; embryonic stem cells (hESCs) and induced pluripotent stem cells (hiPSCs). hESCs have provided multiple powerful platforms to study human biology, including human development and diseases; however, there were difficulties in the establishment of hESCs from human embryo and concerns over its ethical issues. The discovery of hiPSCs has expanded to various applications in no time because hiPSCs had already overcome these problems. Many hPSC-based studies have been performed using two-dimensional monocellular culture methods at the cellular level. However, in many physiological and pathophysiological conditions, intra- and inter-organ interactions play an essential role, which has hampered the establishment of an appropriate study model. Therefore, the application of recently developed technologies, such as three-dimensional organoids, bioengineering, and organ-on-a-chip technology, has great potential for constructing multicellular tissues, generating the functional organs from hPSCs, and recapitulating complex tissue functions for better biological research and disease modeling. Moreover, emerging techniques, such as single-cell transcriptomics, spatial transcriptomics, and artificial intelligence (AI) allowed for a denser and more precise analysis of such heterogeneous and complex tissues. Here, we review the applications of hPSCs to construct complex organs and discuss further prospects of disease modeling and drug discovery based on these PSC-derived organs.
Collapse
|
8
|
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
|
9
|
Raza MU, Sivarao DV. Test-retest reliability of tone- and 40 Hz train-evoked gamma oscillations in female rats and their sensitivity to low-dose NMDA channel blockade. Psychopharmacology (Berl) 2021; 238:2325-2334. [PMID: 33944972 DOI: 10.1007/s00213-021-05856-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 04/15/2021] [Indexed: 10/21/2022]
Abstract
RATIONALE Schizophrenia patients consistently show deficits in sensory-evoked broadband gamma oscillations and click-evoked entrainment at 40 Hz, called the 40-Hz auditory steady-state response (ASSR). Since such evoked oscillations depend on cortical N-methyl D-aspartic acid (NMDA)-mediated network activity, they can serve as pharmacodynamic biomarkers in the preclinical and clinical development of drug candidates engaging these circuits. However, there are few test-retest reliability data in preclinical species, a prerequisite for within-subject testing paradigms. OBJECTIVE We investigated the long-term psychometric stability of these measures in a rodent model. METHODS Female rats with chronic epidural implants were used to record tone- and 40 Hz click-evoked responses at multiple time points and across six sessions, spread over 3 weeks. We assessed reliability using intraclass correlation coefficients (ICC). Separately, we used mixed-effects ANOVA to examine time and session effects. Individual subject variability was determined using the coefficient of variation (CV). Lastly, to illustrate the importance of long-term measure stability for within-subject testing design, we used low to moderate doses of an NMDA antagonist MK801 (0.025-0.15 mg/kg) to disrupt the evoked response. RESULTS We found that 40-Hz ASSR showed good reliability (ICC=0.60-0.75), while the reliability of tone-evoked gamma ranged from poor to good (0.33-0.67). We noted time but no session effects. Subjects showed a lower variance for ASSR over tone-evoked gamma. Both measures were dose-dependently attenuated by NMDA antagonism. CONCLUSION Overall, while both evoked gamma measures use NMDA transmission, 40-Hz ASSR showed superior psychometric properties of higher ICC and lower CV, relative to tone-evoked gamma.
Collapse
Affiliation(s)
- Muhammad Ummear Raza
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, VA Building 7, Room 324, Maple Ave, Johnson City, TN, 37604, USA
| | - Digavalli V Sivarao
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, VA Building 7, Room 324, Maple Ave, Johnson City, TN, 37604, USA.
| |
Collapse
|
10
|
Local Oscillatory Brain Dynamics of Mind Wandering in Schizophrenia. Brain Sci 2021; 11:brainsci11070910. [PMID: 34356145 PMCID: PMC8304325 DOI: 10.3390/brainsci11070910] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 11/29/2022] Open
Abstract
A number of studies have focused on brain dynamics underlying mind wandering (MW) states in healthy people. However, there is limited understanding of how the oscillatory dynamics accompanying MW states and task-focused states are characterized in clinical populations. In this study, we explored EEG local synchrony of MW associated with schizophrenia, under the premise that changes in attention that arise during MW are associated with a different pattern of brain activity. To this end, we measured the power of EEG oscillations in different frequency bands, recorded while participants watched short video clips. In the group of participants diagnosed with schizophrenia, the power in MW states was significantly lower than during task-focused states, mainly in the frontal and posterior regions. However, in the group of healthy controls, the differences in power between the task-focused and MW states occurred exclusively in the posterior region. Furthermore, the power of the frequency bands during MW and during episodes of task-focused attention correlated with cognitive variables such as processing speed and working memory. These findings on dynamic changes of local synchronization in different frequency bands and areas of the cortex can improve our understanding of mental disorders, such as schizophrenia.
Collapse
|
11
|
Investigating neurophysiological markers of impaired cognition in schizophrenia. Schizophr Res 2021; 233:34-43. [PMID: 34225025 DOI: 10.1016/j.schres.2021.06.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 04/21/2021] [Accepted: 06/21/2021] [Indexed: 01/22/2023]
Abstract
Cognitive impairment is highly prevalent in schizophrenia and treatment options are severely limited. A greater understanding of the pathophysiology of impaired cognition would have broad implications, including for the development of effective treatments. In the current study we used a multimodal approach to identify neurophysiological markers of cognitive impairment in schizophrenia. Fifty-seven participants (30 schizophrenia, 27 controls) underwent neurobiological assessment (electroencephalography [EEG] and Transcranial Magnetic Stimulation combined with EEG [TMS-EEG]) and assessment of cognitive functioning using an n-back task and the MATRICS Consensus Cognitive Battery. Neurobiological outcome measures included oscillatory power during a 2-back task, TMS-related oscillations and TMS-evoked potentials (TEPs). Cognitive outcome measures were d prime and accurate reaction time on the 2-back and MATRICS domain scores. Compared to healthy controls, participants with schizophrenia showed significantly reduced theta oscillations in response to TMS, and trend level decreases in task-related theta and cortical reactivity (i.e. reduced N100 and N40 TEPs). Participants with schizophrenia also showed significantly impaired cognitive performance across all measures. Correlational analysis identified significant associations between cortical reactivity and TMS-related oscillations in both groups; and trend level associations between task-related oscillations and impaired cognition in schizophrenia. The current study provides experimental support for possible neurophysiological markers of cognitive impairment in schizophrenia. The potential implications of these findings, including for treatment development, are discussed.
Collapse
|
12
|
Delavari F, Sandini C, Zöller D, Mancini V, Bortolin K, Schneider M, Van De Ville D, Eliez S. Dysmaturation Observed as Altered Hippocampal Functional Connectivity at Rest Is Associated With the Emergence of Positive Psychotic Symptoms in Patients With 22q11 Deletion Syndrome. Biol Psychiatry 2021; 90:58-68. [PMID: 33771350 DOI: 10.1016/j.biopsych.2020.12.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 12/03/2020] [Accepted: 12/21/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Hippocampal alterations are among the most replicated neuroimaging findings across the psychosis spectrum. Moreover, there is strong translational evidence that preserving the maturation of hippocampal networks in mice models prevents the progression of cognitive deficits. However, the developmental trajectory of hippocampal functional connectivity (HFC) and its contribution to psychosis is not well characterized in the human population. 22q11 deletion syndrome (22q11DS) offers a unique model for characterizing early neural correlates of schizophrenia. METHODS We acquired resting-state functional magnetic resonance imaging in 242 longitudinally repeated scans from 84 patients with 22q11DS (30 with moderate to severe positive psychotic symptoms) and 94 healthy control subjects in the age span of 6 to 32 years. We obtained bilateral hippocampus to whole-brain functional connectivity and employed a novel longitudinal multivariate approach by means of partial least squares correlation to evaluate the developmental trajectory of HFC across groups. RESULTS Relative to control subjects, patients with 22q11DS failed to increase HFC with frontal regions such as the dorsal part of the anterior cingulate cortex, prefrontal cortex, and supplementary motor area. Concurrently, carriers of the deletion had abnormally higher HFC with subcortical dopaminergic areas. Remarkably, this aberrant maturation of HFC was more prominent during midadolescence and was mainly driven by patients exhibiting subthreshold positive psychotic symptoms. CONCLUSIONS Our findings suggest a critical period of prefrontal cortex-hippocampal-striatal circuit dysmaturation, particularly during late adolescence, which in light of current translation evidence could be a target for short-term interventions to potentially achieve long-lasting rescue of circuit dysfunctions associated with psychosis.
Collapse
Affiliation(s)
- Farnaz Delavari
- Developmental Imaging and Psychopathology Laboratory, University of Geneva School of Medicine, Geneva, Switzerland; Medical Image Processing Laboratory, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.
| | - Corrado Sandini
- Developmental Imaging and Psychopathology Laboratory, University of Geneva School of Medicine, Geneva, Switzerland
| | - Daniela Zöller
- Developmental Imaging and Psychopathology Laboratory, University of Geneva School of Medicine, Geneva, Switzerland; Medical Image Processing Laboratory, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Valentina Mancini
- Developmental Imaging and Psychopathology Laboratory, University of Geneva School of Medicine, Geneva, Switzerland
| | - Karin Bortolin
- Developmental Imaging and Psychopathology Laboratory, University of Geneva School of Medicine, Geneva, Switzerland; Medical Image Processing Laboratory, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Maude Schneider
- Developmental Imaging and Psychopathology Laboratory, University of Geneva School of Medicine, Geneva, Switzerland; Department of Neuroscience, Center for Contextual Psychiatry, Research Group Psychiatry, KU Leuven, Leuven, Belgium
| | - Dimitri Van De Ville
- Department of Radiology and Medical Informatics, University of Geneva, Geneva, Switzerland; Medical Image Processing Laboratory, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Stephan Eliez
- Developmental Imaging and Psychopathology Laboratory, University of Geneva School of Medicine, Geneva, Switzerland; Department of Genetic Medicine and Development, University of Geneva School of Medicine, Geneva, Switzerland
| |
Collapse
|
13
|
Nguyen QA, Rubchinsky LL. Temporal patterns of synchrony in a pyramidal-interneuron gamma (PING) network. CHAOS (WOODBURY, N.Y.) 2021; 31:043134. [PMID: 34251236 DOI: 10.1063/5.0042451] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 04/05/2021] [Indexed: 06/13/2023]
Abstract
Synchronization in neural systems plays an important role in many brain functions. Synchronization in the gamma frequency band (30-100 Hz) is involved in a variety of cognitive phenomena; abnormalities of the gamma synchronization are found in schizophrenia and autism spectrum disorder. Frequently, the strength of synchronization is not high, and synchronization is intermittent even on short time scales (few cycles of oscillations). That is, the network exhibits intervals of synchronization followed by intervals of desynchronization. Neural circuit dynamics may show different distributions of desynchronization durations even if the synchronization strength is fixed. We use a conductance-based neural network exhibiting pyramidal-interneuron gamma rhythm to study the temporal patterning of synchronized neural oscillations. We found that changes in the synaptic strength (as well as changes in the membrane kinetics) can alter the temporal patterning of synchrony. Moreover, we found that the changes in the temporal pattern of synchrony may be independent of the changes in the average synchrony strength. Even though the temporal patterning may vary, there is a tendency for dynamics with short (although potentially numerous) desynchronizations, similar to what was observed in experimental studies of neural synchronization in the brain. Recent studies suggested that the short desynchronizations dynamics may facilitate the formation and the breakup of transient neural assemblies. Thus, the results of this study suggest that changes of synaptic strength may alter the temporal patterning of the gamma synchronization as to make the neural networks more efficient in the formation of neural assemblies and the facilitation of cognitive phenomena.
Collapse
Affiliation(s)
- Quynh-Anh Nguyen
- Department of Mathematical Sciences, Indiana University Purdue University Indianapolis, Indianapolis, Indiana 46202, USA
| | - Leonid L Rubchinsky
- Department of Mathematical Sciences, Indiana University Purdue University Indianapolis, Indianapolis, Indiana 46202, USA
| |
Collapse
|
14
|
Morales L, Castro-Robles B, Abellán A, Desfilis E, Medina L. A novel telencephalon-opto-hypothalamic morphogenetic domain coexpressing Foxg1 and Otp produces most of the glutamatergic neurons of the medial extended amygdala. J Comp Neurol 2021; 529:2418-2449. [PMID: 33386618 DOI: 10.1002/cne.25103] [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: 07/02/2020] [Revised: 12/28/2020] [Accepted: 12/29/2020] [Indexed: 12/14/2022]
Abstract
Deficits in social cognition and behavior are a hallmark of many psychiatric disorders. The medial extended amygdala, including the medial amygdala and the medial bed nucleus of the stria terminalis, is a key component of functional networks involved in sociality. However, this nuclear complex is highly heterogeneous and contains numerous GABAergic and glutamatergic neuron subpopulations. Deciphering the connections of different neurons is essential in order to understand how this structure regulates different aspects of sociality, and it is necessary to evaluate their differential implication in distinct mental disorders. Developmental studies in different vertebrates are offering new venues to understand neuronal diversity of the medial extended amygdala and are helping to establish a relation between the embryonic origin and molecular signature of distinct neurons with the functional subcircuits in which they are engaged. These studies have provided many details on the distinct GABAergic neurons of the medial extended amygdala, but information on the glutamatergic neurons is still scarce. Using an Otp-eGFP transgenic mouse and multiple fluorescent labeling, we show that most glutamatergic neurons of the medial extended amygdala originate in a distinct telencephalon-opto-hypothalamic embryonic domain (TOH), located at the transition between telencephalon and hypothalamus, which produces Otp-lineage neurons expressing the telencephalic marker Foxg1 but not Nkx2.1 during development. These glutamatergic cells include a subpopulation of projection neurons of the medial amygdala, which activation has been previously shown to promote autistic-like behavior. Our data open new venues for studying the implication of this neuron subtype in neurodevelopmental disorders producing social deficits.
Collapse
Affiliation(s)
- Lorena Morales
- Laboratory of Evolutionary and Developmental Neurobiology, Lleida's Institute for Biomedical Research-Dr. Pifarré Foundation (IRBLleida), Catalonia, Spain
| | - Beatriz Castro-Robles
- Laboratory of Cerebrovascular, Neurodegenerative and Neuro-oncology Diseases, Research Unit, Complejo Hospitalario Universitario de Albacete, Castilla-La Mancha, Spain
| | - Antonio Abellán
- Laboratory of Evolutionary and Developmental Neurobiology, Lleida's Institute for Biomedical Research-Dr. Pifarré Foundation (IRBLleida), Catalonia, Spain
| | - Ester Desfilis
- Laboratory of Evolutionary and Developmental Neurobiology, Lleida's Institute for Biomedical Research-Dr. Pifarré Foundation (IRBLleida), Catalonia, Spain
| | - Loreta Medina
- Laboratory of Evolutionary and Developmental Neurobiology, Lleida's Institute for Biomedical Research-Dr. Pifarré Foundation (IRBLleida), Catalonia, Spain
| |
Collapse
|
15
|
Dysbindin-1 regulates mitochondrial fission and gamma oscillations. Mol Psychiatry 2021; 26:4633-4651. [PMID: 33589740 PMCID: PMC8364574 DOI: 10.1038/s41380-021-01038-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 01/08/2021] [Accepted: 01/25/2021] [Indexed: 12/11/2022]
Abstract
Mitochondria are cellular ATP generators. They are dynamic structures undergoing fission and fusion. While much is known about the mitochondrial fission machinery, the mechanism of initiating fission and the significance of fission to neurophysiology are largely unclear. Gamma oscillations are synchronized neural activities that impose a great energy challenge to synapses. The cellular mechanism of fueling gamma oscillations has yet to be defined. Here, we show that dysbindin-1, a protein decreased in the brain of individuals with schizophrenia, is required for neural activity-induced fission by promoting Drp1 oligomerization. This process is engaged by gamma-frequency activities and in turn, supports gamma oscillations. Gamma oscillations and novel object recognition are impaired in dysbindin-1 null mice. These defects can be ameliorated by increasing mitochondrial fission. These findings identify a molecular mechanism for activity-induced mitochondrial fission, a role of mitochondrial fission in gamma oscillations, and mitochondrial fission as a potential target for improving cognitive functions.
Collapse
|
16
|
Dolleman-van der Weel MJ, Witter MP. The thalamic midline nucleus reuniens: potential relevance for schizophrenia and epilepsy. Neurosci Biobehav Rev 2020; 119:422-439. [PMID: 33031816 DOI: 10.1016/j.neubiorev.2020.09.033] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 09/03/2020] [Accepted: 09/28/2020] [Indexed: 01/08/2023]
Abstract
Anatomical, electrophysiological and behavioral studies in rodents have shown that the thalamic midline nucleus reuniens (RE) is a crucial link in the communication between hippocampal formation (HIP, i.e., CA1, subiculum) and medial prefrontal cortex (mPFC), important structures for cognitive and executive functions. A common feature in neurodevelopmental and neurodegenerative brain diseases is a dysfunctional connectivity/communication between HIP and mPFC, and disturbances in the cognitive domain. Therefore, it is assumed that aberrant functioning of RE may contribute to behavioral/cognitive impairments in brain diseases characterized by cortico-thalamo-hippocampal circuit dysfunctions. In the human brain the connections of RE are largely unknown. Yet, recent studies have found important similarities in the functional connectivity of HIP-mPFC-RE in humans and rodents, making cautious extrapolating experimental findings from animal models to humans justifiable. The focus of this review is on a potential involvement of RE in schizophrenia and epilepsy.
Collapse
Affiliation(s)
- M J Dolleman-van der Weel
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, NTNU Norwegian University of Science and Technology, Trondheim NO-7491, Norway.
| | - M P Witter
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, NTNU Norwegian University of Science and Technology, Trondheim NO-7491, Norway.
| |
Collapse
|
17
|
Rürup L, Mathes B, Schmiedt-Fehr C, Wienke AS, Özerdem A, Brand A, Basar-Eroglu C. Altered gamma and theta oscillations during multistable perception in schizophrenia. Int J Psychophysiol 2020; 155:127-139. [DOI: 10.1016/j.ijpsycho.2020.06.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 06/01/2020] [Accepted: 06/05/2020] [Indexed: 12/24/2022]
|
18
|
Guirado R, Perez-Rando M, Ferragud A, Gutierrez-Castellanos N, Umemori J, Carceller H, Nacher J, Castillo-Gómez E. A Critical Period for Prefrontal Network Configurations Underlying Psychiatric Disorders and Addiction. Front Behav Neurosci 2020; 14:51. [PMID: 32317945 PMCID: PMC7155216 DOI: 10.3389/fnbeh.2020.00051] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 03/19/2020] [Indexed: 12/24/2022] Open
Abstract
The medial prefrontal cortex (mPFC) has been classically defined as the brain region responsible for higher cognitive functions, including the decision-making process. Ample information has been gathered during the last 40 years in an attempt to understand how it works. We now know extensively about the connectivity of this region and its relationship with neuromodulatory ascending projection areas, such as the dorsal raphe nucleus (DRN) or the ventral tegmental area (VTA). Both areas are well-known regulators of the reward-based decision-making process and hence likely to be involved in processes like evidence integration, impulsivity or addiction biology, but also in helping us to predict the valence of our future actions: i.e., what is “good” and what is “bad.” Here we propose a hypothesis of a critical period, during which the inputs of the mPFC compete for target innervation, establishing specific prefrontal network configurations in the adult brain. We discuss how these different prefrontal configurations are linked to brain diseases such as addiction or neuropsychiatric disorders, and especially how drug abuse and other events during early life stages might lead to the formation of more vulnerable prefrontal network configurations. Finally, we show different promising pharmacological approaches that, when combined with the appropriate stimuli, will be able to re-establish these functional prefrontocortical configurations during adulthood.
Collapse
Affiliation(s)
- Ramon Guirado
- Neurobiology Unit, Department of Cell Biology, Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de Valencia, Valencia, Spain.,Neuroscience Center, University of Helsinki, Helsinki, Finland.,Spanish National Network for Research in Mental Health, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.,Dirección General de Universidades, Gobierno de Aragón, Zaragoza, Spain
| | - Marta Perez-Rando
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Antonio Ferragud
- Department of Psychology, Cambridge University, Cambridge, United Kingdom
| | | | - Juzoh Umemori
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Hector Carceller
- Neurobiology Unit, Department of Cell Biology, Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de Valencia, Valencia, Spain
| | - Juan Nacher
- Neurobiology Unit, Department of Cell Biology, Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de Valencia, Valencia, Spain.,Spanish National Network for Research in Mental Health, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.,Fundación Investigación Hospital Clínico de Valencia, INCLIVA, Valencia, Spain
| | - Esther Castillo-Gómez
- Spanish National Network for Research in Mental Health, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.,Department of Medicine, School of Medical Sciences, Universitat Jaume I, Valencia, Spain
| |
Collapse
|
19
|
Ehlers CL, Phillips E, Wills D, Benedict J, Sanchez-Alavez M. Phase locking of event-related oscillations is decreased in both young adult humans and rats with a history of adolescent alcohol exposure. Addict Biol 2020; 25:e12732. [PMID: 30884076 PMCID: PMC6751029 DOI: 10.1111/adb.12732] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 01/23/2019] [Accepted: 01/24/2019] [Indexed: 01/25/2023]
Abstract
Alcohol exposure typically begins in adolescence, and frequent binge drinking has been associated with health risk behaviors including alcohol use disorders (AUDs). Few studies have documented the effects of a history of adolescent binge drinking on neurophysiological consequences in young adulthood. Synchrony of phase (phase locking (PL)) of event-related oscillations (EROs) within and between different brain areas reflects communication exchange between neural networks and is a sensitive measure of adolescent development in both rats and humans, and thus may be a good translational measure of the potential harmful effects of alcohol exposure during adolescence. In this study, EROs were collected from 1041 young adults of Mexican American and American Indian ancestry (age 18-30 years) with and without a history of adolescent binge drinking (five drinks for boys and four for girls per occasion at least once per month) and in 74 young adult rats with and without a history of 5 weeks of adolescent alcohol vapor exposure. PL of theta and beta frequencies between frontal and parietal cortex were estimated using an auditory-oddball paradigm in the rats and a visual facial expression paradigm in the humans. Significantly lower PL between frontal and parietal cortices in the theta frequencies was seen in both the humans and the rats with a history of adolescent alcohol exposure as compared with their controls. These findings suggest that alcohol exposure during adolescence may result in decreases in synchrony between cortical neuronal networks, suggesting a developmental delay, in young adult humans and in rats.
Collapse
Affiliation(s)
- Cindy L Ehlers
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | - Evie Phillips
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | - Derek Wills
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | - Jessica Benedict
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | | |
Collapse
|
20
|
Kawe TNJ, Shadli SM, McNaughton N. Higuchi's fractal dimension, but not frontal or posterior alpha asymmetry, predicts PID-5 anxiousness more than depressivity. Sci Rep 2019; 9:19666. [PMID: 31873184 PMCID: PMC6928148 DOI: 10.1038/s41598-019-56229-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 12/06/2019] [Indexed: 12/28/2022] Open
Abstract
Depression is a major cause of health disability. EEG measures may provide one or more economical biomarkers for the diagnosis of depression. Here we compared frontal alpha asymmetry (FAA), posterior alpha asymmetry (PAA), and Higuchi's fractal dimension (HFD) for their capacity to predict PID-5 depressivity and for the specificity of these predictions relative to PID-5 anxiousness. University students provided 8 or 10 minutes of resting EEG and PID-5 depressivity and PID-5 anxiousness questionnaire scores. FAA and PAA had no significant correlations with the measures at any electrode pair. There were distinct frontal and posterior factors underlying HFD that correlated significantly with anxiousness and with each other. Posterior HFD also correlated significantly with depressivity, though this was weaker than the correlation with anxiousness. The portion of depressivity variance accounted for by posterior HFD was not unique but shared with anxiousness. Inclusion of anxiety disorder patients into the sample rendered the frontal factor somewhat more predictive than the posterior one but generally strengthened the prior conclusions. Contrary to our predictions, none of our measures specifically predicted depressivity. Previous reports of links with depression may involve confounds with concurrent anxiety. Indeed, HFD may be a better measure of anxiety than depression; and its previous linkage to depression may be due to a confound between the two, given the high incidence of depression in cases of severe anxiety.
Collapse
Affiliation(s)
- Tame N J Kawe
- Department of Psychology, University of Otago, Dunedin, New Zealand
| | - Shabah M Shadli
- Department of Psychology, University of Otago, Dunedin, New Zealand
| | - Neil McNaughton
- Department of Psychology, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
21
|
Long-Lasting Rescue of Network and Cognitive Dysfunction in a Genetic Schizophrenia Model. Cell 2019; 178:1387-1402.e14. [DOI: 10.1016/j.cell.2019.07.023] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 06/01/2019] [Accepted: 07/11/2019] [Indexed: 01/03/2023]
|
22
|
Abnormal synchronization of functional and structural networks in schizophrenia. Brain Imaging Behav 2019; 14:2232-2241. [PMID: 31376115 DOI: 10.1007/s11682-019-00175-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Synchronization is believed to play an important role in information processing of the brain. Mounting evidence supports the hypothesis that schizophrenia is related to impaired neural synchrony. However, most previous studies characterize brain synchronization from the perspective of temporal coordination of distributed neural activity, rather than network properties. Our aim was to investigate the network synchronization alterations in schizophrenia using publically available data. Resting-state functional magnetic resonance imaging (fMRI) and diffusion tensor imaging (DTI) were performed in 96 schizophrenia patients and 120 healthy controls. The whole-brain functional and structural networks were constructed and analyzed using graph theoretical approaches. Inter-group differences in network synchronization were investigated. Both the binary and weighted functional networks of schizophrenia patients exhibited decreased synchronizability (increased eigenratio) than those of healthy controls. With respect to the structural binary networks, schizophrenia patients showed a trend towards excessive synchronizability (decreased eigenratio). In addition, the excessive synchronizability of the structural binary networks was associated with more severe negative symptoms in schizophrenia patients. Our findings provide novel biological evidence that schizophrenia involves a disruption of neural synchrony from the perspective of network properties.
Collapse
|
23
|
Habermacher C, Angulo MC, Benamer N. Glutamate versus GABA in neuron-oligodendroglia communication. Glia 2019; 67:2092-2106. [PMID: 30957306 DOI: 10.1002/glia.23618] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 02/28/2019] [Accepted: 03/19/2019] [Indexed: 12/20/2022]
Abstract
In the central nervous system (CNS), myelin sheaths around axons are formed by glial cells named oligodendrocytes (OLs). In turn, OLs are generated by oligodendrocyte precursor cells (OPCs) during postnatal development and in adults, according to a process that depends on the proliferation and differentiation of these progenitors. The maturation of OL lineage cells as well as myelination by OLs are complex and highly regulated processes in the CNS. OPCs and OLs express an array of receptors for neurotransmitters, in particular for the two main CNS neurotransmitters glutamate and GABA, and are therefore endowed with the capacity to respond to neuronal activity. Initial studies in cell cultures demonstrated that both glutamate and GABA signaling mechanisms play important roles in OL lineage cell development and function. However, much remains to be learned about the communication of glutamatergic and GABAergic neurons with oligodendroglia in vivo. This review focuses on recent major advances in our understanding of the neuron-oligodendroglia communication mediated by glutamate and GABA in the CNS, and highlights the present controversies in the field. We discuss the expression, activation modes and potential roles of synaptic and extrasynaptic receptors along OL lineage progression. We review the properties of OPC synaptic connectivity with presynaptic glutamatergic and GABAergic neurons in the brain and consider the implication of glutamate and GABA signaling in activity-driven adaptive myelination.
Collapse
Affiliation(s)
- Chloé Habermacher
- Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Paris, France.,Université Paris Descartes, Paris, France
| | - María C Angulo
- Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Paris, France.,Université Paris Descartes, Paris, France
| | - Najate Benamer
- Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Paris, France.,Université Paris Descartes, Paris, France
| |
Collapse
|
24
|
Lucas KM, Warrington J, Lewis TJ, Lewis JE. Neuronal Dynamics Underlying Communication Signals in a Weakly Electric Fish: Implications for Connectivity in a Pacemaker Network. Neuroscience 2019; 401:21-34. [PMID: 30641115 DOI: 10.1016/j.neuroscience.2019.01.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 12/19/2018] [Accepted: 01/04/2019] [Indexed: 11/19/2022]
Abstract
Neuronal networks can produce stable oscillations and synchrony that are under tight control yet flexible enough to rapidly switch between dynamical states. The pacemaker nucleus in the weakly electric fish comprises a network of electrically coupled neurons that fire synchronously at high frequency. This activity sets the timing for an oscillating electric organ discharge with the lowest cycle-to-cycle variability of all known biological oscillators. Despite this high temporal precision, pacemaker activity is behaviorally modulated on millisecond time-scales for the generation of electrocommunication signals. The network mechanisms that allow for this combination of stability and flexibility are not well understood. In this study, we use an in vitro pacemaker preparation from Apteronotus leptorhynchus to characterize the neural responses elicited by the synaptic inputs underlying electrocommunication. These responses involve a variable increase in firing frequency and a prominent desynchronization of neurons that recovers within 5 oscillation cycles. Using a previously developed computational model of the pacemaker network, we show that the frequency changes and rapid resynchronization observed experimentally are most easily explained when model neurons are interconnected more densely and with higher coupling strengths than suggested by published data. We suggest that the pacemaker network achieves both stability and flexibility by balancing coupling strength with interconnectivity and that variation in these network features may provide a substrate for species-specific evolution of electrocommunication signals.
Collapse
Affiliation(s)
- Kathleen M Lucas
- Department of Biology, University of Ottawa, Ottawa K1N 6N5, Canada
| | - Julie Warrington
- Department of Biology, University of Ottawa, Ottawa K1N 6N5, Canada
| | - Timothy J Lewis
- Department of Mathematics, University of California Davis, Davis, CA 95616, USA
| | - John E Lewis
- Department of Biology, University of Ottawa, Ottawa K1N 6N5, Canada; University of Ottawa Brain and Mind Research Institute, Ottawa K1N 6N5, Canada.
| |
Collapse
|
25
|
Parvalbumin-containing GABA cells and schizophrenia: experimental model based on targeted gene delivery through adeno-associated viruses. Behav Pharmacol 2018; 28:630-641. [PMID: 29120948 DOI: 10.1097/fbp.0000000000000360] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Understanding the contribution of transmitter systems in behavioural pharmacology has a long tradition. Multiple techniques such as transmitter-specific lesions, and also localized administration of pharmacological toxins including agonists and antagonists of selected receptors have been applied. More recently, modern genetic tools have permitted cell-type selective interferences, for example by expression of light-sensitive channels followed by optogenetic stimulation in behaviourally meaningful settings or by engineered channels termed DREADDS that respond to peripherally administered drugs. We here took a similar approach and employed a Cre recombinase-dependent viral delivery system (adeno-associated virus) to express tetanus toxin light chain (TeLc) and thus, block neural transmission specifically in parvalbumin-positive (PV+) neurons of the limbic and infralimbic prefrontal circuitry. PV-TeLc cohorts presented with normal circadian activity as recorded in PhenoTyper home cages, but a reproducible increase in anxiety was extracted in both the open field and light-dark box. Interestingly, working memory assessed in a spontaneous alternation Y-maze task was impaired in PV-TeLc mice. We also recorded local field potentials from a separate cohort and found no global changes in brain activity, but found a behaviourally relevant lack of modulation in the gamma spectral band. These anomalies are reminiscent of endophenotypes of schizophrenia and appear to be critically dependent on GABAergic signalling through PV neurones. At the same time, these observations validate the use of viral vector delivery and its expression in Cre-lines as a useful tool for understanding the role of selective components of the brain in behaviour and the underpinning physiology.
Collapse
|
26
|
Phase-Amplitude Coupling of the Electroencephalogram in the Auditory Cortex in Schizophrenia. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2017; 3:69-76. [PMID: 29397081 DOI: 10.1016/j.bpsc.2017.09.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 09/01/2017] [Accepted: 09/03/2017] [Indexed: 01/13/2023]
Abstract
BACKGROUND Cross-frequency interactions may coordinate neural circuits operating at different frequencies. While neural oscillations associated with particular circuits in schizophrenia (SZ) are impaired, few studies have examined cross-frequency interactions. Here we examined phase-amplitude coupling (PAC) in the electroencephalograms of individuals with SZ and healthy control subjects (HCs). We computed PAC during the baseline period of 40-Hz auditory steady-state stimulation and rest. We hypothesized that subjects with SZ would show abnormal theta/gamma coupling during stimulation, especially in the left auditory cortex, and coupling with high frequencies would be higher during stimulation than during rest. METHODS We reanalyzed data from 18 subjects with SZ and 18 HCs. Auditory cortex electroencephalogram activity was estimated using dipole source localization. PAC was computed using the debiased PAC measure, calculated with the generalized Morse wavelet transform. PAC clusters were identified using cluster-corrected permutation testing and interrogated in analyses of variance with correction for multiple tests. RESULTS Overall, coupling of high beta and gamma amplitude was higher during the auditory steady-state response, while alpha/beta PAC was higher during rest. Theta/alpha PAC was higher in subjects with SZ than in HCs. Theta/gamma PAC was lateralized to the left hemisphere in HCs but was not lateralized in subjects with SZ. CONCLUSIONS PAC involving high frequencies was state dependent and not abnormal in SZ. Increased theta/alpha PAC in subjects with SZ was consistent with other evidence of increased low-frequency activity. Hemispheric lateralization of theta/gamma PAC was reduced in subjects with SZ, consistent with evidence for left hemisphere auditory cortex abnormalities in subjects with SZ. PAC may reveal new insights into neural circuitry abnormalities in SZ and other neuropsychiatric disorders.
Collapse
|
27
|
Atanasova T, Kharybina Z, Kaarela T, Huupponen J, Luchkina NV, Taira T, Lauri SE. GluA4 Dependent Plasticity Mechanisms Contribute to Developmental Synchronization of the CA3-CA1 Circuitry in the Hippocampus. Neurochem Res 2017; 44:562-571. [PMID: 28856535 DOI: 10.1007/s11064-017-2392-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 08/14/2017] [Accepted: 08/16/2017] [Indexed: 10/24/2022]
Abstract
During the course of development, molecular mechanisms underlying activity-dependent synaptic plasticity change considerably. At immature CA3-CA1 synapses in the hippocampus, PKA-driven synaptic insertion of GluA4 AMPA receptors is the predominant mechanism for synaptic strengthening. However, the physiological significance of the developmentally restricted GluA4-dependent plasticity mechanisms is poorly understood. Here we have used microelectrode array (MEA) recordings in GluA4 deficient slice cultures to study the role of GluA4 in early development of the hippocampal circuit function. We find that during the first week in culture (DIV2-6) when GluA4 expression is restricted to pyramidal neurons, loss of GluA4 has no effect on the overall excitability of the immature network, but significantly impairs synchronization of the CA3 and CA1 neuronal populations. In the absence of GluA4, the temporal correlation of the population spiking activity between CA3-CA1 neurons was significantly lower as compared to wild-types at DIV6. Our data show that synapse-level defects in transmission and plasticity mechanisms are efficiently compensated for to normalize population firing rate at the immature hippocampal network. However, lack of the plasticity mechanisms typical for the immature synapses may perturb functional coupling between neuronal sub-populations, a defect frequently implicated in the context of developmentally originating neuropsychiatric disorders.
Collapse
Affiliation(s)
- Tsvetomira Atanasova
- Neuroscience Center, University of Helsinki, Helsinki, Finland.,Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
| | - Zoya Kharybina
- Neuroscience Center, University of Helsinki, Helsinki, Finland.,Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
| | - Tiina Kaarela
- Neuroscience Center, University of Helsinki, Helsinki, Finland.,Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
| | - Johanna Huupponen
- Neuroscience Center, University of Helsinki, Helsinki, Finland.,Department of Biosciences, University of Helsinki, Helsinki, Finland
| | - Natalia V Luchkina
- Neuroscience Center, University of Helsinki, Helsinki, Finland.,Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
| | - Tomi Taira
- Neuroscience Center, University of Helsinki, Helsinki, Finland.,Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
| | - Sari E Lauri
- Neuroscience Center, University of Helsinki, Helsinki, Finland. .,Department of Biosciences, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
28
|
Allen WE, Kauvar IV, Chen MZ, Richman EB, Yang SJ, Chan K, Gradinaru V, Deverman BE, Luo L, Deisseroth K. Global Representations of Goal-Directed Behavior in Distinct Cell Types of Mouse Neocortex. Neuron 2017; 94:891-907.e6. [PMID: 28521139 PMCID: PMC5723385 DOI: 10.1016/j.neuron.2017.04.017] [Citation(s) in RCA: 240] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 01/26/2017] [Accepted: 04/11/2017] [Indexed: 11/27/2022]
Abstract
The successful planning and execution of adaptive behaviors in mammals may require long-range coordination of neural networks throughout cerebral cortex. The neuronal implementation of signals that could orchestrate cortex-wide activity remains unclear. Here, we develop and apply methods for cortex-wide Ca2+ imaging in mice performing decision-making behavior and identify a global cortical representation of task engagement encoded in the activity dynamics of both single cells and superficial neuropil distributed across the majority of dorsal cortex. The activity of multiple molecularly defined cell types was found to reflect this representation with type-specific dynamics. Focal optogenetic inhibition tiled across cortex revealed a crucial role for frontal cortex in triggering this cortex-wide phenomenon; local inhibition of this region blocked both the cortex-wide response to task-initiating cues and the voluntary behavior. These findings reveal cell-type-specific processes in cortex for globally representing goal-directed behavior and identify a major cortical node that gates the global broadcast of task-related information.
Collapse
Affiliation(s)
- William E Allen
- Neurosciences Graduate Program, Stanford University, Stanford, CA 94305, USA; Department of Biology, Stanford University, Stanford, CA 94305, USA; Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Isaac V Kauvar
- Electrical Engineering Graduate Program, Stanford University, Stanford, CA 94305, USA; Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Michael Z Chen
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Ethan B Richman
- Neurosciences Graduate Program, Stanford University, Stanford, CA 94305, USA; Department of Biology, Stanford University, Stanford, CA 94305, USA; Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Samuel J Yang
- Electrical Engineering Graduate Program, Stanford University, Stanford, CA 94305, USA; Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Ken Chan
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Viviana Gradinaru
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Benjamin E Deverman
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Liqun Luo
- Department of Biology, Stanford University, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA.
| | - Karl Deisseroth
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA; Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
29
|
Ahn S, Rubchinsky LL. Potential Mechanisms and Functions of Intermittent Neural Synchronization. Front Comput Neurosci 2017; 11:44. [PMID: 28611618 PMCID: PMC5447717 DOI: 10.3389/fncom.2017.00044] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 05/15/2017] [Indexed: 11/26/2022] Open
Abstract
Neural synchronization is believed to play an important role in different brain functions. Synchrony in cortical and subcortical circuits is frequently variable in time and not perfect. Few long intervals of desynchronized dynamics may be functionally different from many short desynchronized intervals although the average synchrony may be the same. Recent analysis of imperfect synchrony in different neural systems reported one common feature: neural oscillations may go out of synchrony frequently, but primarily for a short time interval. This study explores potential mechanisms and functional advantages of this short desynchronizations dynamics using computational neuroscience techniques. We show that short desynchronizations are exhibited in coupled neurons if their delayed rectifier potassium current has relatively large values of the voltage-dependent activation time-constant. The delayed activation of potassium current is associated with generation of quickly-rising action potential. This “spikiness” is a very general property of neurons. This may explain why very different neural systems exhibit short desynchronization dynamics. We also show how the distribution of desynchronization durations may be independent of the synchronization strength. Finally, we show that short desynchronization dynamics requires weaker synaptic input to reach a pre-set synchrony level. Thus, this dynamics allows for efficient regulation of synchrony and may promote efficient formation of synchronous neural assemblies.
Collapse
Affiliation(s)
- Sungwoo Ahn
- Department of Mathematical Sciences, Indiana University Purdue University IndianapolisIndianapolis, IN, United States
| | - Leonid L Rubchinsky
- Department of Mathematical Sciences, Indiana University Purdue University IndianapolisIndianapolis, IN, United States.,Stark Neurosciences Research Institute, Indiana University School of MedicineIndianapolis, IN, United States
| |
Collapse
|
30
|
Alamian G, Hincapié AS, Combrisson E, Thiery T, Martel V, Althukov D, Jerbi K. Alterations of Intrinsic Brain Connectivity Patterns in Depression and Bipolar Disorders: A Critical Assessment of Magnetoencephalography-Based Evidence. Front Psychiatry 2017; 8:41. [PMID: 28367127 PMCID: PMC5355450 DOI: 10.3389/fpsyt.2017.00041] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 02/28/2017] [Indexed: 12/21/2022] Open
Abstract
Despite being the object of a thriving field of clinical research, the investigation of intrinsic brain network alterations in psychiatric illnesses is still in its early days. Because the pathological alterations are predominantly probed using functional magnetic resonance imaging (fMRI), many questions about the electrophysiological bases of resting-state alterations in psychiatric disorders, particularly among mood disorder patients, remain unanswered. Alongside important research using electroencephalography (EEG), the specific recent contributions and future promise of magnetoencephalography (MEG) in this field are not fully recognized and valued. Here, we provide a critical review of recent findings from MEG resting-state connectivity within major depressive disorder (MDD) and bipolar disorder (BD). The clinical MEG resting-state results are compared with those previously reported with fMRI and EEG. Taken together, MEG appears to be a promising but still critically underexploited technique to unravel the neurophysiological mechanisms that mediate abnormal (both hyper- and hypo-) connectivity patterns involved in MDD and BD. In particular, a major strength of MEG is its ability to provide source-space estimations of neuromagnetic long-range rhythmic synchronization at various frequencies (i.e., oscillatory coupling). The reviewed literature highlights the relevance of probing local and interregional rhythmic synchronization to explore the pathophysiological underpinnings of each disorder. However, before we can fully take advantage of MEG connectivity analyses in psychiatry, several limitations inherent to MEG connectivity analyses need to be understood and taken into account. Thus, we also discuss current methodological challenges and outline paths for future research. MEG resting-state studies provide an important window onto perturbed spontaneous oscillatory brain networks and hence supply an important complement to fMRI-based resting-state measurements in psychiatric populations.
Collapse
Affiliation(s)
- Golnoush Alamian
- Department of Psychology, University of Montreal , Montreal, QC , Canada
| | - Ana-Sofía Hincapié
- Department of Psychology, University of Montreal, Montreal, QC, Canada; Department of Computer Science, Pontificia Universidad Católica de Chile, Santiago de Chile, Chile; Interdisciplinary Center for Neurosciences, School of Psychology, Pontificia Universidad Católica de Chile, Santiago de Chile, Chile
| | - Etienne Combrisson
- Department of Psychology, University of Montreal, Montreal, QC, Canada; Center of Research and Innovation in Sport, Mental Processes and Motor Performance, University Claude Bernard Lyon I, University of Lyon, Villeurbanne, France; Brain Dynamics and Cognition, Lyon Neuroscience Research Center, INSERM U1028, UMR 5292, University of Lyon, Villeurbanne, France
| | - Thomas Thiery
- Department of Psychology, University of Montreal , Montreal, QC , Canada
| | - Véronique Martel
- Department of Psychology, University of Montreal , Montreal, QC , Canada
| | - Dmitrii Althukov
- Department of Psychology, University of Montreal, Montreal, QC, Canada; Department of Computer Sciences, National Research Institution Higher School of Economics, Moscow, Russia; MEG Center, Moscow State University of Pedagogics and Education, Moscow, Russia
| | - Karim Jerbi
- Department of Psychology, University of Montreal, Montreal, QC, Canada; Centre de recherche de l'Institut universitaire en santé mentale de Montréal, Montreal, QC, Canada
| |
Collapse
|
31
|
Abstract
Defined genetic models based on human pluripotent stem cells have opened new avenues for understanding disease mechanisms and drug screening. Many of these models assume cell-autonomous mechanisms of disease but it is possible that disease phenotypes or drug responses will only be evident if all cellular and extracellular components of a tissue are present and functionally mature. To derive optimal benefit from such models, complex multicellular structures with vascular components that mimic tissue niches will thus likely be necessary. Here we consider emerging research creating human tissue mimics and provide some recommendations for moving the field forward.
Collapse
|
32
|
Using model systems to understand errant plasticity mechanisms in psychiatric disorders. Nat Neurosci 2016; 19:1418-1425. [PMID: 27786180 DOI: 10.1038/nn.4413] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 09/13/2016] [Indexed: 02/08/2023]
Abstract
In vivo model systems are a critical tool for gaining insight into the pathology underlying psychiatric disorders. Although modern functional imaging tools allow study of brain correlates of behavior in clinical groups and genome-wide association studies are beginning to uncover the complex genetic architecture of psychiatric disorders, there is less understanding of pathology at intervening levels of organization. Several psychiatric disorders derive from pathological neural plasticity, and studying the mechanisms that underlie these processes, including reinforcement learning and spike-timing-dependent plasticity, requires the use of animals. It will be particularly important to understand how individual differences in plasticity mechanisms at a cellular level confer resilience on some but lead to disease in others.
Collapse
|
33
|
Sigurdsson T. Neural circuit dysfunction in schizophrenia: Insights from animal models. Neuroscience 2016; 321:42-65. [DOI: 10.1016/j.neuroscience.2015.06.059] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 06/15/2015] [Accepted: 06/26/2015] [Indexed: 12/17/2022]
|
34
|
Fuccillo MV. Striatal Circuits as a Common Node for Autism Pathophysiology. Front Neurosci 2016; 10:27. [PMID: 26903795 PMCID: PMC4746330 DOI: 10.3389/fnins.2016.00027] [Citation(s) in RCA: 190] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 01/22/2016] [Indexed: 12/17/2022] Open
Abstract
Autism spectrum disorders (ASD) are characterized by two seemingly unrelated symptom domains-deficits in social interactions and restrictive, repetitive patterns of behavioral output. Whether the diverse nature of ASD symptomatology represents distributed dysfunction of brain networks or abnormalities within specific neural circuits is unclear. Striatal dysfunction is postulated to underlie the repetitive motor behaviors seen in ASD, and neurological and brain-imaging studies have supported this assumption. However, as our appreciation of striatal function expands to include regulation of behavioral flexibility, motivational state, goal-directed learning, and attention, we consider whether alterations in striatal physiology are a central node mediating a range of autism-associated behaviors, including social and cognitive deficits that are hallmarks of the disease. This review investigates multiple genetic mouse models of ASD to explore whether abnormalities in striatal circuits constitute a common pathophysiological mechanism in the development of autism-related behaviors. Despite the heterogeneity of genetic insult investigated, numerous genetic ASD models display alterations in the structure and function of striatal circuits, as well as abnormal behaviors including repetitive grooming, stereotypic motor routines, deficits in social interaction and decision-making. Comparative analysis in rodents provides a unique opportunity to leverage growing genetic association data to reveal canonical neural circuits whose dysfunction directly contributes to discrete aspects of ASD symptomatology. The description of such circuits could provide both organizing principles for understanding the complex genetic etiology of ASD as well as novel treatment routes. Furthermore, this focus on striatal mechanisms of behavioral regulation may also prove useful for exploring the pathogenesis of other neuropsychiatric diseases, which display overlapping behavioral deficits with ASD.
Collapse
Affiliation(s)
- Marc V. Fuccillo
- Department of Neuroscience, Perelman School of Medicine, University of PennsylvaniaPhiladelphia, PA, USA
| |
Collapse
|
35
|
Sigurdsson T, Duvarci S. Hippocampal-Prefrontal Interactions in Cognition, Behavior and Psychiatric Disease. Front Syst Neurosci 2016; 9:190. [PMID: 26858612 PMCID: PMC4727104 DOI: 10.3389/fnsys.2015.00190] [Citation(s) in RCA: 165] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 12/23/2015] [Indexed: 12/22/2022] Open
Abstract
The hippocampus and prefrontal cortex (PFC) have long been known to play a central role in various behavioral and cognitive functions. More recently, electrophysiological and functional imaging studies have begun to examine how interactions between the two structures contribute to behavior during various tasks. At the same time, it has become clear that hippocampal-prefrontal interactions are disrupted in psychiatric disease and may contribute to their pathophysiology. These impairments have most frequently been observed in schizophrenia, a disease that has long been associated with hippocampal and prefrontal dysfunction. Studies in animal models of the illness have also begun to relate disruptions in hippocampal-prefrontal interactions to the various risk factors and pathophysiological mechanisms of the illness. The goal of this review is to summarize what is known about the role of hippocampal-prefrontal interactions in normal brain function and compare how these interactions are disrupted in schizophrenia patients and animal models of the disease. Outstanding questions for future research on the role of hippocampal-prefrontal interactions in both healthy brain function and disease states are also discussed.
Collapse
Affiliation(s)
- Torfi Sigurdsson
- Institute of Neurophysiology, Neuroscience Center, Goethe University FrankfurtFrankfurt, Germany
| | - Sevil Duvarci
- Institute of Neurophysiology, Neuroscience Center, Goethe University FrankfurtFrankfurt, Germany
| |
Collapse
|
36
|
White RS, Siegel SJ. Cellular and circuit models of increased resting-state network gamma activity in schizophrenia. Neuroscience 2015; 321:66-76. [PMID: 26577758 DOI: 10.1016/j.neuroscience.2015.11.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Revised: 10/27/2015] [Accepted: 11/04/2015] [Indexed: 02/05/2023]
Abstract
Schizophrenia (SCZ) is a disorder characterized by positive symptoms (hallucinations, delusions), negative symptoms (blunted affect, alogia, reduced sociability, and anhedonia), as well as persistent cognitive deficits (memory, concentration, and learning). While the biology underlying subjective experiences is difficult to study, abnormalities in electroencephalographic (EEG) measures offer a means to dissect potential circuit and cellular changes in brain function. EEG is indispensable for studying cerebral information processing due to the introduction of techniques for the decomposition of event-related activity into its frequency components. Specifically, brain activity in the gamma frequency range (30-80Hz) is thought to underlie cognitive function and may be used as an endophenotype to aid in diagnosis and treatment of SCZ. In this review we address evidence indicating that there is increased resting-state gamma power in SCZ. We address how modeling this aspect of the illness in animals may help treatment development as well as providing insights into the etiology of SCZ.
Collapse
Affiliation(s)
- R S White
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - S J Siegel
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, United States.
| |
Collapse
|
37
|
Heckenast JR, Wilkinson LS, Jones MW. Decoding Advances in Psychiatric Genetics: A Focus on Neural Circuits in Rodent Models. ADVANCES IN GENETICS 2015; 92:75-106. [PMID: 26639916 DOI: 10.1016/bs.adgen.2015.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Appropriately powered genome-wide association studies combined with deep-sequencing technologies offer the prospect of real progress in revealing the complex biological underpinnings of schizophrenia and other psychiatric disorders. Meanwhile, recent developments in genome engineering, including CRISPR, constitute better tools to move forward with investigating these genetic leads. This review aims to assess how these advances can inform the development of animal models for psychiatric disease, with a focus on schizophrenia and in vivo electrophysiological circuit-level measures with high potential as disease biomarkers.
Collapse
Affiliation(s)
- Julia R Heckenast
- School of Psychology, Cardiff University, Cardiff, UK; School of Medicine, Cardiff University, Cardiff, UK; Behavioural Genetics Group, MRC Centre for Neuropsychiatric Genetics and Genomics, Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, UK
| | - Lawrence S Wilkinson
- School of Psychology, Cardiff University, Cardiff, UK; School of Medicine, Cardiff University, Cardiff, UK; Behavioural Genetics Group, MRC Centre for Neuropsychiatric Genetics and Genomics, Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, UK
| | - Matthew W Jones
- School of Physiology and Pharmacology, University of Bristol, University Walk, Bristol, UK
| |
Collapse
|
38
|
Cohen SM, Tsien RW, Goff DC, Halassa MM. The impact of NMDA receptor hypofunction on GABAergic neurons in the pathophysiology of schizophrenia. Schizophr Res 2015; 167:98-107. [PMID: 25583246 PMCID: PMC4724170 DOI: 10.1016/j.schres.2014.12.026] [Citation(s) in RCA: 173] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 11/25/2014] [Accepted: 12/18/2014] [Indexed: 02/07/2023]
Abstract
While the dopamine hypothesis has dominated schizophrenia research for several decades, more recent studies have highlighted the role of fast synaptic transmitters and their receptors in schizophrenia etiology. Here we review evidence that schizophrenia is associated with a reduction in N-methyl-d-aspartate receptor (NMDAR) function. By highlighting postmortem, neuroimaging and electrophysiological studies, we provide evidence for preferential disruption of GABAergic circuits in the context of NMDAR hypo-activity states. The functional relationship between NMDARs and GABAergic neurons is realized at the molecular, cellular, microcircuit and systems levels. A synthesis of findings across these levels explains how NMDA-mediated inhibitory dysfunction may lead to aberrant interactions among brain regions, accounting for key clinical features of schizophrenia. This synthesis of schizophrenia unifies observations from diverse fields and may help chart pathways for developing novel diagnostics and therapeutics.
Collapse
Affiliation(s)
- Samuel M. Cohen
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY 10016, USA
| | - Richard W. Tsien
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY 10016, USA
| | - Donald C. Goff
- Department of Psychiatry, NYU Langone Medical Center, 550 First Avenue, New York City, NY 10016, USA
,Nathan Kline Institute for Psychiatric Research, 140 Old Orangeburg Road, Orangeburg, NY 10962, USA
| | - Michael M. Halassa
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY 10016, USA
,Department of Psychiatry, NYU Langone Medical Center, 550 First Avenue, New York City, NY 10016, USA
,To whom correspondence should be addressed:
| |
Collapse
|
39
|
Solanka L, van Rossum MCW, Nolan MF. Noise promotes independent control of gamma oscillations and grid firing within recurrent attractor networks. eLife 2015; 4. [PMID: 26146940 PMCID: PMC4508578 DOI: 10.7554/elife.06444] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 07/04/2015] [Indexed: 11/25/2022] Open
Abstract
Neural computations underlying cognitive functions require calibration of the strength of excitatory and inhibitory synaptic connections and are associated with modulation of gamma frequency oscillations in network activity. However, principles relating gamma oscillations, synaptic strength and circuit computations are unclear. We address this in attractor network models that account for grid firing and theta-nested gamma oscillations in the medial entorhinal cortex. We show that moderate intrinsic noise massively increases the range of synaptic strengths supporting gamma oscillations and grid computation. With moderate noise, variation in excitatory or inhibitory synaptic strength tunes the amplitude and frequency of gamma activity without disrupting grid firing. This beneficial role for noise results from disruption of epileptic-like network states. Thus, moderate noise promotes independent control of multiplexed firing rate- and gamma-based computational mechanisms. Our results have implications for tuning of normal circuit function and for disorders associated with changes in gamma oscillations and synaptic strength. DOI:http://dx.doi.org/10.7554/eLife.06444.001 When electrodes are placed on the scalp, or lowered into the brain itself, rhythmic waves of electrical activity are seen that reflect the coordinated firing of large numbers of neurons. The pattern of the waves varies between different brain regions, and according to what the animal or person is doing. During sleep and quiet wakefulness, slower brain waves predominate, whereas faster waves called gamma oscillations emerge during cognition—the act of processing knowledge. Gamma waves can be readily detected in a region of the brain called the medial entorhinal cortex (MEC). This brain region is also known for its role in forming the spatial memories that allow an individual to remember how to navigate around an area they have previously visited. Individual MEC cells increase their firing rates whenever an individual is at specific locations. When these locations are plotted in two dimensions, they form a hexagonal grid: this ‘grid cell map’ enables the animal to keep track of its position as it navigates through its environment. To determine how MEC neurons can simultaneously encode spatial locations and generate the gamma waves implicated in cognition, Solanka et al. have used supercomputing to simulate the activity of more than 1.5 million connections between MEC cells. Changing the strength of these connections had different effects on the ability of the MEC to produce gamma waves or spatial maps. However, adjusting the model to include random fluctuations in neuronal firing, or ‘noise’, was beneficial for both types of output. This is partly because noise prevented neuronal firing from becoming excessively synchronized, which would otherwise have caused seizures. Although noise is generally regarded as disruptive, the results of Solanka et al. suggest that it helps the MEC to perform its two distinct roles. Specifically, the presence of noise enables relatively small changes in the strength of the connections between neurons to alter gamma waves—and thus affect cognition—without disrupting the neurons' ability to encode spatial locations. Given that noise reduces the likelihood of seizures, the results also raise the possibility that introducing noise into the brain in a controlled way could have therapeutic benefits for individuals with epilepsy. DOI:http://dx.doi.org/10.7554/eLife.06444.002
Collapse
Affiliation(s)
- Lukas Solanka
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Matthew F Nolan
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
40
|
Rosen AM, Spellman T, Gordon JA. Electrophysiological endophenotypes in rodent models of schizophrenia and psychosis. Biol Psychiatry 2015; 77:1041-9. [PMID: 25910423 PMCID: PMC4444383 DOI: 10.1016/j.biopsych.2015.03.021] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 03/03/2015] [Accepted: 03/23/2015] [Indexed: 02/06/2023]
Abstract
Schizophrenia is caused by a diverse array of risk factors and results in a similarly diverse set of symptoms. Electrophysiological endophenotypes lie between risks and symptoms and have the potential to link the two. Electrophysiological studies in rodent models, described here, demonstrate that widely differing risk factors result in a similar set of core electrophysiological endophenotypes, suggesting the possibility of a shared neurobiological substrate.
Collapse
Affiliation(s)
- Andrew M. Rosen
- Department of Psychiatry, College of Physicians and Surgeons Columbia University New York, NY 10032
| | - Timothy Spellman
- Department of Physiology, College of Physicians and Surgeons Columbia University New York, NY 10032
| | - Joshua A. Gordon
- Department of Psychiatry, College of Physicians and Surgeons Columbia University New York, NY 10032,Division of Integrative Neuroscience New York State Psychiatric Institute New York NY 10032,Correspondence to: Joshua A. Gordon 1051 Riverside Drive Unit 87 Kolb Annex Room 140 New York, NY 10032 Ph. 646 774-7116 Fax. 646 774-7101
| |
Collapse
|
41
|
Venkatasubramanian G. Understanding schizophrenia as a disorder of consciousness: biological correlates and translational implications from quantum theory perspectives. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE : THE OFFICIAL SCIENTIFIC JOURNAL OF THE KOREAN COLLEGE OF NEUROPSYCHOPHARMACOLOGY 2015; 13:36-47. [PMID: 25912536 PMCID: PMC4423156 DOI: 10.9758/cpn.2015.13.1.36] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 12/07/2014] [Accepted: 12/08/2014] [Indexed: 11/18/2022]
Abstract
From neurophenomenological perspectives, schizophrenia has been conceptualized as "a disorder with heterogeneous manifestations that can be integrally understood to involve fundamental perturbations in consciousness". While these theoretical constructs based on consciousness facilitate understanding the 'gestalt' of schizophrenia, systematic research to unravel translational implications of these models is warranted. To address this, one needs to begin with exploration of plausible biological underpinnings of "perturbed consciousness" in schizophrenia. In this context, an attractive proposition to understand the biology of consciousness is "the orchestrated object reduction (Orch-OR) theory" which invokes quantum processes in the microtubules of neurons. The Orch-OR model is particularly important for understanding schizophrenia especially due to the shared 'scaffold' of microtubules. The initial sections of this review focus on the compelling evidence to support the view that "schizophrenia is a disorder of consciousness" through critical summary of the studies that have demonstrated self-abnormalities, aberrant time perception as well as dysfunctional intentional binding in this disorder. Subsequently, these findings are linked with 'Orch-OR theory' through the research evidence for aberrant neural oscillations as well as microtubule abnormalities observed in schizophrenia. Further sections emphasize the applicability and translational implications of Orch-OR theory in the context of schizophrenia and elucidate the relevance of quantum biology to understand the origins of this puzzling disorder as "fundamental disturbances in consciousness".
Collapse
Affiliation(s)
- Ganesan Venkatasubramanian
- The Schizophrenia Clinic, Department of Psychiatry and Translational Psychiatry Laboratory, Neurobiology Research Centre, National Institute of Mental Health and Neurosciences, Bangalore,
India
| |
Collapse
|
42
|
Ho SM, Topol A, Brennand KJ. From "directed differentiation" to "neuronal induction": modeling neuropsychiatric disease. Biomark Insights 2015; 10:31-41. [PMID: 26045654 PMCID: PMC4444490 DOI: 10.4137/bmi.s20066] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 02/22/2015] [Accepted: 02/24/2015] [Indexed: 11/23/2022] Open
Abstract
Aberrant behavior and function of neurons are believed to be the primary causes of most neurological diseases and psychiatric disorders. Human postmortem samples have limited availability and, while they provide clues to the state of the brain after a prolonged illness, they offer limited insight into the factors contributing to disease onset. Conversely, animal models cannot recapitulate the polygenic origins of neuropsychiatric disease. Novel methods, such as somatic cell reprogramming, deliver nearly limitless numbers of pathogenic human neurons for the study of the mechanism of neuropsychiatric disease initiation and progression. First, this article reviews the advent of human induced pluripotent stem cell (hiPSC) technology and introduces two major methods, “directed differentiation” and “neuronal induction,” by which it is now possible to generate neurons for modeling neuropsychiatric disease. Second, it discusses the recent applications, and the limitations, of these technologies to in vitro studies of psychiatric disorders.
Collapse
Affiliation(s)
- Seok-Man Ho
- Icahn School of Medicine at Mount Sinai, Department of Psychiatry, New York, NY, USA
| | - Aaron Topol
- Icahn School of Medicine at Mount Sinai, Department of Psychiatry, New York, NY, USA
| | - Kristen J Brennand
- Icahn School of Medicine at Mount Sinai, Department of Psychiatry, New York, NY, USA
| |
Collapse
|
43
|
Crabtree GW, Gogos JA. Synaptic plasticity, neural circuits, and the emerging role of altered short-term information processing in schizophrenia. Front Synaptic Neurosci 2014; 6:28. [PMID: 25505409 PMCID: PMC4243504 DOI: 10.3389/fnsyn.2014.00028] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 10/22/2014] [Indexed: 01/01/2023] Open
Abstract
Synaptic plasticity alters the strength of information flow between presynaptic and postsynaptic neurons and thus modifies the likelihood that action potentials in a presynaptic neuron will lead to an action potential in a postsynaptic neuron. As such, synaptic plasticity and pathological changes in synaptic plasticity impact the synaptic computation which controls the information flow through the neural microcircuits responsible for the complex information processing necessary to drive adaptive behaviors. As current theories of neuropsychiatric disease suggest that distinct dysfunctions in neural circuit performance may critically underlie the unique symptoms of these diseases, pathological alterations in synaptic plasticity mechanisms may be fundamental to the disease process. Here we consider mechanisms of both short-term and long-term plasticity of synaptic transmission and their possible roles in information processing by neural microcircuits in both health and disease. As paradigms of neuropsychiatric diseases with strongly implicated risk genes, we discuss the findings in schizophrenia and autism and consider the alterations in synaptic plasticity and network function observed in both human studies and genetic mouse models of these diseases. Together these studies have begun to point toward a likely dominant role of short-term synaptic plasticity alterations in schizophrenia while dysfunction in autism spectrum disorders (ASDs) may be due to a combination of both short-term and long-term synaptic plasticity alterations.
Collapse
Affiliation(s)
- Gregg W. Crabtree
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia UniversityNew York, NY, USA
| | - Joseph A. Gogos
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia UniversityNew York, NY, USA
- Department of Neuroscience, College of Physicians and Surgeons, Columbia UniversityNew York, NY, USA
| |
Collapse
|