1
|
Molla HM, Miguelez Fernández AMM, Tseng KY. Late-adolescent onset of prefrontal endocannabinoid control of hippocampal and amygdalar inputs and its impact on trace-fear conditioning behavior. Neuropsychopharmacology 2024; 49:1417-1424. [PMID: 38467844 PMCID: PMC11250818 DOI: 10.1038/s41386-024-01844-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/28/2024] [Accepted: 03/04/2024] [Indexed: 03/13/2024]
Abstract
Prefrontal cortex (PFC) maturation during adolescence is characterized by structural and functional changes, which involve the remodeling of GABA and glutamatergic synapses, as well as changes in the endocannabinoid system. Yet, the way PFC endocannabinoid signaling interacts with local GABA and glutamatergic function to impact its processing of afferent transmission during the adolescent transition to adulthood remains unknown. Here we combined PFC local field potential recordings with local manipulations of 2-AG and anandamide levels to assess how PFC endocannabinoid signaling is recruited to modulate ventral hippocampal and basolateral amygdalar inputs in vivo in adolescent and adult male rats. We found that the PFC endocannabinoid signaling does not fully emerge until late-adolescence/young adulthood. Once present, both 2-AG and anandamide can be recruited in the PFC to limit the impact of hippocampal drive through a CB1R-mediated mechanism whereas basolateral amygdalar inputs are only inhibited by 2-AG. Similarly, the behavioral effects of increasing 2-AG and anandamide in the PFC do not emerge until late-adolescence/young adulthood. Using a trace fear conditioning paradigm, we found that elevating PFC 2-AG levels preferentially reduced freezing behavior during acquisition without affecting its extinction. In contrast, increasing anandamide levels in the PFC selectively disrupted the extinction of trace fear memory without affecting its acquisition. Collectively, these results indicate a protracted recruitment of PFC endocannabinoid signaling, which becomes online in late adolescence/young adulthood as revealed by its impact on hippocampal and amygdalar-evoked local field potential responses and trace fear memory behavior.
Collapse
Affiliation(s)
- Hanna M Molla
- Department of Anatomy and Cell Biology, University of Illinois Chicago - College of Medicine, Chicago, IL, 60612, USA
| | - Anabel M M Miguelez Fernández
- Department of Anatomy and Cell Biology, University of Illinois Chicago - College of Medicine, Chicago, IL, 60612, USA
| | - Kuei Y Tseng
- Department of Anatomy and Cell Biology, University of Illinois Chicago - College of Medicine, Chicago, IL, 60612, USA.
| |
Collapse
|
2
|
Cody CR, de la Villarmois EA, Fernandez AM, Lardizabal J, McKnight C, Tseng K, Brenhouse HC. Effects of early life adversity and adolescent basolateral amygdala activity on corticolimbic connectivity and anxiety behaviors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.26.586708. [PMID: 38853948 PMCID: PMC11160567 DOI: 10.1101/2024.03.26.586708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Early postnatal development of corticolimbic circuitry is shaped by the environment and is vulnerable to early life challenges. Prior work has shown that early life adversity (ELA) leads to hyperinnervation of glutamatergic basolateral amygdala (BLA) projections to the prefrontal cortex (PFC) in adolescence. While hyperinnervation is associated with later-life anxiety behaviors, the physiological changes underpinning corticolimbic and behavioral impacts of ELA are not understood. We tested whether postsynaptic BLA-driven PFC activity is enhanced in ELA-exposed animals, using the maternal separation (MS) model of ELA. PFC local-field potential following BLA stimulation was facilitated in MS-exposed adolescents. Since ELA increases activity of the early-developing BLA, while the PFC exhibits protracted development, we further examined impacts of glutamatergic BLA activity during early adolescence on later-life PFC innervation and heightened anxiety. In early adolescence, MS-exposed animals exhibited decreased anxiety-like behavior, and acute adolescent BLA inhibition induced behaviors that resembled those of MS animals. To examine long-lasting impacts of adolescent BLA activity on innervation, BLA-originating axonal boutons in the PFC were quantified in late adolescence after early adolescent BLA inhibition. We further tested whether late adolescent BLA-PFC changes were associated with anxious reactivity expressed as heightened acoustic startle responses. MS rearing increased BLA-PFC innervation and threat reactivity in late adolescence, however early adolescent BLA inhibition was insufficient to prevent MS effects, suggesting that earlier BLA activity or post-synaptic receptor rearrangement in the PFC drives altered innervation. Taken together, these findings highlight both pre- and postsynaptic changes in the adolescent BLA-PFC circuit following ELA.
Collapse
Affiliation(s)
- Caitlyn R Cody
- Psychology Department, Northeastern University, Boston MA 02115
| | | | | | | | - Chaney McKnight
- Psychology Department, Northeastern University, Boston MA 02115
| | - Kuei Tseng
- Department of Anatomy and Cell Biology, University of Illinois, Chicago IL 60612
| | | |
Collapse
|
3
|
Grace AA, Uliana DL. Insights into the Mechanism of Action of Antipsychotic Drugs Derived from Animal Models: Standard of Care versus Novel Targets. Int J Mol Sci 2023; 24:12374. [PMID: 37569748 PMCID: PMC10418544 DOI: 10.3390/ijms241512374] [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: 06/28/2023] [Revised: 08/01/2023] [Accepted: 08/01/2023] [Indexed: 08/13/2023] Open
Abstract
Therapeutic intervention for schizophrenia relies on blockade of dopamine D2 receptors in the associative striatum; however, there is little evidence for baseline overdrive of the dopamine system. Instead, the dopamine system is in a hyper-responsive state due to excessive drive by the hippocampus. This causes more dopamine neurons to be in a spontaneously active, hyper-responsive state. Antipsychotic drugs alleviate this by causing depolarization block, or excessive depolarization-induced dopamine neuron inactivation. Indeed, both first- and second-generation antipsychotic drugs cause depolarization block in the ventral tegmentum to relieve positive symptoms, whereas first-generation drugs also cause depolarization in the nigrostriatal dopamine system to lead to extrapyramidal side effects. However, by blocking dopamine receptors, these drugs are activating multiple synapses downstream from the proposed site of pathology: the loss of inhibitory influence over the hippocampus. An overactive hippocampus not only drives the dopamine-dependent positive symptoms, but via its projections to the amygdala and the neocortex can also drive negative and cognitive symptoms, respectively. On this basis, a novel class of drugs that can reverse schizophrenia at the site of pathology, i.e., the hippocampal overdrive, could be effective in alleviating all three classes of symptoms of schizophrenia while also being better tolerated.
Collapse
Affiliation(s)
- Anthony A. Grace
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA 15260, USA;
| | | |
Collapse
|
4
|
Di Domenico D, Mapelli L. Dopaminergic Modulation of Prefrontal Cortex Inhibition. Biomedicines 2023; 11:biomedicines11051276. [PMID: 37238947 DOI: 10.3390/biomedicines11051276] [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: 03/31/2023] [Revised: 04/21/2023] [Accepted: 04/23/2023] [Indexed: 05/28/2023] Open
Abstract
The prefrontal cortex is the highest stage of integration in the mammalian brain. Its functions vary greatly, from working memory to decision-making, and are primarily related to higher cognitive functions. This explains the considerable effort devoted to investigating this area, revealing the complex molecular, cellular, and network organization, and the essential role of various regulatory controls. In particular, the dopaminergic modulation and the impact of local interneurons activity are critical for prefrontal cortex functioning, controlling the excitatory/inhibitory balance and the overall network processing. Though often studied separately, the dopaminergic and GABAergic systems are deeply intertwined in influencing prefrontal network processing. This mini review will focus on the dopaminergic modulation of GABAergic inhibition, which plays a significant role in shaping prefrontal cortex activity.
Collapse
Affiliation(s)
- Danila Di Domenico
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy
| | - Lisa Mapelli
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
5
|
Acoustic startle and prepulse inhibition deficits in adult monkeys with neonatal lesions of the hippocampus, amygdala and orbital frontal cortex. Behav Brain Res 2023; 438:114170. [PMID: 36283567 DOI: 10.1016/j.bbr.2022.114170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 10/17/2022] [Accepted: 10/19/2022] [Indexed: 12/05/2022]
Abstract
Sensory-motor gating, the process of filtering sensory stimuli to modulate motor responses, is impaired in many psychiatric diseases but especially schizophrenia. Sensory-motor gating assessed with the prepulse inhibition paradigm (PPI) measures startle in response to preceding acoustic stimuli. PPI studies in rodents have consistently found that neonatal hippocampal lesions impair sensory-motor gating in adult animals, but its applicability to primates has yet to be tested. The study examined acoustic startle responses and PPI in adult rhesus monkeys with neonatal lesions of the hippocampus (Neo-Hibo), amygdala (Neo-Aibo), and orbital frontal cortex areas 11 and 13 (Neo-Oasp) and with sham-operations (Neo-C). All monkeys were initially habituated to the startle apparatus and assayed for acoustic startle response curves. Subsequently, PPI was measured with the prepulse occurring at 60, 120, 240, 480, 1000 and 5000 msec prior to the pulse onset. No significant group differences in baseline startle were found. Compared to Neo-C monkeys, Neo-Hibo monkeys showed normal startle curves as well as normal PPI at short prepulse delays but prepulse facilitation (PPF) at longer prepulse intervals. Neo-Aibo monkeys displayed enhanced startle responses with only minor changes in PPI, whereas Neo-Oasp monkeys had severe dampening of startle responses and impaired PPI at shorter prepulse intervals. These results support prior evidence from rodent literature of the involvement of each of these areas in the development of the complex cortico-limbic circuit modulating sensory-motor gating and may shade light on the specific neural structures associated with deficits in PPI reported in neuropsychiatric disorders, such as schizophrenia, autism spectrum disorders, and post-traumatic disorders.
Collapse
|
6
|
Bruno JP. Enhancing the resolution of behavioral measures: Key observations during a forty year career in behavioral neuroscience. Neurosci Biobehav Rev 2023; 145:105004. [PMID: 36549379 DOI: 10.1016/j.neubiorev.2022.105004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 12/04/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022]
Abstract
This manuscript reviews several key observations from the research program of Professor John P. Bruno that are believed to have significantly advanced our understanding of the brain's mediation of behavior. This review focuses on findings within several important research areas in behavioral neuroscience, including a) age-dependent neurobehavioral plasticity following brain damage; b) the role of the cortical cholinergic system in attentional processing and cognitive flexibility; and c) the design and validation of animal models of cognitive deficits in schizophrenia. In selecting these observations, emphasis was given to examples in which the heuristic potency was increased by maximizing the resolution and microanalysis of behavioral assays in the same fashion as one typically refines neuronal manipulations. Professor Bruno served the International Behavioral Neuroscience Society (IBNS) as an IBNS Fellow (1995-present) and President of the IBNS (2001-02).
Collapse
Affiliation(s)
- John P Bruno
- Department of Psychology, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
7
|
Nath M, Bhardwaj SK, Srivastava LK, Wong TP. Altered excitatory and decreased inhibitory transmission in the prefrontal cortex of male mice with early developmental disruption to the ventral hippocampus. Cereb Cortex 2023; 33:865-880. [PMID: 35297476 PMCID: PMC9890473 DOI: 10.1093/cercor/bhac107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/20/2022] [Accepted: 02/22/2022] [Indexed: 02/04/2023] Open
Abstract
Ventral hippocampal (vHPC)-prefrontal cortical (PFC) pathway dysfunction is a core neuroimaging feature of schizophrenia. However, mechanisms underlying impaired connectivity within this pathway remain poorly understood. The vHPC has direct projections to the PFC that help shape its maturation. Here, we wanted to investigate the effects of early developmental vHPC perturbations on long-term functional PFC organization. Using whole-cell recordings to assess PFC cellular activity in transgenic male mouse lines, we show early developmental disconnection of vHPC inputs, by excitotoxic lesion or cell-specific ablations, impairs pyramidal cell firing output and produces a persistent increase in excitatory and decrease in inhibitory synaptic inputs onto pyramidal cells. We show this effect is specific to excitatory vHPC projection cell ablation. We further identify PV-interneurons as a source of deficit in inhibitory transmission. We find PV-interneurons are reduced in density, show a reduced ability to sustain high-frequency firing, and show deficits in excitatory inputs that emerge over time. We additionally show differences in vulnerabilities to early developmental vHPC disconnection, wherein PFC PV-interneurons but not pyramidal cells show deficits in NMDA receptor-mediated current. Our results highlight mechanisms by which the PFC adapts to early developmental vHPC perturbations, providing insights into schizophrenia circuit pathology.
Collapse
Affiliation(s)
- Moushumi Nath
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 0G4, Canada.,Basic Neuroscience Division, Douglas Hospital Research Centre, Montreal, QC H4H 1R3, Canada
| | - Sanjeev K Bhardwaj
- Basic Neuroscience Division, Douglas Hospital Research Centre, Montreal, QC H4H 1R3, Canada
| | - Lalit K Srivastava
- Basic Neuroscience Division, Douglas Hospital Research Centre, Montreal, QC H4H 1R3, Canada.,Department of Psychiatry, McGill University, Montreal, QC H3A 1A1, Canada
| | - Tak Pan Wong
- Basic Neuroscience Division, Douglas Hospital Research Centre, Montreal, QC H4H 1R3, Canada.,Department of Psychiatry, McGill University, Montreal, QC H3A 1A1, Canada
| |
Collapse
|
8
|
Kaki S, DeRosa H, Timmerman B, Brummelte S, Hunter RG, Kentner AC. Developmental Manipulation-Induced Changes in Cognitive Functioning. Curr Top Behav Neurosci 2023; 63:241-289. [PMID: 36029460 PMCID: PMC9971379 DOI: 10.1007/7854_2022_389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
Schizophrenia is a complex neurodevelopmental disorder with as-yet no identified cause. The use of animals has been critical to teasing apart the potential individual and intersecting roles of genetic and environmental risk factors in the development of schizophrenia. One way to recreate in animals the cognitive impairments seen in people with schizophrenia is to disrupt the prenatal or neonatal environment of laboratory rodent offspring. This approach can result in congruent perturbations in brain physiology, learning, memory, attention, and sensorimotor domains. Experimental designs utilizing such animal models have led to a greatly improved understanding of the biological mechanisms that could underlie the etiology and symptomology of schizophrenia, although there is still more to be discovered. The implementation of the Research and Domain Criterion (RDoC) has been critical in taking a more comprehensive approach to determining neural mechanisms underlying abnormal behavior in people with schizophrenia through its transdiagnostic approach toward targeting mechanisms rather than focusing on symptoms. Here, we describe several neurodevelopmental animal models of schizophrenia using an RDoC perspective approach. The implementation of animal models, combined with an RDoC framework, will bolster schizophrenia research leading to more targeted and likely effective therapeutic interventions resulting in better patient outcomes.
Collapse
Affiliation(s)
- Sahith Kaki
- School of Arts and Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, USA
| | - Holly DeRosa
- School of Arts and Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, USA
- University of Massachusetts Boston, Boston, MA, USA
| | - Brian Timmerman
- Department of Psychology, Wayne State University, Detroit, MI, USA
| | - Susanne Brummelte
- Department of Psychology, Wayne State University, Detroit, MI, USA
- Translational Neuroscience Program, Wayne State University, Detroit, MI, USA
| | | | - Amanda C Kentner
- School of Arts and Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, USA.
| |
Collapse
|
9
|
Micale V, Di Bartolomeo M, Di Martino S, Stark T, Dell'Osso B, Drago F, D'Addario C. Are the epigenetic changes predictive of therapeutic efficacy for psychiatric disorders? A translational approach towards novel drug targets. Pharmacol Ther 2023; 241:108279. [PMID: 36103902 DOI: 10.1016/j.pharmthera.2022.108279] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 09/01/2022] [Accepted: 09/01/2022] [Indexed: 02/06/2023]
Abstract
The etiopathogenesis of mental disorders is not fully understood and accumulating evidence support that clinical symptomatology cannot be assigned to a single gene mutation, but it involves several genetic factors. More specifically, a tight association between genes and environmental risk factors, which could be mediated by epigenetic mechanisms, may play a role in the development of mental disorders. Several data suggest that epigenetic modifications such as DNA methylation, post-translational histone modification and interference of microRNA (miRNA) or long non-coding RNA (lncRNA) may modify the severity of the disease and the outcome of the therapy. Indeed, the study of these mechanisms may help to identify patients particularly vulnerable to mental disorders and may have potential utility as biomarkers to facilitate diagnosis and treatment of psychiatric disorders. This article summarizes the most relevant preclinical and human data showing how epigenetic modifications can be central to the therapeutic efficacy of antidepressant and/or antipsychotic agents, as possible predictor of drugs response.
Collapse
Affiliation(s)
- Vincenzo Micale
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy.
| | - Martina Di Bartolomeo
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Serena Di Martino
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy
| | - Tibor Stark
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czech Republic; Scientific Core Unit Neuroimaging, Max Planck Institute of Psychiatry, Munich, Germany
| | - Bernardo Dell'Osso
- Department of Biomedical and Clinical Sciences 'Luigi Sacco', University of Milan, Milan, Italy, Department of Mental Health, ASST Fatebenefratelli-Sacco, Milan, Italy; "Aldo Ravelli" Research Center for Neurotechnology and Experimental Brain Therapeutics, Department of Health Sciences, University of Milan Medical School, Milan, Italy; Department of Psychiatry and Behavioral Sciences, Stanford University, CA, USA
| | - Filippo Drago
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy.
| | - Claudio D'Addario
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy; Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
10
|
Fry BR, Roberts D, Thakkar KN, Johnson AW. Variables influencing conditioning-evoked hallucinations: overview and future applications. Psychol Med 2022; 52:2937-2949. [PMID: 36138518 PMCID: PMC9693682 DOI: 10.1017/s0033291722002100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 06/13/2022] [Accepted: 06/20/2022] [Indexed: 01/05/2023]
Abstract
Hallucinations occur in the absence of sensory stimulation and result in vivid perceptual experiences of nonexistent events that manifest across a range of sensory modalities. Approaches from the field of experimental and cognitive psychology have leveraged the idea that associative learning experiences can evoke conditioning-induced hallucinations in both animals and humans. In this review, we describe classical and contemporary findings and highlight the variables eliciting these experiences. We also provide an overview of the neurobiological mechanisms, along with the associative and computational factors that may explain hallucinations that are generated by representation-mediated conditioning phenomena. Through the integration of animal and human research, significant advances into the psychobiology of hallucinations are possible, which may ultimately translate to more effective clinical applications.
Collapse
Affiliation(s)
- Benjamin R. Fry
- Department of Psychology, Michigan State University, East Lansing, MI, USA
| | - Dominic Roberts
- Department of Psychology, Michigan State University, East Lansing, MI, USA
| | - Katharine N. Thakkar
- Department of Psychology, Michigan State University, East Lansing, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Alexander W. Johnson
- Department of Psychology, Michigan State University, East Lansing, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
11
|
Apam-Castillejos DJ, Tendilla-Beltrán H, Vázquez-Roque RA, Vázquez-Hernández AJ, Fuentes-Medel E, García-Dolores F, Díaz A, Flores G. Second-generation antipsychotic olanzapine attenuates behavioral and prefrontal cortex synaptic plasticity deficits in a neurodevelopmental schizophrenia-related rat model. J Chem Neuroanat 2022; 125:102166. [PMID: 36156295 DOI: 10.1016/j.jchemneu.2022.102166] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 09/17/2022] [Accepted: 09/20/2022] [Indexed: 11/19/2022]
Abstract
Second-generation antipsychotics are the drugs of choice for the treatment of neurodevelopmental-related mental diseases such as schizophrenia. Despite the effectiveness of these drugs to ameliorate some of the symptoms of schizophrenia, specifically the positive ones, the mechanisms beyond their antipsychotic effect are still poorly understood. Specifically, second-generation antipsychotics are reported to have anti-inflammatory, antioxidant and neuroplastic properties. Using the neonatal ventral hippocampus lesion (nVHL) in the rat, an accepted schizophrenia-related model, we evaluated the effect of the second-generation antipsychotic olanzapine (OLZ) in the behavioral, neuroplastic, and neuroinflammatory alterations exhibited in the nVHL animals. OLZ corrected the hyperlocomotion and impaired working memory of the nVHL animals but failed to enhance social disturbances of these animals. In the prefrontal cortex (PFC), OLZ restored the pyramidal cell structural plasticity in the nVHL rats, enhancing the dendritic arbor length, the spinogenesis and the proportion of mature spines. Moreover, OLZ attenuated astrogliosis as well as some pro-inflammatory, oxidative stress, and apoptosis-related molecules in the PFC. These findings reinforce the evidence of anti-inflammatory, antioxidant, and neurotrophic mechanisms of second-generation antipsychotics in the nVHL schizophrenia-related model, which allows for the possibility of developing more specific drugs for this disorder and thus avoiding the side effects of current schizophrenia treatments.
Collapse
Affiliation(s)
| | | | | | | | - Estefania Fuentes-Medel
- Facultad de Ciencias Químicas (FCQ), Benemérita Universidad Autónoma de Puebla (BUAP), Mexico
| | - Fernando García-Dolores
- Instituto de Ciencias Forenses del Tribunal Superior de Justicia de la Ciudad de México (TSJCDMX), Mexico
| | - Alfonso Díaz
- Facultad de Ciencias Químicas (FCQ), Benemérita Universidad Autónoma de Puebla (BUAP), Mexico
| | - Gonzalo Flores
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla (BUAP), Mexico.
| |
Collapse
|
12
|
Uliana DL, Zhu X, Gomes FV, Grace AA. Using animal models for the studies of schizophrenia and depression: The value of translational models for treatment and prevention. Front Behav Neurosci 2022; 16:935320. [PMID: 36090659 PMCID: PMC9449416 DOI: 10.3389/fnbeh.2022.935320] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 08/04/2022] [Indexed: 11/29/2022] Open
Abstract
Animal models of psychiatric disorders have been highly effective in advancing the field, identifying circuits related to pathophysiology, and identifying novel therapeutic targets. In this review, we show how animal models, particularly those based on development, have provided essential information regarding circuits involved in disorders, disease progression, and novel targets for intervention and potentially prevention. Nonetheless, in recent years there has been a pushback, largely driven by the US National Institute of Mental Health (NIMH), to shift away from animal models and instead focus on circuits in normal subjects. This has been driven primarily from a lack of discovery of new effective therapeutic targets, and the failure of targets based on preclinical research to show efficacy. We discuss why animal models of complex disorders, when strongly cross-validated by clinical research, are essential to understand disease etiology as well as pathophysiology, and direct new drug discovery. Issues related to shortcomings in clinical trial design that confound translation from animal models as well as the failure to take patient pharmacological history into account are proposed to be a source of the failure of what are likely effective compounds from showing promise in clinical trials.
Collapse
Affiliation(s)
- Daniela L. Uliana
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Xiyu Zhu
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Felipe V. Gomes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Anthony A. Grace
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
13
|
McCunn P, Chen X, Gimi B, Green AI, Khokhar JY. Glutamine and GABA alterations in cingulate cortex may underlie alcohol drinking in a rat model of co-occurring alcohol use disorder and schizophrenia: an 1H-MRS study. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2022; 8:67. [PMID: 35999232 PMCID: PMC9399110 DOI: 10.1038/s41537-022-00272-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 08/01/2022] [Indexed: 06/15/2023]
Abstract
Alcohol use disorder commonly occurs in patients with schizophrenia and significantly worsens the clinical course of the disorder. The neurobiological underpinnings of alcohol drinking are not well understood. Magnetic resonance spectroscopy (MRS) has been used to assess the neurochemical substrates that may be associated with alcohol drinking in patients; however, the causal impact of these findings remains elusive, highlighting the need for studies in animal models. This study performed MRS in the neonatal ventral hippocampal lesioned (NVHL) rat model, a model of co-occurring schizophrenia and substance use disorders. NVHL lesions (or sham surgeries) were performed on post-natal day 7 and animals were given brief exposure to alcohol during adolescence (10% v/v in a 2-bottle choice design). Animals were re-exposed to alcohol during adulthood (20% v/v) until a stable drinking baseline was established, and then forced into abstinence to control for the effects of differential alcohol drinking. Animals were scanned for MRS after one month of abstinence. NVHL rats consumed significantly more alcohol than sham rats and in the cingulate cortex showed significantly higher levels of GABA and glutamine. Significantly lower GABA levels were observed in the nucleus accumbens. No differences between the NVHL and sham animals were observed in the hippocampus. Correlation analysis revealed that GABA and glutamine concentrations in the cingulate cortex significantly correlated with the rats' alcohol drinking prior to 30 days of forced abstinence. These findings suggest that a potential dysfunction in the glutamate/GABA-glutamine cycle may contribute to alcohol drinking in a rat model of schizophrenia, and this dysfunction could be targeted in future treatment-focused studies.
Collapse
Affiliation(s)
- Patrick McCunn
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Xi Chen
- Department of Radiology, Biomedical NMR Research Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
- Psychotic Disorders Division, McLean Hospital, Belmont, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Barjor Gimi
- Department of Radiology, Biomedical NMR Research Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Alan I Green
- Department of Psychiatry, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Jibran Y Khokhar
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada.
- Department of Psychiatry, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA.
| |
Collapse
|
14
|
Hazani R, Lavidor M, Weller A. Treatments for Social Interaction Impairment in Animal Models of Schizophrenia: A Critical Review and Meta-analysis. Schizophr Bull 2022; 48:1179-1193. [PMID: 35925025 PMCID: PMC9673263 DOI: 10.1093/schbul/sbac093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND While pharmacological treatments for positive symptoms of schizophrenia are widely used, their beneficial effect on negative symptoms, particularly social impairment, is insufficiently studied. Therefore, there is an increasing interest in preclinical research of potentially beneficial treatments, with mixed results. The current review aims to evaluate the efficacy of available treatments for social deficits in different animal models of schizophrenia. STUDY DESIGN A systematic literature search generated 145 outcomes for the measures "total time" and "number" of social interactions. Standardized mean differences (SMD) and 95% confidence interval (CI) were calculated, and heterogeneity was tested using Q statistics in a random-effect meta-analytic model. Given the vast heterogeneity in effect sizes, the animal model, treatment group, and sample size were all examined as potential moderators. STUDY RESULTS The results showed that in almost all models, treatment significantly improved social deficit (total time: SMD = 1.24; number: SMD = 1.1). The moderator analyses discovered significant subgroup differences across models and treatment subgroups. Perinatal and adult pharmacological models showed the most substantial influence of treatments on social deficits, reflecting relative pharmacological validity. Furthermore, atypical antipsychotic drugs had the highest SMD within each model subgroup. CONCLUSIONS Our findings indicate that the improvement in social interaction behaviors is dependent on the animal model and treatment family used. Implications for the preclinical and clinical fields are discussed.
Collapse
Affiliation(s)
- Reut Hazani
- To whom correspondence should be addressed; Department of Psychology, Bar-Ilan University, Ramat-Gan 5290002, Israel; tel: 972-3-531-8548, fax: 972-3-738-4173, e-mail:
| | - Michal Lavidor
- Psychology Department and Gonda Brain Research Center, Bar-Ilan University, Ramat Gan, Israel
| | - Aron Weller
- Psychology Department and Gonda Brain Research Center, Bar-Ilan University, Ramat Gan, Israel
| |
Collapse
|
15
|
Oliveras I, Soria-Ruiz OJ, Sampedro-Viana D, Cañete T, Río-Álamos C, Tobeña A, Fernández-Teruel A. Different maturation patterns for sensorimotor gating and startle habituation deficits in male and female RHA vs RLA rats. Behav Brain Res 2022; 434:114021. [PMID: 35872331 DOI: 10.1016/j.bbr.2022.114021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 05/17/2022] [Accepted: 07/19/2022] [Indexed: 11/28/2022]
Abstract
Neurodevelopmental anomalies are thought to play a crucial role in the emergence of schizophrenia. The Roman high-avoidance (RHA) rats exhibit impaired prepulse inhibition (PPI), as well as other behavioral and cognitive singularities related to schizophrenia syndromes compared to the Roman low-avoidance (RLA) rats. In the present study, we aimed at elucidating whether PPI deficits in the RHA rats take place during prepubescence, adolescence, or adulthood. Thus, we evaluated the levels of PPI of both strains and both sexes during these three developmental phases. Additionally, we also investigated the onset of startle habituation deficits in the same groups. The results showed that male RHA rats exhibit a clear-cut PPI reduction compared to their RLA counterparts in adulthood. In female RHA rats, we observed lower levels of PPI since adolescence and through adulthood. We also found no differences between PPI percentages among the three ages in RHA male rats. Contrarily, in male RLA rats, PPI levels were increased in adults compared to their adolescent and prepubescent counterparts. Finally, a deficit in startle habituation was observed in adulthood of both male and female RHA rats, although in the latter case the disturbance in startle habituation was more profound. These results further the description of the maturational trajectory of cognitive markers relevant to schizophrenia prodrome and they add face validity to the RHA rats as a model of schizophrenia-relevant phenotypes.
Collapse
Affiliation(s)
- Ignasi Oliveras
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine & Institute of Neurosciences, School of Medicine, Autonomous University of Barcelona, Bellaterra, Barcelona 08193, Spain.
| | - Oscar J Soria-Ruiz
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine & Institute of Neurosciences, School of Medicine, Autonomous University of Barcelona, Bellaterra, Barcelona 08193, Spain
| | - Daniel Sampedro-Viana
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine & Institute of Neurosciences, School of Medicine, Autonomous University of Barcelona, Bellaterra, Barcelona 08193, Spain
| | - Toni Cañete
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine & Institute of Neurosciences, School of Medicine, Autonomous University of Barcelona, Bellaterra, Barcelona 08193, Spain
| | | | - Adolf Tobeña
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine & Institute of Neurosciences, School of Medicine, Autonomous University of Barcelona, Bellaterra, Barcelona 08193, Spain
| | - Alberto Fernández-Teruel
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine & Institute of Neurosciences, School of Medicine, Autonomous University of Barcelona, Bellaterra, Barcelona 08193, Spain.
| |
Collapse
|
16
|
Sánchez‐Olguin CP, Zamudio SR, Guzmán‐Velázquez S, Márquez‐Portillo M, Caba‐Flores MD, Camacho‐Abrego I, Flores G, Melo AI. Neonatal ventral hippocampus lesion disrupts maternal behavior in rats: An animal model of schizophrenia. Dev Psychobiol 2022; 64:e22283. [DOI: 10.1002/dev.22283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/08/2022] [Accepted: 04/17/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Claudia P. Sánchez‐Olguin
- Departamento de Fisiología Escuela Nacional de Ciencias Biológicas Instituto Politécnico Nacional Mexico City Mexico
- Maestría en Ciencias Biológicas Universidad Autónoma de Tlaxcala Tlaxcala Mexico
| | - Sergio R. Zamudio
- Departamento de Fisiología Escuela Nacional de Ciencias Biológicas Instituto Politécnico Nacional Mexico City Mexico
| | - Sonia Guzmán‐Velázquez
- Departamento de Fisiología Escuela Nacional de Ciencias Biológicas Instituto Politécnico Nacional Mexico City Mexico
| | - Mariana Márquez‐Portillo
- Centro de Investigación en Reproducción Animal CINVESTAV Laboratorio Tlaxcala Universidad Autónoma de Tlaxcala Tlaxcala Mexico
| | | | - Israel Camacho‐Abrego
- Laboratorio de Neuropsiquiatría Instituto de Fisiología Benemérita Universidad Autónoma de Puebla Puebla Mexico
- Doctorado en Ciencias Biológicas Universidad Autónoma de Tlaxcala Tlaxcala Mexico
| | - Gonzalo Flores
- Laboratorio de Neuropsiquiatría Instituto de Fisiología Benemérita Universidad Autónoma de Puebla Puebla Mexico
| | - Angel I. Melo
- Centro de Investigación en Reproducción Animal CINVESTAV Laboratorio Tlaxcala Universidad Autónoma de Tlaxcala Tlaxcala Mexico
| |
Collapse
|
17
|
Jung SY, Kim YE, Park WS, Ahn SY, Sung DK, Sung SI, Joo KM, Kim SG, Chang YS. Thrombin Preconditioning Improves the Therapeutic Efficacy of Mesenchymal Stem Cells in Severe Intraventricular Hemorrhage Induced Neonatal Rats. Int J Mol Sci 2022; 23:ijms23084447. [PMID: 35457266 PMCID: PMC9030410 DOI: 10.3390/ijms23084447] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/11/2022] [Accepted: 04/14/2022] [Indexed: 02/05/2023] Open
Abstract
Severe intraventricular hemorrhage (IVH) remains a major cause of high mortality and morbidity in extremely preterm infants. Mesenchymal stem cell (MSC) transplantation is a possible therapeutic option, and development of therapeutics with enhanced efficacy is necessary. This study investigated whether thrombin preconditioning improves the therapeutic efficacy of human Wharton’s jelly-derived MSC transplantation for severe neonatal IVH, using a rat model. Severe neonatal IVH was induced by injecting 150 μL blood into each lateral ventricle on postnatal day (P) 4 in Sprague-Dawley rats. After 2 days (P6), naïve MSCs or thrombin-preconditioned MSCs (1 × 105/10 μL) were transplanted intraventricularly. After behavioral tests, brain tissues and cerebrospinal fluid of P35 rats were obtained for histological and biochemical analyses, respectively. Thrombin-preconditioned MSC transplantation significantly reduced IVH-induced ventricular dilatation on in vivo magnetic resonance imaging, which was coincident with attenuations of reactive gliosis, cell death, and the number of activated microglia and levels of inflammatory cytokines after IVH induction, compared to naïve MSC transplantation. In the behavioral tests, the sensorimotor and memory functions significantly improved after transplantation of thrombin-preconditioned MSCs, compared to naïve MSCs. Overall, thrombin preconditioning significantly improves the therapeutic potential and more effectively attenuates brain injury, including progressive ventricular dilatation, gliosis, cell death, inflammation, and neurobehavioral functional impairment, in newborn rats with induced severe IVH than does naïve MSC transplantation.
Collapse
Affiliation(s)
- So Yeon Jung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (S.Y.J.); (W.S.P.); (S.Y.A.); (D.K.S.); (S.I.S.)
- Department of Anatomy & Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, Korea;
| | - Young Eun Kim
- Samsung Medical Center, Cell and Gene Therapy Institute, Seoul 06351, Korea;
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Korea
| | - Won Soon Park
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (S.Y.J.); (W.S.P.); (S.Y.A.); (D.K.S.); (S.I.S.)
- Samsung Medical Center, Cell and Gene Therapy Institute, Seoul 06351, Korea;
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Korea
| | - So Yoon Ahn
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (S.Y.J.); (W.S.P.); (S.Y.A.); (D.K.S.); (S.I.S.)
| | - Dong Kyung Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (S.Y.J.); (W.S.P.); (S.Y.A.); (D.K.S.); (S.I.S.)
| | - Se In Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (S.Y.J.); (W.S.P.); (S.Y.A.); (D.K.S.); (S.I.S.)
| | - Kyeung Min Joo
- Department of Anatomy & Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, Korea;
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Korea
| | - Seong Gi Kim
- Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science (IBS), Suwon 16419, Korea;
- Department of Biomedical Engineering, Sungkyunkwan University, Suwon 16419, Korea
| | - Yun Sil Chang
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (S.Y.J.); (W.S.P.); (S.Y.A.); (D.K.S.); (S.I.S.)
- Samsung Medical Center, Cell and Gene Therapy Institute, Seoul 06351, Korea;
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Korea
- Correspondence: ; Tel.: +82-2-3410-3528; Fax: +82-2-3410-0049
| |
Collapse
|
18
|
Dion-Albert L, Bandeira Binder L, Daigle B, Hong-Minh A, Lebel M, Menard C. Sex differences in the blood-brain barrier: Implications for mental health. Front Neuroendocrinol 2022; 65:100989. [PMID: 35271863 DOI: 10.1016/j.yfrne.2022.100989] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 02/07/2022] [Accepted: 02/19/2022] [Indexed: 12/13/2022]
Abstract
Prevalence of mental disorders, including major depressive disorder (MDD), bipolar disorder (BD) and schizophrenia (SZ) are increasing at alarming rates in our societies. Growing evidence points toward major sex differences in these conditions, and high rates of treatment resistance support the need to consider novel biological mechanisms outside of neuronal function to gain mechanistic insights that could lead to innovative therapies. Blood-brain barrier alterations have been reported in MDD, BD and SZ. Here, we provide an overview of sex-specific immune, endocrine, vascular and transcriptional-mediated changes that could affect neurovascular integrity and possibly contribute to the pathogenesis of mental disorders. We also identify pitfalls in current literature and highlight promising vascular biomarkers. Better understanding of how these adaptations can contribute to mental health status is essential not only in the context of MDD, BD and SZ but also cardiovascular diseases and stroke which are associated with higher prevalence of these conditions.
Collapse
Affiliation(s)
- Laurence Dion-Albert
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Canada
| | - Luisa Bandeira Binder
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Canada
| | - Beatrice Daigle
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Canada
| | - Amandine Hong-Minh
- Smurfit Institute of Genetics, Trinity College Dublin, Lincoln Place Gate, Dublin 2, Ireland
| | - Manon Lebel
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Canada
| | - Caroline Menard
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Canada.
| |
Collapse
|
19
|
Borjeni MS, Korani M, Meftahi GH, Davoodian N, Hadipour M, Jahromi GP. Laterality dissociation of ventral hippocampus inhibition in learning and memory, glial activation and neural arborization in response to chronic stress in male Wistar rats. J Chem Neuroanat 2022; 121:102090. [DOI: 10.1016/j.jchemneu.2022.102090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 03/05/2022] [Accepted: 03/10/2022] [Indexed: 11/26/2022]
|
20
|
Rogóż Z, Kamińska K, Lech MA, Lorenc-Koci E. N-Acetylcysteine and Aripiprazole Improve Social Behavior and Cognition and Modulate Brain BDNF Levels in a Rat Model of Schizophrenia. Int J Mol Sci 2022; 23:ijms23042125. [PMID: 35216241 PMCID: PMC8877560 DOI: 10.3390/ijms23042125] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/18/2022] [Accepted: 02/11/2022] [Indexed: 02/01/2023] Open
Abstract
Treatment of negative symptoms and cognitive disorders in patients with schizophrenia is still a serious clinical problem. The aim of our study was to compare the efficacy of chronic administration of the atypical antipsychotic drug aripiprazole (7-{4-[4-(2,3-dichlorophenyl)-1-piperazinyl] butoxy}-3,4-dihydro-2(1H)-quinolinone; ARI) and the well-known antioxidant N-acetylcysteine (NAC) both in alleviating schizophrenia-like social and cognitive deficits and in reducing the decreases in the levels of the brain-derived neurotrophic factor (BDNF) in the prefrontal cortex (PFC) and hippocampus (HIP) of adult Sprague-Dawley rats, that have been induced by chronic administration of the model compound L-buthionine-(S, R)-sulfoximine (BSO) during the early postnatal development (p5–p16). ARI was administered at doses of 0.1 and 0.3 mg/kg while NAC at doses of 10 and 30 mg/kg, alone or in combination. Administration of higher doses of ARI or NAC alone, or co-treatment with lower, ineffective doses of these drugs significantly improved social and cognitive performance as assessed in behavioral tests. Both doses of NAC and 0.3 mg/kg of ARI increased the expression of BDNF mRNA in the PFC, while all doses of these drugs and their combinations enhanced the levels of BDNF protein in this brain structure. In the HIP, only 0,3 mg/kg ARI increased the levels of both BDNF mRNA and its protein. These data show that in the rat BSO-induced neurodevelopmental model of schizophrenia, ARI and NAC differently modulated BDNF levels in the PFC and HIP.
Collapse
Affiliation(s)
- Zofia Rogóż
- Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, 31-343 Kraków, Poland; (Z.R.); (K.K.); (M.A.L.)
| | - Kinga Kamińska
- Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, 31-343 Kraków, Poland; (Z.R.); (K.K.); (M.A.L.)
| | - Marta Anna Lech
- Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, 31-343 Kraków, Poland; (Z.R.); (K.K.); (M.A.L.)
| | - Elżbieta Lorenc-Koci
- Department of Neuro-Psychopharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, 31-343 Kraków, Poland
- Correspondence: ; Tel.: +48-126-623-272
| |
Collapse
|
21
|
Yang S, Tseng KY. Maturation of Corticolimbic Functional Connectivity During Sensitive Periods of Brain Development. Curr Top Behav Neurosci 2022; 53:37-53. [PMID: 34386969 DOI: 10.1007/7854_2021_239] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The maturation of key corticolimbic structures and the prefrontal cortex during sensitive periods of brain development from early life through adolescence is crucial for the acquisition of a variety of cognitive and affective processes associated with adult behavior. In this chapter, we first review how key cellular and circuit level changes during adolescence dictate the development of the prefrontal cortex and its capacity to integrate contextual and emotional information from the ventral hippocampus and the amygdala. We further discuss how afferent transmission from ventral hippocampal and amygdala inputs displays unique age-dependent trajectories that directly impact prefrontal functional maturation through adolescence. We conclude by proposing that time-sensitive strengthening of specific corticolimbic synapses is a critical contributing factor for the protracted maturation of cognitive and emotional regulation by the prefrontal cortex.
Collapse
Affiliation(s)
- Shaolin Yang
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kuei Y Tseng
- Department of Anatomy and Cell Biology, University of Illinois at Chicago - College of Medicine, Chicago, IL, USA.
| |
Collapse
|
22
|
Jenkins BW, Buckhalter S, Perreault ML, Khokhar JY. Cannabis Vapor Exposure Alters Neural Circuit Oscillatory Activity in a Neurodevelopmental Model of Schizophrenia: Exploring the Differential Impact of Cannabis Constituents. SCHIZOPHRENIA BULLETIN OPEN 2022; 3:sgab052. [PMID: 35036917 PMCID: PMC8752653 DOI: 10.1093/schizbullopen/sgab052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Cannabis use is highly prevalent in patients with schizophrenia and worsens the course of the disorder. To understand how exposure to cannabis changes schizophrenia-related oscillatory disruptions, we investigated the impact of administering cannabis vapor containing either Δ9-tetrahydrocannabinol (THC) or balanced THC/cannabidiol (CBD) on oscillatory activity in the neonatal ventral hippocampal lesion (NVHL) rat model of schizophrenia. Male Sprague Dawley rats underwent lesion or sham surgeries on postnatal day 7. In adulthood, electrodes were implanted targeting the cingulate cortex (Cg), the prelimbic cortex (PrLC), the hippocampus (HIP), and the nucleus accumbens (NAc). Local field potential recordings were obtained after rats were administered either the "THC-only" cannabis vapor (8-18% THC/0% CBD) or the "Balanced THC:CBD" cannabis vapor (4-11% THC/8.5-15.5% CBD) in a cross-over design with a 2-week wash-out period between exposures. Compared to controls, NVHL rats had reduced baseline gamma power in the Cg, HIP, and NAc, and reduced HIP-Cg high-gamma coherence. THC-only vapor exposure broadly suppressed oscillatory power and coherence, even beyond the baseline reductions observed in NHVL rats. Balanced THC:CBD vapor, however, did not suppress oscillatory power and coherence, and in some instances enhanced power. For NVHL rats, THC-only vapor normalized the baseline HIP-Cg high-gamma coherence deficits. NHVL rats demonstrated a 20 ms delay in HIP theta to high-gamma phase coupling, which was not apparent in the PrLC and NAc after both exposures. In conclusion, cannabis vapor exposure has varying impacts on oscillatory activity in NVHL rats, and the relative composition of naturally occurring cannabinoids may contribute to this variability.
Collapse
Affiliation(s)
- Bryan W Jenkins
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, Canada
| | - Shoshana Buckhalter
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | | | - Jibran Y Khokhar
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
23
|
Pilarzyk K, Farmer R, Porcher L, Kelly MP. The Role of PDE11A4 in Social Isolation-Induced Changes in Intracellular Signaling and Neuroinflammation. Front Pharmacol 2021; 12:749628. [PMID: 34887755 PMCID: PMC8650591 DOI: 10.3389/fphar.2021.749628] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 10/25/2021] [Indexed: 12/29/2022] Open
Abstract
Phosphodiesterase 11A (PDE11A), an enzyme that degrades cyclic nucleotides (cAMP and cGMP), is the only PDE whose mRNA expression in brain is restricted to the hippocampal formation. Previously, we showed that chronic social isolation changes subsequent social behaviors in adult mice by reducing expression of PDE11A4 in the membrane fraction of the ventral hippocampus (VHIPP). Here we seek extend these findings by determining 1) if isolation-induced decreases in PDE11A4 require chronic social isolation or if they occur acutely and are sustained long-term, 2) if isolation-induced decreases occur uniquely in adults (i.e., not adolescents), and 3) how the loss of PDE11 signaling may increase neuroinflammation. Both acute and chronic social isolation decrease PDE11A4 expression in adult but not adolescent mice. This decrease in PDE11A4 is specific to the membrane compartment of the VHIPP, as it occurs neither in the soluble nor nuclear fractions of the VHIPP nor in any compartment of the dorsal HIPP. The effect of social isolation on membrane PDE11A4 is also selective in that PDE2A and PDE10A expression remain unchanged. Isolation-induced decreases in PDE11A4 expression appear to be functional as social isolation elicited changes in PDE11A-relevant signal transduction cascades (i.e., decreased pCamKIIα and pS6-235/236) and behavior (i.e., increased remote long-term memory for social odor recognition). Interestingly, we found that isolation-induced decreases in membrane PDE11A4 correlated with increased expression of interleukin-6 (IL-6) in the soluble fraction, suggesting pro-inflammatory signaling for this cytokine. This effect on IL-6 is consistent with the fact that PDE11A deletion increased microglia activation, although it left astrocytes unchanged. Together, these data suggest that isolation-induced decreases in PDE11A4 may alter subsequent social behavior via increased neuroinflammatory processes in adult mice.
Collapse
Affiliation(s)
- Katy Pilarzyk
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Reagan Farmer
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Latarsha Porcher
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Michy P Kelly
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States.,Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States.,Center for Aging Research, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
24
|
Fernández-Teruel A, Oliveras I, Cañete T, Rio-Álamos C, Tapias-Espinosa C, Sampedro-Viana D, Sánchez-González A, Sanna F, Torrubia R, González-Maeso J, Driscoll P, Morón I, Torres C, Aznar S, Tobeña A, Corda MG, Giorgi O. Neurobehavioral and neurodevelopmental profiles of a heuristic genetic model of differential schizophrenia- and addiction-relevant features: The RHA vs. RLA rats. Neurosci Biobehav Rev 2021; 131:597-617. [PMID: 34571119 DOI: 10.1016/j.neubiorev.2021.09.042] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/20/2021] [Accepted: 09/21/2021] [Indexed: 12/26/2022]
Abstract
The Roman High- (RHA) and Low-(RLA) avoidance rat lines/strains were generated through bidirectional selective breeding for rapid (RHA) vs. extremely poor (RLA) two-way active avoidance acquisition. Compared with RLAs and other rat strains/stocks, RHAs are characterized by increased impulsivity, deficits in social behavior, novelty-induced hyper-locomotion, impaired attentional/cognitive abilities, vulnerability to psychostimulant sensitization and drug addiction. RHA rats also exhibit decreased function of the prefrontal cortex (PFC) and hippocampus, increased functional activity of the mesolimbic dopamine system and a dramatic deficit of central metabotropic glutamate-2 (mGlu2) receptors (due to a stop codon mutation at cysteine 407 in Grm2 -cys407*-), along with increased density of 5-HT2A receptors in the PFC, alterations of several synaptic markers and increased density of pyramidal "thin" (immature) dendrític spines in the PFC. These characteristics suggest an immature brain of RHA rats, and are reminiscent of schizophrenia features like hypofrontality and disruption of the excitation/inhibition cortical balance. RHA rats represent a promising heuristic model of neurodevelopmental schizophrenia-relevant features and comorbidity with drug addiction vulnerability.
Collapse
Affiliation(s)
- Alberto Fernández-Teruel
- Medical Psychology Unit, Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Autonomous University of Barcelona, 08193, Bellaterra, Barcelona, Spain.
| | - Ignasi Oliveras
- Medical Psychology Unit, Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Autonomous University of Barcelona, 08193, Bellaterra, Barcelona, Spain
| | - Toni Cañete
- Medical Psychology Unit, Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Autonomous University of Barcelona, 08193, Bellaterra, Barcelona, Spain
| | | | - Carles Tapias-Espinosa
- Medical Psychology Unit, Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Autonomous University of Barcelona, 08193, Bellaterra, Barcelona, Spain
| | - Daniel Sampedro-Viana
- Medical Psychology Unit, Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Autonomous University of Barcelona, 08193, Bellaterra, Barcelona, Spain
| | - Ana Sánchez-González
- Medical Psychology Unit, Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Autonomous University of Barcelona, 08193, Bellaterra, Barcelona, Spain
| | - Francesco Sanna
- Department of Life and Environmental Sciences (DiSVA), University of Cagliari, Italy
| | - Rafael Torrubia
- Medical Psychology Unit, Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Autonomous University of Barcelona, 08193, Bellaterra, Barcelona, Spain
| | - Javier González-Maeso
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | | | - Ignacio Morón
- Department of Psychobiology and Centre of Investigation of Mind, Brain, and Behaviour (CIMCYC), University of Granada, Spain
| | - Carmen Torres
- Department of Psychology, University of Jaén, 23071, Jaén, Spain.
| | - Susana Aznar
- Research Laboratory for Stereology and Neuroscience, Bispebjerg Copenhagen University Hospital, 2400, Copenhagen, Denmark.
| | - Adolf Tobeña
- Medical Psychology Unit, Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Autonomous University of Barcelona, 08193, Bellaterra, Barcelona, Spain.
| | - Maria G Corda
- Department of Life and Environmental Sciences (DiSVA), University of Cagliari, Italy.
| | - Osvaldo Giorgi
- Department of Life and Environmental Sciences (DiSVA), University of Cagliari, Italy.
| |
Collapse
|
25
|
Tendilla-Beltrán H, Coatl-Cuaya H, Meneses-Prado S, Vázquez-Roque RA, Brambila E, Tapia-Rodríguez M, Martín-Hernández D, Garcés-Ramírez L, Madrigal JLM, Leza JC, Flores G. Neuroplasticity and inflammatory alterations in the nucleus accumbens are corrected after risperidone treatment in a schizophrenia-related developmental model in rats. Schizophr Res 2021; 235:17-28. [PMID: 34298239 DOI: 10.1016/j.schres.2021.07.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 07/04/2021] [Accepted: 07/11/2021] [Indexed: 12/18/2022]
Abstract
Increased dopaminergic activity in the striatum underlies the neurobiology of psychotic symptoms in schizophrenia (SZ). Beyond the impaired connectivity among the limbic system, the excess of dopamine could lead to inflammation and oxidative/nitrosative stress. It has been suggested that atypical antipsychotic drugs attenuate psychosis not only due to their modulatory activity on the dopaminergic/serotonergic neurotransmission but also due to their anti-inflammatory/antioxidant effects. In such a manner, we assessed the effects of the atypical antipsychotic risperidone (RISP) on the structural neuroplasticity and biochemistry of the striatum in adult rats with neonatal ventral hippocampus lesion (NVHL), which is a developmental SZ-related model. RISP administration (0.25 mg/kg, i.p.) ameliorated the neuronal atrophy and the impairments in the morphology of the dendritic spines in the spiny projection neurons (SPNs) of the ventral striatum (nucleus accumbens: NAcc) in the NVHL rats. Also, RISP treatment normalized the pro-inflammatory pathways and induced the antioxidant activity of the nuclear factor (erythroid-derived 2)-like 2 (Nrf2) in this model. Our results point to the neurotrophic, anti-inflammatory, and antioxidant effects of RISP, together with its canonical antipsychotic mechanism, to enhance striatum function in animals with NVHL.
Collapse
Affiliation(s)
- Hiram Tendilla-Beltrán
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla 72570, Mexico; Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional (IPN), CDMX 11340, Mexico; Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid 28040, Spain; Instituto Universitario de Investigación en Neuroquímica (IUIN), UCM, Spain
| | - Heriberto Coatl-Cuaya
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla 72570, Mexico; Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional (IPN), CDMX 11340, Mexico
| | - Silvia Meneses-Prado
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla 72570, Mexico
| | | | | | - Miguel Tapia-Rodríguez
- Instituto de Investigaciones Biomédicas (IIBO), Universidad Nacional Autónoma de México (UNAM), CDMX 04510, Mexico
| | - David Martín-Hernández
- Servicio de Psiquiatría del Niño y del Adolescente, Instituto de Psiquiatría y Salud Mental, Hospital General Universitario Gregorio Marañón, Facultad de Medicina, Universidad Complutense, IiSGM, CIBERSAM, Madrid, Spain
| | - Linda Garcés-Ramírez
- Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional (IPN), CDMX 11340, Mexico
| | - José L M Madrigal
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid 28040, Spain; Instituto Universitario de Investigación en Neuroquímica (IUIN), UCM, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Madrid 28029, Spain
| | - Juan C Leza
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid 28040, Spain; Instituto Universitario de Investigación en Neuroquímica (IUIN), UCM, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Madrid 28029, Spain.
| | - Gonzalo Flores
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla 72570, Mexico.
| |
Collapse
|
26
|
Alteration of NMDA receptor trafficking as a cellular hallmark of psychosis. Transl Psychiatry 2021; 11:444. [PMID: 34462417 PMCID: PMC8405679 DOI: 10.1038/s41398-021-01549-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 07/24/2021] [Accepted: 08/04/2021] [Indexed: 02/07/2023] Open
Abstract
A dysfunction of the glutamatergic transmission, especially of the NMDA receptor (NMDAR), constitutes one of the main biological substrate of psychotic disorders, such as schizophrenia. The NMDAR signaling hypofunction, through genetic and/or environmental insults, would cause a neurodevelopmental myriad of molecular, cellular, and network alterations that persist throughout life. Yet, the mechanisms underpinning NMDAR dysfunctions remain elusive. Here, we compared the membrane trafficking of NMDAR in three gold-standard models of schizophrenia, i.e., patient's cerebrospinal fluids, genetic manipulations of susceptibility genes, and prenatal developmental alterations. Using a combination of single nanoparticle tracking, electrophysiological, biochemical, and behavioral approaches in rodents, we identified that the NMDAR trafficking in hippocampal neurons was consistently altered in all these different models. Artificial manipulations of the NMDAR surface dynamics with competing ligands or antibody-induced receptor cross-link in the developing rat brain were sufficient to regulate the adult acoustic startle reflex and compensate for an early pathological challenge. Collectively, we show that the NMDAR trafficking is markedly altered in all clinically relevant models of psychosis, opening new avenues of therapeutical strategies.
Collapse
|
27
|
Minchew HM, Radabaugh HL, LaPorte ML, Free KE, Cheng JP, Bondi CO. A combined therapeutic regimen of citalopram and environmental enrichment ameliorates attentional set-shifting performance after brain trauma. Eur J Pharmacol 2021; 904:174174. [PMID: 34004206 PMCID: PMC8906929 DOI: 10.1016/j.ejphar.2021.174174] [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] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 04/29/2021] [Accepted: 05/11/2021] [Indexed: 01/09/2023]
Abstract
Traumatic brain injuries (TBI) have led to lasting deficits for an estimated 5.3 million American patients. Effective therapies for these patients remain scarce and each of the clinical trials stemming from success in experimental models has failed. We believe that the failures may be, in part, due to the lack of preclinical assessment of cognitive domains that widely affect clinical TBI. Specifically, the behavioral tasks in the TBI literature often do not focus on common executive impairments related to the frontal lobe such as cognitive flexibility. In previous work, we have demonstrated that the attentional set-shifting test (AST), a task analogous to the clinically-employed Wisconsin Card Sorting Test (WCST), could be used to identify cognitive flexibility impairments following controlled cortical impact (CCI) injury. In this study, we hypothesized that both the administration of the antidepressant drug citalopram (CIT) and exposure to a preclinical model of neurorehabilitation, environmental enrichment (EE), would attenuate cognitive performance deficits on AST when provided alone and lead to greater benefits when administered in combination. Adult male rats were subjected to a moderate-severe CCI or sham injury. Rats were randomly divided into experimental groups that included surgical injury, drug therapy, and housing condition. We observed that both CIT and EE provided significant cognitive recovery when administered alone and reversal learning performance recovery increased the most when the therapies were combined (p < 0.05). Ongoing studies continue to evaluate novel ways of assessing more clinically relevant measurements of high order cognitive TBI-related impairments in the rat model.
Collapse
Affiliation(s)
- Heather M Minchew
- Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, 15213, USA; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Hannah L Radabaugh
- Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, 15213, USA; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Megan L LaPorte
- Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, 15213, USA; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Kristin E Free
- Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, 15213, USA; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Jeffrey P Cheng
- Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, 15213, USA; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Corina O Bondi
- Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, 15213, USA; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, 15213, USA; Neurobiology, University of Pittsburgh, Pittsburgh, PA, 15213, USA; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
28
|
Peters KZ, Zlebnik NE, Cheer JF. Cannabis exposure during adolescence: A uniquely sensitive period for neurobiological effects. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 161:95-120. [PMID: 34801175 DOI: 10.1016/bs.irn.2021.07.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Adolescence is a crucial developmental period where neural circuits are refined and the brain is especially vulnerable to external insults. The endocannabinoid (eCB) system undergoes changes during adolescence which affect the way in which it modulates the development of other systems, in particular dopamine circuits, which show protracted development into adolescence. Given the rise of cannabis use by adolescents and young people, as well as variants containing increasingly higher concentrations of THC, it is now crucial to understand the unique effects of adolescent exposure to cannabis on the developing brain and it might shape future adult vulnerabilities to conditions such as psychosis, schizophrenia, addiction and more. Here we discuss the development of the eCB system across the lifespan, how CB1 receptors modulate dopamine release and potential neurobiological and behavioral effects of adolescent THC exposure on the developing brain such as alterations in excitatory/inhibitory balance during this developmental period.
Collapse
Affiliation(s)
- K Z Peters
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States; Sussex Neuroscience, School of Psychology, University of Sussex, Falmer, United Kingdom.
| | - N E Zlebnik
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - J F Cheer
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States; Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States; Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
29
|
Klink PC, Aubry JF, Ferrera VP, Fox AS, Froudist-Walsh S, Jarraya B, Konofagou EE, Krauzlis RJ, Messinger A, Mitchell AS, Ortiz-Rios M, Oya H, Roberts AC, Roe AW, Rushworth MFS, Sallet J, Schmid MC, Schroeder CE, Tasserie J, Tsao DY, Uhrig L, Vanduffel W, Wilke M, Kagan I, Petkov CI. Combining brain perturbation and neuroimaging in non-human primates. Neuroimage 2021; 235:118017. [PMID: 33794355 PMCID: PMC11178240 DOI: 10.1016/j.neuroimage.2021.118017] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 03/07/2021] [Accepted: 03/22/2021] [Indexed: 12/11/2022] Open
Abstract
Brain perturbation studies allow detailed causal inferences of behavioral and neural processes. Because the combination of brain perturbation methods and neural measurement techniques is inherently challenging, research in humans has predominantly focused on non-invasive, indirect brain perturbations, or neurological lesion studies. Non-human primates have been indispensable as a neurobiological system that is highly similar to humans while simultaneously being more experimentally tractable, allowing visualization of the functional and structural impact of systematic brain perturbation. This review considers the state of the art in non-human primate brain perturbation with a focus on approaches that can be combined with neuroimaging. We consider both non-reversible (lesions) and reversible or temporary perturbations such as electrical, pharmacological, optical, optogenetic, chemogenetic, pathway-selective, and ultrasound based interference methods. Method-specific considerations from the research and development community are offered to facilitate research in this field and support further innovations. We conclude by identifying novel avenues for further research and innovation and by highlighting the clinical translational potential of the methods.
Collapse
Affiliation(s)
- P Christiaan Klink
- Department of Vision & Cognition, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands.
| | - Jean-François Aubry
- Physics for Medicine Paris, Inserm U1273, CNRS UMR 8063, ESPCI Paris, PSL University, Paris, France
| | - Vincent P Ferrera
- Department of Neuroscience & Department of Psychiatry, Columbia University Medical Center, New York, NY, USA; Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Andrew S Fox
- Department of Psychology & California National Primate Research Center, University of California, Davis, CA, USA
| | | | - Béchir Jarraya
- NeuroSpin, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Institut National de la Santé et de la Recherche Médicale (INSERM), Cognitive Neuroimaging Unit, Université Paris-Saclay, France; Foch Hospital, UVSQ, Suresnes, France
| | - Elisa E Konofagou
- Ultrasound and Elasticity Imaging Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY, USA; Department of Radiology, Columbia University, New York, NY, USA
| | - Richard J Krauzlis
- Laboratory of Sensorimotor Research, National Eye Institute, Bethesda, MD, USA
| | - Adam Messinger
- Laboratory of Brain and Cognition, National Institute of Mental Health, Bethesda, MD, USA
| | - Anna S Mitchell
- Department of Experimental Psychology, Oxford University, Oxford, United Kingdom
| | - Michael Ortiz-Rios
- Newcastle University Medical School, Newcastle upon Tyne NE1 7RU, United Kingdom; German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| | - Hiroyuki Oya
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, USA; Department of Neurosurgery, University of Iowa, Iowa city, IA, USA
| | - Angela C Roberts
- Department of Physiology, Development and Neuroscience, Cambridge University, Cambridge, United Kingdom
| | - Anna Wang Roe
- Interdisciplinary Institute of Neuroscience and Technology, School of Medicine, Zhejiang University, Hangzhou 310029, China
| | | | - Jérôme Sallet
- Department of Experimental Psychology, Oxford University, Oxford, United Kingdom; Univ Lyon, Université Lyon 1, Inserm, Stem Cell and Brain Research Institute, U1208 Bron, France; Wellcome Centre for Integrative Neuroimaging, Department of Experimental Psychology, University of Oxford, Oxford, United Kingdom
| | - Michael Christoph Schmid
- Newcastle University Medical School, Newcastle upon Tyne NE1 7RU, United Kingdom; Faculty of Science and Medicine, University of Fribourg, Chemin du Musée 5, CH-1700 Fribourg, Switzerland
| | - Charles E Schroeder
- Nathan Kline Institute, Orangeburg, NY, USA; Columbia University, New York, NY, USA
| | - Jordy Tasserie
- NeuroSpin, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Institut National de la Santé et de la Recherche Médicale (INSERM), Cognitive Neuroimaging Unit, Université Paris-Saclay, France
| | - Doris Y Tsao
- Division of Biology and Biological Engineering, Tianqiao and Chrissy Chen Institute for Neuroscience; Howard Hughes Medical Institute; Computation and Neural Systems, Caltech, Pasadena, CA, USA
| | - Lynn Uhrig
- NeuroSpin, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Institut National de la Santé et de la Recherche Médicale (INSERM), Cognitive Neuroimaging Unit, Université Paris-Saclay, France
| | - Wim Vanduffel
- Laboratory for Neuro- and Psychophysiology, Neurosciences Department, KU Leuven Medical School, Leuven, Belgium; Leuven Brain Institute, KU Leuven, Leuven Belgium; Harvard Medical School, Boston, MA, USA; Massachusetts General Hospital, Martinos Center for Biomedical Imaging, Charlestown, MA, USA
| | - Melanie Wilke
- German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany; Department of Cognitive Neurology, University Medicine Göttingen, Göttingen, Germany
| | - Igor Kagan
- German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany.
| | - Christopher I Petkov
- Newcastle University Medical School, Newcastle upon Tyne NE1 7RU, United Kingdom.
| |
Collapse
|
30
|
Ferretjans R, de Souza RP, Panizzutti B, Ferrari P, Mantovani L, de Campos-Carli SM, Santos RR, Guimarães FC, Teixeira AL, Gama CS, Salgado JV. Cannabinoid receptor gene polymorphisms and cognitive performance in patients with schizophrenia and controls. ACTA ACUST UNITED AC 2021; 44:26-34. [PMID: 34190825 PMCID: PMC8827365 DOI: 10.1590/1516-4446-2020-1650] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 02/20/2021] [Indexed: 12/18/2022]
Abstract
Objective: To test the hypothesis that genetic variations of cannabinoid receptors contribute to the pathophysiology of cognitive deficits in schizophrenia. Methods: In this genetic association case-control study, cannabinoid receptor polymorphisms CNR1 rs12720071 and CNR2 rs2229579 were tested for association with neurocognitive performance in 69 patients with schizophrenia and 45 healthy controls. Neurocognition was assessed by the Brief Assessment of Cognition in Schizophrenia (BACS). Results: We found a consistent association between CNR1 rs12720071 polymorphism and the cognitive performance of patients in several cognitive domains. Patients with C/C polymorphism presented significantly worse performance in motor speed, verbal fluency, attention/processing speed and reasoning/problem solving. Conclusion: Although limited, our data support the hypothesis that CNR1 variations may be associated with the pathogenesis of cognitive deficits of schizophrenia.
Collapse
Affiliation(s)
- Rodrigo Ferretjans
- Programa Interdisciplinar de Pós-Graduação em Neurociências, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Renan P de Souza
- Programa de Pós-Graduação em Genética, Instituto de Ciências Biológicas, UFMG, Belo Horizonte, MG, Brazil
| | - Bruna Panizzutti
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Barwon Health, Geelong, Australia.,Centre for Molecular and Medical Research, School of Medicine, Deakin University, Geelong, Australia
| | - Pâmela Ferrari
- Programa de Pós-Graduação em Psiquiatria e Ciências do Comportamento (PPGPSIQ), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.,Laboratório de Psiquiatria Molecular, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, RS, Brazil
| | - Lucas Mantovani
- Programa Interdisciplinar de Pós-Graduação em Neurociências, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Salvina M de Campos-Carli
- Programa Interdisciplinar de Pós-Graduação em Neurociências, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Rafael R Santos
- Programa Interdisciplinar de Pós-Graduação em Neurociências, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Fernanda C Guimarães
- Programa Interdisciplinar de Pós-Graduação em Neurociências, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Antonio L Teixeira
- Instituto de Ensino e Pesquisa, Santa Casa BH, Belo Horizonte, MG, Brazil.,Neuropsychiatry Program, UTHealth Houston, TX, USA
| | - Clarissa S Gama
- Centre for Molecular and Medical Research, School of Medicine, Deakin University, Geelong, Australia.,Programa de Pós-Graduação em Psiquiatria e Ciências do Comportamento (PPGPSIQ), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - João V Salgado
- Programa Interdisciplinar de Pós-Graduação em Neurociências, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil.,Departamento de Morfologia, Instituto de Ciências Biológicas, UFMG, Belo Horizonte, MG, Brazil
| |
Collapse
|
31
|
Wang P, Li M, Zhao A, Ma J. Application of animal experimental models in the research of schizophrenia. Am J Med Genet B Neuropsychiatr Genet 2021; 186:209-227. [PMID: 34155806 DOI: 10.1002/ajmg.b.32863] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 05/04/2021] [Accepted: 05/31/2021] [Indexed: 12/14/2022]
Abstract
Schizophrenia is a relatively common but serious mental illness that results in a heavy burden to patients, their families, and society. The disease can be triggered by multiple factors, while the specific pathogenesis remains unclear. The development of effective therapeutic drugs for schizophrenia relies on a comprehensive understanding of the basic biology and pathophysiology of the disease. Therefore, effective animal experimental models play a vital role in the study of schizophrenia. Based on different molecular mechanisms and modeling methods, the currently used experimental animal experimental models of schizophrenia can be divided into four categories that can better simulate the clinical symptoms and the interplay between susceptible genes and the environment: neurodevelopmental, drug-induced, genetic-engineering, and genetic-environmental interaction of animal experimental models. Each of these categories contains multiple subtypes, which has its own advantages and disadvantages and therefore requires careful selection in a research application. The emergence and utilization of these models are promising in the prediction of the risk of schizophrenia at the molecular level, which will shed light on effective and targeted treatment at the genetic level.
Collapse
Affiliation(s)
- Pengjie Wang
- Medical Research Center, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, China.,Department of Electron Microscope, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Manling Li
- School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Gui Yang, Guizhou, China
| | - Aizhen Zhao
- Medical Research Center, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, China
| | - Jie Ma
- Medical Research Center, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, China.,Department of Electron Microscope, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| |
Collapse
|
32
|
Ahn S, Choi Y, Choi W, Jo YT, Kim H, Lee J, Joo SW. Effects of comorbid alcohol use disorder on the clinical outcomes of first-episode schizophrenia: a nationwide population-based study. Ann Gen Psychiatry 2021; 20:32. [PMID: 34051807 PMCID: PMC8164281 DOI: 10.1186/s12991-021-00353-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/24/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Alcohol use disorder (AUD) is a common psychiatric comorbidity in schizophrenia, associated with poor clinical outcomes and medication noncompliance. Most previous studies on the effect of alcohol use in patients with schizophrenia had limitations of small sample size or a cross-sectional design. Therefore, we used a nationwide population database to investigate the impact of AUD on clinical outcomes of schizophrenia. METHODS Data from the Health Insurance Review Agency database in South Korea from January 1, 2007 to December 31, 2016 were used. Among 64,442 patients with first-episode schizophrenia, 1598 patients with comorbid AUD were selected based on the diagnostic code F10. We performed between- and within-group analyses to compare the rates of psychiatric admissions and emergency room (ER) visits, and medication possession ratio (MPR) between the patients with comorbid AUD and control patients matched for the onset age, sex, and observation period. RESULTS The rates of psychiatric admissions and ER visits in both groups decreased after the time point of diagnosis of AUD; however, the decrease was significantly greater in the patients with comorbid AUD compared to the control patients. While the comorbid AUD group showed an increase in MPR after the diagnosis of AUD, MPR decreased in the control group. The rates of psychiatric admissions, ER visits, and MPR were worse in the comorbid AUD group both before and after the diagnosis of AUD. CONCLUSIONS The results emphasize an importance of psychiatric comorbidities, especially AUD, in first-episode schizophrenia and the necessity of further research for confirmative findings of the association of AUD with clinical outcomes of schizophrenia.
Collapse
Affiliation(s)
- Soojin Ahn
- Department of Psychiatry, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-Ro 43-Gil, SongPa-Gu, Seoul, 05505, Republic of Korea
| | - Youngjae Choi
- Department of Psychiatry, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-Ro 43-Gil, SongPa-Gu, Seoul, 05505, Republic of Korea
| | - Woohyeok Choi
- Department of Psychiatry, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-Ro 43-Gil, SongPa-Gu, Seoul, 05505, Republic of Korea
| | - Young Tak Jo
- Department of Psychiatry, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-Ro 43-Gil, SongPa-Gu, Seoul, 05505, Republic of Korea
| | - Harin Kim
- Department of Psychiatry, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-Ro 43-Gil, SongPa-Gu, Seoul, 05505, Republic of Korea
| | - Jungsun Lee
- Department of Psychiatry, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-Ro 43-Gil, SongPa-Gu, Seoul, 05505, Republic of Korea
| | - Sung Woo Joo
- Department of Psychiatry, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-Ro 43-Gil, SongPa-Gu, Seoul, 05505, Republic of Korea.
| |
Collapse
|
33
|
Martínez-Torres NI, Vázquez-Hernández N, Martín-Amaya-Barajas FL, Flores-Soto M, González-Burgos I. Ibotenic acid induced lesions impair the modulation of dendritic spine plasticity in the prefrontal cortex and amygdala, a phenomenon that underlies working memory and social behavior. Eur J Pharmacol 2021; 896:173883. [PMID: 33513334 DOI: 10.1016/j.ejphar.2021.173883] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/12/2021] [Accepted: 01/14/2021] [Indexed: 01/08/2023]
Abstract
The lesions induced by Ibotenic acid (IA) emulate some of the symptoms associated with schizophrenia, such as impaired working memory that is predominantly organized by the medial prefrontal cortex (mPFC), or difficulties in social interactions that aremainly organized by the amygdala (AMG). The plastic capacity of dendritic spines in neurons of the mPFC and AMG is modulated by molecules that participate in the known deterioration of working memory, although the influence of these on the socialization of schizophrenic patients is unknown. Here, the effect of a neonatal IA induced lesion on social behavior and working memory was evaluated in adult rats, along with the changes in cytoarchitecture of dendritic spines and their protein content, specifically the postsynaptic density protein 95 (PSD-95), Synaptophysin (Syn), AMPA receptors, and brain-derived neurotrophic factor (BDNF). Both working memory and social behavior were impaired, and the density of the spines, as well as their PSD-95, Syn, AMPA receptor and BDNF content was lower in IA lesioned animals. The proportional density of thin, mushroom, stubby and wide spines resulted in plastic changes that suggest the activation of compensatory processes in the face of the adverse effects of the lesion. In addition, the reduction in the levels of the modulating factors also suggests that the signaling pathways in which such factors are implicated would be altered in the brains of patients with schizophrenia. Accordingly, the experimental study of such signaling pathways is likely to aid the development of more effective pharmacological strategies for the treatment of schizophrenia.
Collapse
Affiliation(s)
- Néstor I Martínez-Torres
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, Guadalajara, Jal., Mexico; Centro Universitario del Norte, Universidad de Guadalajara, Colotlán, Jal., Mexico
| | - Nallely Vázquez-Hernández
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, Guadalajara, Jal., Mexico
| | | | - Mario Flores-Soto
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, Guadalajara, Jal., Mexico
| | - Ignacio González-Burgos
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, Guadalajara, Jal., Mexico.
| |
Collapse
|
34
|
Damme KSF, Schiffman J, Ellman LM, Mittal VA. Sensorimotor and Activity Psychosis-Risk (SMAP-R) Scale: An Exploration of Scale Structure With Replication and Validation. Schizophr Bull 2021; 47:332-343. [PMID: 33047134 PMCID: PMC7965079 DOI: 10.1093/schbul/sbaa138] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Sensorimotor abnormalities precede and predict the onset of psychosis. Despite the practical utility of sensorimotor abnormalities for early identification, prediction, and individualized medicine applications, there is currently no dedicated self-report instrument designed to capture these important behaviors. The current study assessed and validated a questionnaire designed for use in individuals at clinical high-risk for psychosis (CHR). METHODS The current study included both exploratory (n = 3009) and validation (n = 439) analytic datasets-that included individuals identified as meeting criteria for a CHR syndrome (n = 84)-who completed the novel Sensorimotor Abnormalities and Psychosis-Risk (SMAP-R) Scale, clinical interviews and a finger-tapping task. The structure of the scale and reliability of items were consistent across 2 analytic datasets. The resulting scales were assessed for discriminant validity across CHR, community sample non-psychiatric volunteer, and clinical groups. RESULTS The scale showed a consistent structure across 2 analytic datasets subscale structure. The resultant subscale structure was consistent with conceptual models of sensorimotor pathology in psychosis (coordination and dyskinesia) in both the exploratory and the validation analytic dataset. Further, these subscales showed discriminant, predictive, and convergent validity. The sensorimotor abnormality scales discriminated CHR from community sample non-psychiatric controls and clinical samples. Finally, these subscales predicted to risk calculator scores and showed convergent validity with sensorimotor performance on a finger-tapping task. CONCLUSION The SMAP-R scale demonstrated good internal, discriminant, predictive, and convergent validity, and subscales mapped on to conceptually relevant sensorimotor circuits. Features of the scale may facilitate widespread incorporation of sensorimotor screening into psychosis-risk research and practice.
Collapse
Affiliation(s)
- Katherine S F Damme
- Department of Psychology, Northwestern University, Evanston, IL
- Institute for Innovations in Developmental Sciences (DevSci), Northwestern University, Evanston and Chicago, IL
| | | | - Lauren M Ellman
- Department of Psychology, Temple University, Philadelphia, PA
| | - Vijay A Mittal
- Department of Psychology, Northwestern University, Evanston, IL
- Institute for Innovations in Developmental Sciences (DevSci), Northwestern University, Evanston and Chicago, IL
- Department of Psychiatry, Northwestern University, Chicago, IL
- Medical Social Sciences, Northwestern University, Chicago, IL
- Institute for Policy Research (IPR), Northwestern University, Chicago, IL
| |
Collapse
|
35
|
Fernández-Teruel A. Conflict between Threat Sensitivity and Sensation Seeking in the Adolescent Brain: Role of the Hippocampus, and Neurobehavioural Plasticity Induced by Pleasurable Early Enriched Experience. Brain Sci 2021; 11:brainsci11020268. [PMID: 33672653 PMCID: PMC7924176 DOI: 10.3390/brainsci11020268] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 02/05/2021] [Accepted: 02/14/2021] [Indexed: 01/28/2023] Open
Abstract
Adolescence is characterized both by the exacerbation of the experience of anxiety, fear or threat, on one hand, and by increased reward seeking (reward sensitivity) and risk taking on the other hand. The rise of these apparently opposite processes, i.e., threat-related anxiety and reward-related sensation seeking, seems to stem from a relatively decreased top-down inhibition of amygdala and striatal circuits by regulatory systems (e.g., prefrontal cortex, hippocampus) that mature later. The present commentary article aims to discuss recent related literature and focusses on two main issues: (i) the septo-hippocampal system (in particular the ventral hippocampus) might be a crucial region for the regulation of approach–avoidance conflict and also for the selection of the most appropriate responses during adolescence, and (ii) developmental studies involving early-life pleasurable-enriched experience (as opposed to early-life adversity) might be a useful study paradigm in order to decipher whether neuroplasticity induced by such experiences (for example, in the hippocampus and associated circuitry) may lead to better top-down inhibition and more “balanced” adolescent responses to environmental demands.
Collapse
Affiliation(s)
- Alberto Fernández-Teruel
- Department of Psychiatry & Forensic Medicine, Medical Psychology Unit, School of Medicine & Institute of Neurosciences, Autonomous University of Barcelona, Bellaterra, 08193 Barcelona, Spain
| |
Collapse
|
36
|
Jenkins BW, Khokhar JY. Cannabis Use and Mental Illness: Understanding Circuit Dysfunction Through Preclinical Models. Front Psychiatry 2021; 12:597725. [PMID: 33613338 PMCID: PMC7892618 DOI: 10.3389/fpsyt.2021.597725] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 01/11/2021] [Indexed: 12/27/2022] Open
Abstract
Patients with a serious mental illness often use cannabis at higher rates than the general population and are also often diagnosed with cannabis use disorder. Clinical studies reveal a strong association between the psychoactive effects of cannabis and the symptoms of serious mental illnesses. Although some studies purport that cannabis may treat mental illnesses, others have highlighted the negative consequences of use for patients with a mental illness and for otherwise healthy users. As epidemiological and clinical studies are unable to directly infer causality or examine neurobiology through circuit manipulation, preclinical animal models remain a valuable resource for examining the causal effects of cannabis. This is especially true considering the diversity of constituents in the cannabis plant contributing to its effects. In this mini-review, we provide an updated perspective on the preclinical evidence of shared neurobiological mechanisms underpinning the dual diagnosis of cannabis use disorder and a serious mental illness. We present studies of cannabinoid exposure in otherwise healthy rodents, as well as rodent models of schizophrenia, depression, and bipolar disorder, and the resulting impact on electrophysiological indices of neural circuit activity. We propose a consolidated neural circuit-based understanding of the preclinical evidence to generate new hypotheses and identify novel therapeutic targets.
Collapse
Affiliation(s)
| | - Jibran Y. Khokhar
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
37
|
Tendilla-Beltrán H, Sanchez-Islas NDC, Marina-Ramos M, Leza JC, Flores G. The prefrontal cortex as a target for atypical antipsychotics in schizophrenia, lessons of neurodevelopmental animal models. Prog Neurobiol 2020; 199:101967. [PMID: 33271238 DOI: 10.1016/j.pneurobio.2020.101967] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 10/10/2020] [Accepted: 11/22/2020] [Indexed: 02/06/2023]
Abstract
Prefrontal cortex (PFC) inflammatory imbalance, oxidative/nitrosative stress (O/NS) and impaired neuroplasticity in schizophrenia are thought to have neurodevelopmental origins. Animal models are not only useful to test this hypothesis, they are also effective to establish a relationship among brain disturbances and behavior with the atypical antipsychotics (AAPs) effects. Here we review data of PFC post-mortem and in vivo neuroimaging, human induced pluripotent stem cells (hiPSC), and peripheral blood studies of inflammatory, O/NS, and neuroplasticity alterations in the disease as well as about their modulation by AAPs. Moreover, we reviewed the PFC alterations and the AAP mechanisms beyond their canonical antipsychotic action in four neurodevelopmental animal models relevant to the study of schizophrenia with a distinct approach in the generation of schizophrenia-like phenotypes, but all converge in O/NS and altered neuroplasticity in the PFC. These animal models not only reinforce the neurodevelopmental risk factor model of schizophrenia but also arouse some novel potential therapeutic targets for the disease including the reestablishment of the antioxidant response by the perineuronal nets (PNNs) and the nuclear factor erythroid 2-related factor (Nrf2) pathway, as well as the dendritic spine dynamics in the PFC pyramidal cells.
Collapse
Affiliation(s)
- Hiram Tendilla-Beltrán
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico; Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional (IPN), CDMX, Mexico
| | | | - Mauricio Marina-Ramos
- Departamento de Ciencias de la Salud, Universidad Popular Autónoma del Estado de Puebla, Puebla, Mexico
| | - Juan C Leza
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Instituto Universitario de Investigación en Neuroquímica (IUIN), UCM. Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Investigación Sanitaria Hospital, 12 de Octubre (Imas12), Madrid, Spain
| | - Gonzalo Flores
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico.
| |
Collapse
|
38
|
Langova V, Vales K, Horka P, Horacek J. The Role of Zebrafish and Laboratory Rodents in Schizophrenia Research. Front Psychiatry 2020; 11:703. [PMID: 33101067 PMCID: PMC7500259 DOI: 10.3389/fpsyt.2020.00703] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 07/03/2020] [Indexed: 12/11/2022] Open
Abstract
Schizophrenia is a severe disorder characterized by positive, negative and cognitive symptoms, which are still not fully understood. The development of efficient antipsychotics requires animal models of a strong validity, therefore the aims of the article were to summarize the construct, face and predictive validity of schizophrenia models based on rodents and zebrafish, to compare the advantages and disadvantages of these models, and to propose future directions in schizophrenia modeling and indicate when it is reasonable to combine these models. The advantages of rodent models stem primarily from the high homology between rodent and human physiology, neurochemistry, brain morphology and circuitry. The advantages of zebrafish models stem in the high fecundity, fast development and transparency of the embryo. Disadvantages of both models originate in behavioral repertoires not allowing specific symptoms to be modeled, even when the models are combined. Especially modeling the verbal component of certain positive, negative and cognitive symptoms is currently impossible.
Collapse
Affiliation(s)
- Veronika Langova
- Translational Neuroscience, National Institute of Mental Health, Prague, Czechia
- Third Faculty of Medicine, Charles University, Prague, Czechia
| | - Karel Vales
- Translational Neuroscience, National Institute of Mental Health, Prague, Czechia
| | - Petra Horka
- Institute for Environmental Studies, Faculty of Science, Charles University, Prague, Czechia
| | - Jiri Horacek
- Third Faculty of Medicine, Charles University, Prague, Czechia
- Brain Electrophysiology, National Institute of Mental Health, Prague, Czechia
| |
Collapse
|
39
|
Nakagawa K, Yoshino H, Ogawa Y, Yamamuro K, Kimoto S, Noriyama Y, Makinodan M, Yamashita M, Saito Y, Kishimoto T. Maternal Immune Activation Affects Hippocampal Excitatory and Inhibitory Synaptic Transmission in Offspring From an Early Developmental Period to Adulthood. Front Cell Neurosci 2020; 14:241. [PMID: 32903758 PMCID: PMC7438877 DOI: 10.3389/fncel.2020.00241] [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] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 07/09/2020] [Indexed: 12/31/2022] Open
Abstract
One of the risk factors for schizophrenia is maternal infection. We have previously shown that Polyriboinosinic-polyribocytidylic acid (poly I:C) induced maternal immune activation in mice caused histological changes in the hippocampal CA1 area of offspring during the developmental period and impaired sensorimotor gating in offspring during adulthood, resulting in behavioral changes. However, it remains unclear how maternal immune activation functionally impacts the hippocampal neuronal activity of offspring. We studied the effect of prenatal poly I:C treatment on synaptic transmission of hippocampal CA1 pyramidal cells in postnatal and adult offspring. Treatment with poly I:C diminished excitatory and enhanced inhibitory (GABAergic) synaptic transmission on pyramidal cells in adult offspring. During the early developmental period, we still observed that treatment with poly I:C decreased excitatory synaptic transmission and potentially increased GABAergic synaptic transmission, which was uncovered under a condition of high extracellular potassium-activated neurons. In conclusion, we demonstrate that maternal immune activation decreased excitatory and increased inhibitory synaptic transmission on hippocampal pyramidal cells from an early developmental period to adulthood, which could result in net inhibition in conjunction with poor functional organization and integration of hippocampal circuits.
Collapse
Affiliation(s)
- Keiju Nakagawa
- Department of Psychiatry, Nara Medical University, Kashihara, Japan
| | - Hiroki Yoshino
- Department of Psychiatry, Nara Medical University, Kashihara, Japan
| | - Yoichi Ogawa
- Department of Neurophysiology, Nara Medical University, Kashihara, Japan
| | | | - Sohei Kimoto
- Department of Psychiatry, Nara Medical University, Kashihara, Japan
| | | | - Manabu Makinodan
- Department of Psychiatry, Nara Medical University, Kashihara, Japan
| | - Masayuki Yamashita
- Center for Medical Science, International University of Health and Welfare, Otawara, Japan
| | - Yasuhiko Saito
- Department of Neurophysiology, Nara Medical University, Kashihara, Japan
| | | |
Collapse
|
40
|
Penny TR, Pham Y, Sutherland AE, Mihelakis JG, Lee J, Jenkin G, Fahey MC, Miller SL, McDonald CA. Multiple doses of umbilical cord blood cells improve long-term brain injury in the neonatal rat. Brain Res 2020; 1746:147001. [PMID: 32585139 DOI: 10.1016/j.brainres.2020.147001] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 06/11/2020] [Accepted: 06/17/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Hypoxic ischemic (HI) insults during pregnancy and birth can result in neurodevelopmental disorders, such as cerebral palsy. We have previously shown that a single dose of umbilical cord blood (UCB) cells is effective at reducing short-term neuroinflammation and improves short and long-term behavioural outcomes in rat pups. A single dose of UCB was not able to modulate long-term neuroinflammation or brain tissue loss. In this study we examined whether multiple doses of UCB can modulate neuroinflammation, decrease cerebral tissue damage and improve behavioural outcomes when followed up long-term. METHODS HI injury was induced in postnatal day 10 (PND10) rat pups using the Rice-Vannucci method of carotid artery ligation. Pups received either 1 dose (PND11), or 3 doses (PND11, 13, 20) of UCB cells. Rats were followed with behavioural testing, to assess both motor and cognitive outcomes. On PND50, brains were collected for analysis. RESULTS HI brain injury in rat pups caused significant behavioural deficits. These deficits were significantly improved by multiple doses of UCB. HI injury resulted in a significant decrease in brain weight and left hemisphere tissue, which was improved by multiple doses of UCB. HI resulted in increased cerebral apoptosis, loss of neurons and upregulation of activated microglia. Multiple doses of UCB modulated these neuropathologies. A single dose of UCB at PND11 did not improve behavioural or neuropathological outcomes. CONCLUSIONS Treatment with repeated doses of UCB is more effective than a single dose for reducing tissue damage, improving brain pathology and restoring behavioural deficits following perinatal brain injury.
Collapse
Affiliation(s)
- Tayla R Penny
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Yen Pham
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Amy E Sutherland
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Jamie G Mihelakis
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Joohyung Lee
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Australia
| | - Graham Jenkin
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Michael C Fahey
- Department of Paediatrics, Monash University, Clayton, Victoria, Australia
| | - Suzanne L Miller
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Courtney A McDonald
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.
| |
Collapse
|
41
|
Abstract
The onset of schizophrenia is usually in late adolescence or early adulthood. However, accumulating evidence has suggested that the disease condition is an outcome of gene-environment interactions that act in neural development during early life and adolescence. Some children who later develop schizophrenia have early developmental and educational and social challenges. Some patients with schizophrenia have an abundance of nonspecific neurologic soft signs and minor physical anomalies. Adolescence is a sensitive period of increased neuronal plasticity. It is important to consider early detection and intervention from the prodromal stage to early disease to prevent its devastating long-term consequences.
Collapse
|
42
|
Saito M, Smiley JF, Hui M, Masiello K, Betz J, Ilina M, Saito M, Wilson DA. Neonatal Ethanol Disturbs the Normal Maturation of Parvalbumin Interneurons Surrounded by Subsets of Perineuronal Nets in the Cerebral Cortex: Partial Reversal by Lithium. Cereb Cortex 2020; 29:1383-1397. [PMID: 29462278 DOI: 10.1093/cercor/bhy034] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 01/02/2018] [Accepted: 01/25/2018] [Indexed: 02/07/2023] Open
Abstract
Reduction in parvalbumin-positive (PV+) interneurons is observed in adult mice exposed to ethanol at postnatal day 7 (P7), a late gestation fetal alcohol spectrum disorder model. To evaluate whether PV+ cells are lost, or PV expression is reduced, we quantified PV+ and associated perineuronal net (PNN)+ cell densities in barrel cortex. While PNN+ cell density was not reduced by P7 ethanol, PV cell density decreased by 25% at P90 with no decrease at P14. PNN+ cells in controls were virtually all PV+, whereas more than 20% lacked PV in ethanol-treated adult animals. P7 ethanol caused immediate apoptosis in 10% of GFP+ cells in G42 mice, which express GFP in a subset of PV+ cells, and GFP+ cell density decreased by 60% at P90 without reduction at P14. The ethanol effect on PV+ cell density was attenuated by lithium treatment at P7 or at P14-28. Thus, reduced PV+ cell density may be caused by disrupted cell maturation, in addition to acute apoptosis. This effect may be regionally specific: in the dentate gyrus, P7 ethanol reduced PV+ cell density by 70% at P14 and both PV+ and PNN+ cell densities by 50% at P90, and delayed lithium did not alleviate ethanol's effect.
Collapse
Affiliation(s)
- Mariko Saito
- Division of Neurochemistry, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA.,Department of Psychiatry, NYU School of Medicine, New York, NY, USA
| | - John F Smiley
- Division of Neurochemistry, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA.,Department of Psychiatry, NYU School of Medicine, New York, NY, USA
| | - Maria Hui
- Division of Neurochemistry, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Kurt Masiello
- Division of Neurochemistry, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Judith Betz
- Division of Neurochemistry, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Maria Ilina
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Mitsuo Saito
- Department of Psychiatry, NYU School of Medicine, New York, NY, USA.,Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Donald A Wilson
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA.,Department of Child and Adolescent Psychiatry, NYU School of Medicine, New York, NY, USA
| |
Collapse
|
43
|
MK-801 Exposure during Adolescence Elicits Enduring Disruption of Prefrontal E-I Balance and Its Control of Fear Extinction Behavior. J Neurosci 2020; 40:4881-4887. [PMID: 32430298 DOI: 10.1523/jneurosci.0581-20.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/05/2020] [Accepted: 05/11/2020] [Indexed: 01/04/2023] Open
Abstract
Understanding how disruption of prefrontal cortex (PFC) maturation during adolescence is crucial to reveal which neural processes could contribute to the onset of psychiatric disorders that display frontal cortical deficits. Of particular interest is the gain of GABAergic function in the PFC during adolescence and its susceptibility to the impact of transient blockade of NMDA receptor function. Here we assessed whether exposure to MK-801 during adolescence in male rats triggers a state of excitatory-inhibitory imbalance in the PFC that limits its functional capacity to regulate behavior in adulthood. Recordings from PFC brain slices revealed that MK-801 exposure during adolescence preferentially reduces the presynaptic functionality of GABAergic activity over that of excitatory synapses. As a result, an imbalance of excitatory-inhibitory synaptic activity emerges in the PFC that correlates linearly with the GABAergic deficit. Notably, the data also suggest that the diminished prefrontal GABAergic function could arise from a deficit in the recruitment of fast-spiking interneurons by excitatory inputs during adolescence. At the behavioral level, MK-801 exposure during adolescence did not disrupt the acquisition of trace fear conditioning, but markedly increased the level of freezing response during extinction testing. Infusion of the GABAA receptor-positive allosteric modulator Indiplon into the PFC before extinction testing reduced the level of freezing response in MK-801-treated rats to control levels. Collectively, the results indicate NMDA receptor signaling during adolescence enables the gain of prefrontal GABAergic function, which is required for maintaining proper excitatory-inhibitory balance in the PFC and its control of behavioral responses.SIGNIFICANCE STATEMENT A developmental disruption of prefrontal cortex maturation has been implicated in the pathophysiology of cognitive deficits in psychiatric disorders. Of particular interest is the susceptibility of the local GABAergic circuit to the impact of transient disruption of NMDA receptors. Here we found that NMDA receptor signaling is critical to enable the gain of prefrontal GABAergic transmission during adolescence for maintaining proper levels of excitatory-inhibitory balance in the PFC and its control of behavior.
Collapse
|
44
|
Bravo L, Llorca-Torralba M, Suárez-Pereira I, Berrocoso E. Pain in neuropsychiatry: Insights from animal models. Neurosci Biobehav Rev 2020; 115:96-115. [PMID: 32437745 DOI: 10.1016/j.neubiorev.2020.04.029] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 04/11/2020] [Accepted: 04/23/2020] [Indexed: 02/08/2023]
Abstract
Pain is the most common symptom reported in clinical practice, meaning that it is associated with many pathologies as either the origin or a consequence of other illnesses. Furthermore, pain is a complex emotional and sensorial experience, as the correspondence between pain and body damage varies considerably. While these issues are widely acknowledged in clinical pain research, until recently they have not been extensively considered when exploring animal models, important tools for understanding pain pathophysiology. Interestingly, chronic pain is currently considered a risk factor to suffer psychiatric disorders, mainly stress-related disorders like anxiety and depression. Conversely, pain appears to be altered in many psychiatric disorders, such as depression, anxiety and schizophrenia. Thus, pain and psychiatric disorders have been linked in epidemiological and clinical terms, although the neurobiological mechanisms involved in this pathological bidirectional relationship remain unclear. Here we review the evidence obtained from animal models about the co-morbidity of pain and psychiatric disorders, placing special emphasis on the different dimensions of pain.
Collapse
Affiliation(s)
- Lidia Bravo
- Neuropsychopharmacology and Psychobiology Research Group, Department of Neuroscience, University of Cádiz, 11003 Cádiz, Spain; Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Avda. Ana de Viya 21, 11009 Cádiz, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Meritxell Llorca-Torralba
- Neuropsychopharmacology and Psychobiology Research Group, Department of Neuroscience, University of Cádiz, 11003 Cádiz, Spain; Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Avda. Ana de Viya 21, 11009 Cádiz, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Irene Suárez-Pereira
- Neuropsychopharmacology and Psychobiology Research Group, Department of Neuroscience, University of Cádiz, 11003 Cádiz, Spain; Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Avda. Ana de Viya 21, 11009 Cádiz, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Esther Berrocoso
- Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Avda. Ana de Viya 21, 11009 Cádiz, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain; Neuropsychopharmacology and Psychobiology Research Group, Department of Psychology, University of Cádiz, 11510 Puerto Real, Cádiz, Spain.
| |
Collapse
|
45
|
Interneuron NMDA Receptor Ablation Induces Hippocampus-Prefrontal Cortex Functional Hypoconnectivity after Adolescence in a Mouse Model of Schizophrenia. J Neurosci 2020; 40:3304-3317. [PMID: 32205341 DOI: 10.1523/jneurosci.1897-19.2020] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 01/07/2020] [Accepted: 01/18/2020] [Indexed: 12/13/2022] Open
Abstract
Although the etiology of schizophrenia is still unknown, it is accepted to be a neurodevelopmental disorder that results from the interaction of genetic vulnerabilities and environmental insults. Although schizophrenia's pathophysiology is still unclear, postmortem studies point toward a dysfunction of cortical interneurons as a central element. It has been suggested that alterations in parvalbumin-positive interneurons in schizophrenia are the consequence of a deficient signaling through NMDARs. Animal studies demonstrated that early postnatal ablation of the NMDAR in corticolimbic interneurons induces neurobiochemical, physiological, behavioral, and epidemiological phenotypes related to schizophrenia. Notably, the behavioral abnormalities emerge only after animals complete their maturation during adolescence and are absent if the NMDAR is deleted during adulthood. This suggests that interneuron dysfunction must interact with development to impact on behavior. Here, we assess in vivo how an early NMDAR ablation in corticolimbic interneurons impacts on mPFC and ventral hippocampus functional connectivity before and after adolescence. In juvenile male mice, NMDAR ablation results in several pathophysiological traits, including increased cortical activity and decreased entrainment to local gamma and distal hippocampal theta rhythms. In addition, adult male KO mice showed reduced ventral hippocampus-mPFC-evoked potentials and an augmented low-frequency stimulation LTD of the pathway, suggesting that there is a functional disconnection between both structures in adult KO mice. Our results demonstrate that early genetic abnormalities in interneurons can interact with postnatal development during adolescence, triggering pathophysiological mechanisms related to schizophrenia that exceed those caused by NMDAR interneuron hypofunction alone.SIGNIFICANCE STATEMENT NMDAR hypofunction in cortical interneurons has been linked to schizophrenia pathophysiology. How a dysfunction of GABAergic cortical interneurons interacts with maturation during adolescence has not been clarified yet. Here, we demonstrate in vivo that early postnatal ablation of the NMDAR in corticolimbic interneurons results in an overactive but desynchronized PFC before adolescence. Final postnatal maturation during this stage outspreads the impact of the genetic manipulation toward a functional disconnection of the ventral hippocampal-prefrontal pathway, probably as a consequence of an exacerbated propensity toward hippocampal-evoked depotentiation plasticity. Our results demonstrate a complex interaction between genetic and developmental factors affecting cortical interneurons and PFC function.
Collapse
|
46
|
Chambers RA, Sentir AM. Integrated Effects of Neonatal Ventral Hippocampal Lesions and Impoverished Social-Environmental Rearing on Endophenotypes of Mental Illness and Addiction Vulnerability. Dev Neurosci 2020; 41:263-273. [PMID: 32160629 PMCID: PMC8454183 DOI: 10.1159/000506227] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 01/23/2020] [Indexed: 11/19/2022] Open
Abstract
A wide range of mental illnesses show high rates of addiction comorbidities regardless of their genetic, neurodevelopmental, and/or adverse-environmental etiologies. Understanding how the spectrum of mental illnesses produce addiction vulnerability will be key to discovering more effective preventions and integrated treatments for adults with addiction and dual diagnosis comorbidities. A population of 131 rats containing a spectrum of etiological mental illness models and degrees of severity was experimentally generated by crossing neonatal ventral hippocampal lesions (NVHL; n = 68) or controls (SHAM-operated; n = 63) with adolescent rearing in environmentally/socially enriched (ENR; n = 66) or impoverished (IMP; n = 65) conditions. This population was divided into 2 experiments: first, examining NVHL and IMP effects on novelty and mild stress-induced locomotion across 3 adolescent ages; second, looking at initial cocaine reactivity and long-term cocaine behavioral sensitization in adulthood. NVHL and IMP-environmental conditions independently produced remarkably similar and robustly significant abnormalities of hyperreactivity to novelty, mild stress, and long-term cocaine sensitization. The combined NVHL-IMP groups showed the most severe phenotypes across the board, so that the mental illness and addiction vulnerability phenotypes increased together in severity in a consistent stepwise progression from the healthiest rats to those with the greatest loading of etiological models. These findings add weight to our understanding of mental illness and addiction vulnerability as brain disorders that are biologically and developmentally unified in ways that transcend etiological causes, and yet co-intensify with increased loading of etiological conditions. Combining neurodevelopmental and adverse-environmental models of mental illness may provide an approach to identifying and therapeutically targeting cortical-striatal-limbic network mechanisms that generate addiction and dual diagnosis diseases.
Collapse
Affiliation(s)
- Robert Andrew Chambers
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, Indiana, USA,
- Laboratory for Translational Neuroscience of Dual Diagnosis & Development, IU Neuroscience Research Center, Indianapolis, Indiana, USA,
| | - Alena M Sentir
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Laboratory for Translational Neuroscience of Dual Diagnosis & Development, IU Neuroscience Research Center, Indianapolis, Indiana, USA
| |
Collapse
|
47
|
Weeks JJ, Rupprecht LE, Grace AA, Donny EC, Sved AF. Nicotine Self-administration Is Not Increased in the Methylazoxymethanol Acetate Rodent Model of Schizophrenia. Nicotine Tob Res 2020; 22:204-212. [PMID: 30899959 PMCID: PMC7297085 DOI: 10.1093/ntr/ntz048] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 03/20/2019] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Patients with schizophrenia (SCZ) smoke at a rate of 4-5 times higher than the general population, contributing to negative health consequences in this group. One possible explanation for this increased smoking is that individuals with SCZ find nicotine (NIC) more reinforcing. However, data supporting this possibility are limited. METHODS The present experiments examined self-administration of NIC, alone or in combination with other reinforcers, across a range of doses in the methylazoxymethanol acetate (MAM) rodent model of SCZ. RESULTS MAM and control animals did not differ in NIC self-administration across a range of doses and schedules of reinforcement, in both standard 1-hour self-administration sessions and 23-hour extended access sessions. However, MAM animals responded less for sucrose or reinforcing visual stimuli alone or when paired with NIC. CONCLUSIONS To the extent that MAM-treated rats are a valid model of SCZ, these results suggest that increased NIC reinforcement does not account for increased smoking in SCZ patients. IMPLICATIONS This study is the first to utilize nicotine self-administration, the gold standard for studying nicotine reinforcement, in the methylazoxymethanol acetate model of schizophrenia, which is arguably the most comprehensive animal model of the disease currently available. Our assessment found no evidence of increased nicotine reinforcement in methylazoxymethanol acetate animals, suggesting that increased reinforcement may not perpetuate increased smoking in schizophrenia patients.
Collapse
Affiliation(s)
- Jillian J Weeks
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA
| | | | - Anthony A Grace
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA
- Department of Neuroscience, School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA
| | - Eric C Donny
- Department of Physiology and Pharmacology, School of Medicine, Wake Forest University, Winston-Salem, NC
| | - Alan F Sved
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA
- Department of Neuroscience, School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
48
|
Gerardo CM, Manuel MMV. The thalamic reticular nucleus: A common nucleus of neuropsychiatric diseases and deep brain stimulation. J Clin Neurosci 2020; 73:1-7. [PMID: 32001110 DOI: 10.1016/j.jocn.2020.01.061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 01/12/2020] [Indexed: 11/18/2022]
Abstract
This review focuses on the studies that have been reviewed to determine the influence of the thalamic reticular nucleus on neuropsychiatric diseases and deep brain stimulation. The literature reviewed to date describes how alterations in the thalamic reticular nucleus affect several functions that regulated brain rhythms and provokes symptoms of many disorders. The observations as the basis for the renewed interest in the thalamic reticular nucleus in experimental models and testing its effectiveness in patients with resistant neuropsychiatric disorders. The preclinical studies showed that deep brain stimulation in the thalamic reticular nucleus could have beneficial effects on EEG activity, including synchronization and desynchronization activity of the brain, as well as promoting an alleviate to neuropsychiatric diseases. These observations open up the possibility of studying the role played by neurotransmitters in the pathologic process and the deep brain stimulation in the thalamic reticular nucleus in experimental animal models and offer evidence of its possible action in the human brain.
Collapse
Affiliation(s)
- Contreras-Murillo Gerardo
- Laboratorio del Control y la Regulación, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México, Mexico
| | - Magdaleno-Madrigal Víctor Manuel
- Laboratorio del Control y la Regulación, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México, Mexico.
| |
Collapse
|
49
|
Koszła O, Targowska-Duda KM, Kędzierska E, Kaczor AA. In Vitro and In Vivo Models for the Investigation of Potential Drugs Against Schizophrenia. Biomolecules 2020; 10:biom10010160. [PMID: 31963851 PMCID: PMC7022578 DOI: 10.3390/biom10010160] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/12/2020] [Accepted: 01/13/2020] [Indexed: 02/06/2023] Open
Abstract
Schizophrenia (SZ) is a complex psychiatric disorder characterized by positive, negative, and cognitive symptoms, and is not satisfactorily treated by current antipsychotics. Progress in understanding the basic pathomechanism of the disease has been hampered by the lack of appropriate models. In order to develop modern drugs against SZ, efficient methods to study them in in vitro and in vivo models of this disease are required. In this review a short presentation of current hypotheses and concepts of SZ is followed by a description of current progress in the field of SZ experimental models. A critical discussion of advantages and limitations of in vitro models and pharmacological, genetic, and neurodevelopmental in vivo models for positive, negative, and cognitive symptoms of the disease is provided. In particular, this review concerns the important issue of how cellular and animal systems can help to meet the challenges of modeling the disease, which fully manifests only in humans, as experimental studies of SZ in humans are limited. Next, it is emphasized that novel clinical candidates should be evaluated in animal models for treatment-resistant SZ. In conclusion, the plurality of available in vitro and in vivo models is a consequence of the complex nature of SZ, and there are extensive possibilities for their integration. Future development of more efficient antipsychotics reflecting the pleiotropy of symptoms in SZ requires the incorporation of various models into one uniting model of the multifactorial disorder and use of this model for the evaluation of new drugs.
Collapse
Affiliation(s)
- Oliwia Koszła
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances, Faculty of Pharmacy, Medical University of Lublin, 4A Chodźki St., PL-20093 Lublin, Poland;
| | - Katarzyna M. Targowska-Duda
- Department of Biopharmacy, Faculty of Pharmacy, Medical University of Lublin, 4A Chodźki St., PL-20093 Lublin, Poland
| | - Ewa Kędzierska
- Department of Pharmacology and Pharmacodynamics, Faculty of Pharmacy, Medical University of Lublin, 4A Chodźki St., PL-20093 Lublin, Poland;
| | - Agnieszka A. Kaczor
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances, Faculty of Pharmacy, Medical University of Lublin, 4A Chodźki St., PL-20093 Lublin, Poland;
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
- Correspondence:
| |
Collapse
|
50
|
Sentir AM, Bell RL, Engleman EA, Chambers RA. Polysubstance addiction vulnerability in mental illness: Concurrent alcohol and nicotine self-administration in the neurodevelopmental hippocampal lesion rat model of schizophrenia. Addict Biol 2020; 25:e12704. [PMID: 30592364 DOI: 10.1111/adb.12704] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 10/25/2018] [Accepted: 11/10/2018] [Indexed: 01/19/2023]
Abstract
Multiple addictions frequently occur in patients with mental illness. However, basic research on the brain-based linkages between these comorbidities is extremely limited. Toward characterizing the first animal modeling of polysubstance use and addiction vulnerability in schizophrenia, adolescent rats with neonatal ventral hippocampal lesions (NVHLs) and controls had 19 weekdays of 1 hour/day free access to alcohol/sucrose solutions (fading from 10% sucrose to 10% alcohol/2% sucrose on day 10) during postnatal days (PD 35-60). Starting in adulthood (PD 63), rats acquired lever pressing for concurrent oral alcohol (10% with 2% sucrose) and iv nicotine (0.015 mg/kg/injection) across 15 sessions. Subsequently, 10 operant extinction sessions and 3 reinstatement sessions examined drug seeking upon withholding of nicotine, then both nicotine and alcohol, then reintroduction. Adolescent alcohol consumption did not differ between NVHLs and controls. However, in adulthood, NVHLs showed increased lever pressing at alcohol and nicotine levers that progressed more strongly at the nicotine lever, even as most pressing by both groups was at the alcohol lever. In extinction, both groups showed expected declines in effort as drugs were withheld, but NVHLs persisted with greater pressing at both alcohol and nicotine levers. In reinstatement, alcohol reaccess increased pressing, with NVHLs showing greater nicotine lever activity overall. Developmental temporal-limbic abnormalities that produce mental illness can thus generate adult polydrug addiction vulnerability as a mechanism independent from putative cross-sensitization effects between addictive drugs. Further preclinical modeling of third-order (and higher) addiction-mental illness comorbidities may advance our understanding and treatment of these complex, yet common brain illnesses.
Collapse
Affiliation(s)
- Alena M. Sentir
- Department of PsychiatryIndiana University School of Medicine Indianapolis IN
- Laboratory for Translational Neuroscience of Dual Diagnosis & DevelopmentIU Neuroscience Research Center Indianapolis IN
| | - Richard L. Bell
- Department of PsychiatryIndiana University School of Medicine Indianapolis IN
| | - Eric A. Engleman
- Department of PsychiatryIndiana University School of Medicine Indianapolis IN
| | - R. Andrew Chambers
- Department of PsychiatryIndiana University School of Medicine Indianapolis IN
- Laboratory for Translational Neuroscience of Dual Diagnosis & DevelopmentIU Neuroscience Research Center Indianapolis IN
| |
Collapse
|