1
|
Kryst J, Chocyk A, Solarz-Andrzejewska A, Majcher-Maślanka I. Juvenile fluoxetine treatment affects the maturation of the medial prefrontal cortex and behavior of adolescent female rats. Pharmacol Rep 2025; 77:670-688. [PMID: 40063219 DOI: 10.1007/s43440-025-00712-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/24/2025] [Accepted: 03/05/2025] [Indexed: 05/13/2025]
Abstract
BACKGROUND Serotonin is strongly involved in the regulation of brain development, including the proper formation of neuronal circuits and synaptic plasticity. One of the factors that can affect brain serotonin levels is exposure to fluoxetine (FLX), a selective serotonin reuptake inhibitor, the first-line pharmacological treatment for depression and anxiety in the pediatric population. The safety of early-life FLX treatment is still questionable. Women are more prone to anxiety and depression from a young age. We hypothesized that juvenile FLX treatment influences the brain maturation and behavior of adolescent females. METHODS On postnatal days 20 to 28, juvenile female rats were injected once daily with FLX. Five days later, anxiety- and fear-related behaviors and amphetamine-induced locomotor activity were assessed. On postnatal day 40, the numbers of neurons and glial cells in the medial prefrontal cortex (mPFC) and hippocampus were estimated via stereological methods. Additionally, the mRNA expression of cell survival/apoptosis and synaptic plasticity markers was evaluated via RT‒qPCR. RESULTS FLX-treated females showed decreased anxiety level, freezing behavior during fear conditioning and amphetamine-induced locomotor activity when compared to control females. Simultaneously, FLX-injected females presented greater regional volume and numbers of neurons and astrocytes in specific subregions of the mPFC when compared to the control group. Additionally, FLX-treated females showed increased expression of genes regulating cell survival and reduced mRNA levels of AMPA glutamate receptors in the mPFC. CONCLUSIONS Juvenile FLX affects the maturation of the mPFC and attenuates anxiety-like behavior, fear memory and the locomotor response to amphetamine in adolescent females.
Collapse
Affiliation(s)
- Joanna Kryst
- Department of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, Kraków, 31-343, Poland
- Department of Chemistry and Biochemistry, Institute for Basics Sciences, Faculty of Physiotherapy, University of Physical Education, Jana Pawła II 78, Kraków, 31-571, Poland
| | - Agnieszka Chocyk
- Department of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, Kraków, 31-343, Poland.
| | - Anna Solarz-Andrzejewska
- Department of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, Kraków, 31-343, Poland
- Laboratory of Molecular Biology, Institute of Physiotherapy and Health Sciences, Jerzy Kukuczka Academy of Physical Education, Mikołowska 72a, Katowice, 40-065, Poland
| | - Iwona Majcher-Maślanka
- Department of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, Kraków, 31-343, Poland
| |
Collapse
|
2
|
Moreau AL, Hansen I, Bogdan R. A systematic review of structural neuroimaging markers of psychotherapeutic and pharmacological treatment for obsessive-compulsive disorder. Front Psychiatry 2025; 15:1432253. [PMID: 40018086 PMCID: PMC11865061 DOI: 10.3389/fpsyt.2024.1432253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 12/19/2024] [Indexed: 03/01/2025] Open
Abstract
Identifying individual difference factors associated with treatment response and putative mechanisms of therapeutic change may improve treatment for Obsessive Compulsive Disorder (OCD). Our systematic review of structural neuroimaging markers (i.e., morphometry, structural connectivity) of psychotherapy and medication treatment response for OCD identified 26 eligible publications from 20 studies (average study total n=54 ± 41.6 [range: 11-175]; OCD group n=29 ± 19) in child, adolescent, and adult samples evaluating baseline brain structure correlates of treatment response as well as treatment-related changes in brain structure. Findings were inconsistent across studies; significant associations within the anterior cingulate cortex (3/5 regional, 2/8 whole brain studies) and orbitofrontal cortex (5/10 regional, 2/7 whole brain studies) were most common, but laterality and directionality were not always consistent. Structural neuroimaging markers of treatment response do not currently hold clinical utility. Given increasing evidence that associations between complex behavior and brain structure are characterized by small, but potentially meaningful, effects, much larger samples are likely needed. Multivariate approaches (e.g., machine learning) may also improve the clinical predictive utility of neuroimaging data.
Collapse
Affiliation(s)
- Allison L. Moreau
- Department of Psychological and Brain Sciences, Washington University in St. Louis, Saint Louis, MO, United States
| | | | - Ryan Bogdan
- Department of Psychological and Brain Sciences, Washington University in St. Louis, Saint Louis, MO, United States
| |
Collapse
|
3
|
Komaki S, Amiri P, Safari S, Abbasi E, Ramezani-Aliakbari F, Golipoor M, Kourosh-Arami M, Rashno M, Komaki A. Investigation of protective effects of olanzapine on impaired learning and memory using behavioral tests in a rat model of Alzheimer's disease. Front Aging Neurosci 2025; 17:1376074. [PMID: 40018516 PMCID: PMC11865076 DOI: 10.3389/fnagi.2025.1376074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 01/22/2025] [Indexed: 03/01/2025] Open
Abstract
Introduction Evidence suggests that oxidative stress plays a critical role in the pathogenesis and progression of Alzheimer's disease (AD). Consequently, antioxidants may mitigate neurotoxicity induced by beta-amyloid (Aβ) and potentially reduce cell death. Previous research has demonstrated that olanzapine (OLZ) possesses antioxidant and neuroprotective properties. In this study, we investigated the protective and therapeutic effects of OLZ on an animal model of AD induced by Aβ using behavioral assessments. Methods Rats were randomly assigned to one of five groups (n = 10 rats per group): a control group, a sham group that received an intracerebrovascular (ICV) injection of phosphate-buffered saline (the solvent for Aβ), an AD group that received an ICV injection of Aβ, an OLZ group that received OLZ via gavage for two months, and an AD + OLZ group that received OLZ for one month before and one month after AD induction. Results We used the Elevated Plus Maze (EPM), Novel Object Recognition Test (NORT), Barnes Maze (BM), Passive Avoidance Test (PAT), and Morris Water Maze (MWM) to assess behavioral performance in the experimental rats. Aβ administration impaired cognition and increased anxiety-like behavior. Treatment with OLZ improved cognitive decline and reduced anxiety-like behavior in Aβ-infused rats. Conclusion Our findings suggest that OLZ can restore cognitive performance and alleviate anxiety-like behavior following Aβ injection. Thus, OLZ may have both preventive and therapeutic potential for AD and could be considered a viable pharmacological option.
Collapse
Affiliation(s)
- Somayeh Komaki
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Parsa Amiri
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Samaneh Safari
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ebrahim Abbasi
- Department of Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | | | - Mandana Golipoor
- Neuroscience Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Masoumeh Kourosh-Arami
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | | | - Alireza Komaki
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
4
|
Alonso M, Petit AC, Lledo PM. The impact of adult neurogenesis on affective functions: of mice and men. Mol Psychiatry 2024; 29:2527-2542. [PMID: 38499657 DOI: 10.1038/s41380-024-02504-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 02/22/2024] [Accepted: 02/27/2024] [Indexed: 03/20/2024]
Abstract
In most mammals, new neurons are not only produced during embryogenesis but also after birth. Soon after adult neurogenesis was discovered, the influence of recruiting new neurons on cognitive functions, especially on memory, was documented. Likewise, the late process of neuronal production also contributes to affective functions, but this outcome was recognized with more difficulty. This review covers hypes and hopes of discovering the influence of newly-generated neurons on brain circuits devoted to affective functions. If the possibility of integrating new neurons into the adult brain is a commonly accepted faculty in the realm of mammals, the reluctance is strong when it comes to translating this concept to humans. Compiling data suggest now that new neurons are derived not only from stem cells, but also from a population of neuroblasts displaying a protracted maturation and ready to be engaged in adult brain circuits, under specific signals. Here, we discuss the significance of recruiting new neurons in the adult brain circuits, specifically in the context of affective outcomes. We also discuss the fact that adult neurogenesis could be the ultimate cellular process that integrates elements from both the internal and external environment to adjust brain functions. While we must be critical and beware of the unreal promises that Science could generate sometimes, it is important to continue exploring the potential of neural recruitment in adult primates. Reporting adult neurogenesis in humankind contributes to a new vision of humans as mammals whose brain continues to develop throughout life. This peculiar faculty could one day become the target of treatment for mental health, cognitive disorders, and elderly-associated diseases. The vision of an adult brain which never stops integrating new neurons is a real game changer for designing new therapeutic interventions to treat mental disorders associated with substantial morbidity, mortality, and social costs.
Collapse
Affiliation(s)
- Mariana Alonso
- Institut Pasteur, Université Paris Cité, CNRS UMR 3571, Perception and Action Unit, F-75015, Paris, France
| | - Anne-Cécile Petit
- Institut Pasteur, Université Paris Cité, CNRS UMR 3571, Perception and Action Unit, F-75015, Paris, France
- Pôle Hospitalo-Universitaire Psychiatrie Paris 15, GHU Paris Psychiatry and Neurosciences, Hôpital Sainte-Anne, Paris, France
| | - Pierre-Marie Lledo
- Institut Pasteur, Université Paris Cité, CNRS UMR 3571, Perception and Action Unit, F-75015, Paris, France.
| |
Collapse
|
5
|
Martins-Macedo J, Araújo B, Anjo SI, Silveira-Rosa T, Patrício P, Alves ND, Silva JM, Teixeira FG, Manadas B, Rodrigues AJ, Lepore AC, Salgado AJ, Gomes ED, Pinto L. Glial-restricted precursors stimulate endogenous cytogenesis and effectively recover emotional deficits in a model of cytogenesis ablation. Mol Psychiatry 2024; 29:2185-2198. [PMID: 38454085 PMCID: PMC11632613 DOI: 10.1038/s41380-024-02490-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 02/08/2024] [Accepted: 02/15/2024] [Indexed: 03/09/2024]
Abstract
Adult cytogenesis, the continuous generation of newly-born neurons (neurogenesis) and glial cells (gliogenesis) throughout life, is highly impaired in several neuropsychiatric disorders, such as Major Depressive Disorder (MDD), impacting negatively on cognitive and emotional domains. Despite playing a critical role in brain homeostasis, the importance of gliogenesis has been overlooked, both in healthy and diseased states. To examine the role of newly formed glia, we transplanted Glial Restricted Precursors (GRPs) into the adult hippocampal dentate gyrus (DG), or injected their secreted factors (secretome), into a previously validated transgenic GFAP-tk rat line, in which cytogenesis is transiently compromised. We explored the long-term effects of both treatments on physiological and behavioral outcomes. Grafted GRPs reversed anxiety-like deficits and demonstrated an antidepressant-like effect, while the secretome promoted recovery of only anxiety-like behavior. Furthermore, GRPs elicited a recovery of neurogenic and gliogenic levels in the ventral DG, highlighting the unique involvement of these cells in the regulation of brain cytogenesis. Both GRPs and their secretome induced significant alterations in the DG proteome, directly influencing proteins and pathways related to cytogenesis, regulation of neural plasticity and neuronal development. With this work, we demonstrate a valuable and specific contribution of glial progenitors to normalizing gliogenic levels, rescuing neurogenesis and, importantly, promoting recovery of emotional deficits characteristic of disorders such as MDD.
Collapse
Affiliation(s)
- Joana Martins-Macedo
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Center for Translational Health and Medical Biotechnology Research (TBIO), School of Health (ESS), Polytechnic of Porto, Porto, Portugal
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal
| | - Bruna Araújo
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Center for Translational Health and Medical Biotechnology Research (TBIO), School of Health (ESS), Polytechnic of Porto, Porto, Portugal
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal
| | - Sandra I Anjo
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra (IIIUC), Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - Tiago Silveira-Rosa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Patrícia Patrício
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Nuno Dinis Alves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Joana M Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Fábio G Teixeira
- Center for Translational Health and Medical Biotechnology Research (TBIO), School of Health (ESS), Polytechnic of Porto, Porto, Portugal
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal
| | - Bruno Manadas
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra (IIIUC), Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - Ana J Rodrigues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Angelo C Lepore
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Eduardo D Gomes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Center for Translational Health and Medical Biotechnology Research (TBIO), School of Health (ESS), Polytechnic of Porto, Porto, Portugal
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal
| | - Luísa Pinto
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
6
|
Nazzi S, Picchi M, Migliarini S, Maddaloni G, Barsotti N, Pasqualetti M. Reversible Morphological Remodeling of Prefrontal and Hippocampal Serotonergic Fibers by Fluoxetine. ACS Chem Neurosci 2024; 15:1702-1711. [PMID: 38433715 DOI: 10.1021/acschemneuro.3c00837] [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] [Indexed: 03/05/2024] Open
Abstract
Serotonin-releasing fibers depart from the raphe nuclei to profusely innervate the entire central nervous system, displaying in some brain regions high structural plasticity in response to genetically induced abrogation of serotonin synthesis. Chronic fluoxetine treatment used as a tool to model peri-physiological, clinically relevant serotonin elevation is also able to cause structural rearrangements of the serotonergic fibers innervating the hippocampus. Whether this effect is limited to hippocampal-innervating fibers or extends to other populations of axons is not known. Here, we used confocal imaging and three-dimensional (3-D) modeling analysis to expand our morphological investigation of fluoxetine-mediated effects on serotonergic circuitry. We found that chronic treatment with a behaviorally active dose of fluoxetine affects the morphology and reduces the density of serotonergic axons innervating the medial prefrontal cortex, a brain region strongly implicated in the regulation of depressive- and anxiety-like behavior. Axons innervating the somatosensory cortex were unaffected, suggesting differential susceptibility to serotonin changes across cortical areas. Importantly, a 1-month washout period was sufficient to reverse morphological changes in both the medial prefrontal cortex and in the previously characterized hippocampus, as well as to normalize behavior, highlighting an intriguing relationship between axon density and an antidepressant-like effect. Overall, these results further demonstrate the bidirectional plasticity of defined serotonergic axons and provide additional insights into fluoxetine effects on the serotonergic system.
Collapse
Affiliation(s)
- Serena Nazzi
- Department of Biology, Unit of Cell and Developmental Biology, University of Pisa, Pisa 56127, Italy
| | - Marta Picchi
- Department of Biology, Unit of Cell and Developmental Biology, University of Pisa, Pisa 56127, Italy
| | - Sara Migliarini
- Department of Biology, Unit of Cell and Developmental Biology, University of Pisa, Pisa 56127, Italy
| | - Giacomo Maddaloni
- Department of Biology, Unit of Cell and Developmental Biology, University of Pisa, Pisa 56127, Italy
| | - Noemi Barsotti
- Department of Biology, Unit of Cell and Developmental Biology, University of Pisa, Pisa 56127, Italy
- Centro per l'Integrazione della Strumentazione Scientifica dell'Università di Pisa (CISUP), Pisa 56126, Italy
| | - Massimo Pasqualetti
- Department of Biology, Unit of Cell and Developmental Biology, University of Pisa, Pisa 56127, Italy
- Centro per l'Integrazione della Strumentazione Scientifica dell'Università di Pisa (CISUP), Pisa 56126, Italy
- Center for Neuroscience and Cognitive Systems@UniTn, Istituto Italiano di Tecnologia, Rovereto 38068, Italy
| |
Collapse
|
7
|
Hirose Y, Oda Y, Yoshino K, Yano F, Kimura M, Kimura H, Iyo M, Shirayama Y. Reduction of claudin-5 and aquaporin-4 in the rat hippocampal CA-1 and CA-3 regions of a learned helplessness model of depression. Pharmacol Biochem Behav 2024; 234:173676. [PMID: 37992974 DOI: 10.1016/j.pbb.2023.173676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/09/2023] [Accepted: 11/09/2023] [Indexed: 11/24/2023]
Abstract
BACKGROUND Although findings from both animal and clinical research indicate that the blood-brain barrier (BBB) contributes to the pathogenesis of various psychiatric disorders (including depression), the underlying mechanisms are unknown. We investigated the levels of the tight-junction proteins claudin-5 and aquaporin-4 (AQP-4) in astrocytes of learned helplessness (LH) rats (an animal model of depression) and non-LH rats (a model of resilience). METHODS We administered inescapable mild electric shock to rats and then identified the LH and non-LH rats by a post-shock test. The expressions of claudin-5 and AQP-4 in several brain regions of the LH and non-LH rats were then evaluated by a western blot analysis. RESULTS The levels of both claudin-5 and AQP-4 in the CA-1 and CA-3 hippocampal areas of the LH group were significantly lower than those of the control group, whereas those of the non-LH rats were not significantly different from those of the control and LH rats. CONCLUSIONS These results suggest that LH rats but not non-LH rats experienced down-regulations of claudin-5 and AQP-4 in the CA-1 and CA-3. It is possible that a region-specific modulation of claudin-5 and AQP-4 is involved in the mechanisms of vulnerability but not resilience in depression.
Collapse
Affiliation(s)
- Yuki Hirose
- Department of Psychiatry, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuou-ku, Chiba, Chiba 260-8670, Japan
| | - Yasunori Oda
- Department of Psychiatry, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuou-ku, Chiba, Chiba 260-8670, Japan.
| | - Kouhei Yoshino
- Department of Psychiatry, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuou-ku, Chiba, Chiba 260-8670, Japan
| | - Fumiaki Yano
- Department of Psychiatry, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuou-ku, Chiba, Chiba 260-8670, Japan
| | - Makoto Kimura
- Department of Psychiatry, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuou-ku, Chiba, Chiba 260-8670, Japan
| | - Hiroshi Kimura
- Department of Psychiatry, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuou-ku, Chiba, Chiba 260-8670, Japan; Department of Psychiatry, School of Medicine, International University of Health and Welfare, 4-3 Kozunomori, Narita, Chiba 286-8686, Japan
| | - Masaomi Iyo
- Department of Psychiatry, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuou-ku, Chiba, Chiba 260-8670, Japan
| | - Yukihiko Shirayama
- Department of Psychiatry, Teikyo University Chiba Medical Center, 3426-3 Anesaki, Ichihara, Chiba 290-0111, Japan
| |
Collapse
|
8
|
Bruin WB, Abe Y, Alonso P, Anticevic A, Backhausen LL, Balachander S, Bargallo N, Batistuzzo MC, Benedetti F, Bertolin Triquell S, Brem S, Calesella F, Couto B, Denys DAJP, Echevarria MAN, Eng GK, Ferreira S, Feusner JD, Grazioplene RG, Gruner P, Guo JY, Hagen K, Hansen B, Hirano Y, Hoexter MQ, Jahanshad N, Jaspers-Fayer F, Kasprzak S, Kim M, Koch K, Bin Kwak Y, Kwon JS, Lazaro L, Li CSR, Lochner C, Marsh R, Martínez-Zalacaín I, Menchon JM, Moreira PS, Morgado P, Nakagawa A, Nakao T, Narayanaswamy JC, Nurmi EL, Zorrilla JCP, Piacentini J, Picó-Pérez M, Piras F, Piras F, Pittenger C, Reddy JYC, Rodriguez-Manrique D, Sakai Y, Shimizu E, Shivakumar V, Simpson BH, Soriano-Mas C, Sousa N, Spalletta G, Stern ER, Evelyn Stewart S, Szeszko PR, Tang J, Thomopoulos SI, Thorsen AL, Yoshida T, Tomiyama H, Vai B, Veer IM, Venkatasubramanian G, Vetter NC, Vriend C, Walitza S, Waller L, Wang Z, Watanabe A, Wolff N, Yun JY, Zhao Q, van Leeuwen WA, van Marle HJF, van de Mortel LA, van der Straten A, van der Werf YD, Thompson PM, Stein DJ, van den Heuvel OA, van Wingen GA. The functional connectome in obsessive-compulsive disorder: resting-state mega-analysis and machine learning classification for the ENIGMA-OCD consortium. Mol Psychiatry 2023; 28:4307-4319. [PMID: 37131072 PMCID: PMC10827654 DOI: 10.1038/s41380-023-02077-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 04/11/2023] [Accepted: 04/14/2023] [Indexed: 05/04/2023]
Abstract
Current knowledge about functional connectivity in obsessive-compulsive disorder (OCD) is based on small-scale studies, limiting the generalizability of results. Moreover, the majority of studies have focused only on predefined regions or functional networks rather than connectivity throughout the entire brain. Here, we investigated differences in resting-state functional connectivity between OCD patients and healthy controls (HC) using mega-analysis of data from 1024 OCD patients and 1028 HC from 28 independent samples of the ENIGMA-OCD consortium. We assessed group differences in whole-brain functional connectivity at both the regional and network level, and investigated whether functional connectivity could serve as biomarker to identify patient status at the individual level using machine learning analysis. The mega-analyses revealed widespread abnormalities in functional connectivity in OCD, with global hypo-connectivity (Cohen's d: -0.27 to -0.13) and few hyper-connections, mainly with the thalamus (Cohen's d: 0.19 to 0.22). Most hypo-connections were located within the sensorimotor network and no fronto-striatal abnormalities were found. Overall, classification performances were poor, with area-under-the-receiver-operating-characteristic curve (AUC) scores ranging between 0.567 and 0.673, with better classification for medicated (AUC = 0.702) than unmedicated (AUC = 0.608) patients versus healthy controls. These findings provide partial support for existing pathophysiological models of OCD and highlight the important role of the sensorimotor network in OCD. However, resting-state connectivity does not so far provide an accurate biomarker for identifying patients at the individual level.
Collapse
Grants
- R01 AG058854 NIA NIH HHS
- R01 MH126213 NIMH NIH HHS
- R21 MH101441 NIMH NIH HHS
- R01 MH121520 NIMH NIH HHS
- R21 MH093889 NIMH NIH HHS
- R01 MH116147 NIMH NIH HHS
- R01 MH111794 NIMH NIH HHS
- R01 MH085900 NIMH NIH HHS
- P41 EB015922 NIBIB NIH HHS
- IA/CPHE/18/1/503956 DBT-Wellcome Trust India Alliance
- UL1 TR001863 NCATS NIH HHS
- R01 MH081864 NIMH NIH HHS
- R01 MH104648 NIMH NIH HHS
- U54 EB020403 NIBIB NIH HHS
- R01 MH117601 NIMH NIH HHS
- R01 MH116038 NIMH NIH HHS
- R01 MH126981 NIMH NIH HHS
- R01 NS107513 NINDS NIH HHS
- RF1 MH123163 NIMH NIH HHS
- R33 MH107589 NIMH NIH HHS
- K24 MH121571 NIMH NIH HHS
- R01 MH121246 NIMH NIH HHS
- Wellcome Trust
- K23 MH115206 NIMH NIH HHS
- R01 AG059874 NIA NIH HHS
- Funding from Japan Society for the Promotion of Science (KAKENHI Grant No. 18K15523)
- Carlos III Health Institute PI18/00856
- NIMH: 5R01MH116038
- Sara Bertolin was supported by Instituto de Salud Carlos III through the grant CM21/00278 (Co-funded by European Social Fund. ESF investing in your future).
- Hartmann Müller Foundation (no. 1460, principal investigator: S.Brem)
- NIHM: R01MH085900, R01MH121520
- NIH: K23 MH115206 & IOCDF Annual Research Award
- AMED Brain/MINDS Beyond program Grant No. JP22dm0307002, JSPS KAKENHI Grants No. 22H01090, 21K03084, 19K03309, 16K04344
- NIH: R01MH117601, R01AG059874, P41EB015922, R01MH126213, R01MH121246
- Michael Smith Health Research BC
- the Deutsche Forschungsgemeinschaf (KO 3744/11-1)
- This work was supported by the Medical Research Council of South Africa (SAMRC), and the National Research Foundation of South Africa (Christine Lochner), and we acknowledge the contribution of our research assistants.
- NIMH: R21MH093889, R21MH101441 and R01MH104648
- IM-Z was supported by a PFIS grant (FI17/00294) from the Carlos III Health Institute
- This work was supported by National funds, through the Foundation for Science and Technology (project UIDB/50026/2020 and UIDP/50026/2020); by the Norte Portugal Regional Operational Programme (NORTE 2020) under the PORTUGAL 2020 Partnership Agreement, through the European Regional Development Fund (ERDF) (projects NORTE-01-0145-FEDER-000013 and NORTE-01-0145-FEDER-000023), and by the FLAD Science Award Mental Health 2021.
- JSPS KAKENHI (C)21K07547, 22K07598 and 22K15766
- Government of India grants from Department of Science and Technology (DST INSPIRE faculty grant -IFA12-LSBM-26) & Department of Biotechnology (BT/06/IYBA/2012)
- NIMH: R01MH081864
- MPP was supported by the Spanish Ministry of Universities, with funds from the European Union - NextGenerationEU (MAZ/2021/11).
- Italian Ministry of Health, Ricerca Corrente 2022, 2023
- NIMH: K24MH121571
- Government of India grants to: Prof. Reddy [(SR/S0/HS/0016/2011) & (BT/PR13334/Med/30/259/2009)], Dr. Janardhanan Narayanaswamy (DST INSPIRE faculty grant -IFA12-LSBM-26) & (BT/06/IYBA/2012) and the Wellcome-DBT India Alliance grant to Dr. Ganesan Venkatasubramanian (500236/Z/11/Z)
- the Japan Agency for Medical Research and Development: JP22dm0307008
- DBT-Wellcome Trust India Alliance Early Career Fellowship grant (IA/CPHE/18/1/503956)
- NIMH: R21MH093889 and R01MH104648
- Grant #PI19/01171 from the Carlos III Health Institute, and 2017SGR 1247 from AGAUR-Generalitat de Catalunya.
- Italian Ministry of Health grant RC19-20-21-22/A
- Grants R01MH126981, R01MH111794, and R33MH107589 from the National Institute of Mental Health/National Institute of Health awarded to ERS.
- National Natural Science Foundation of China (Nos. 81871057, 82171495), and Key Technologies Research and Development Program of China (Nos.2022YFE0103700)
- Helse Vest Health Authority (Grant ID 911754 and 911880)
- JSPS KAKENHI (C) JP21K07547, 22K07598 and 22K15766.
- Ganesan Venkatasubramanian acknowledges the support of Department of Biotechnology (DBT) - Wellcome Trust India Alliance CRC grant (IA/CRC/19/1/610005) & senior fellowship grant (500236/Z/11/Z)
- Supported by an grant from Amsterdam Neuroscience CIA-2019-03-A
- Swiss National Science Foundation (no. 320030_130237, principal investigator: S.Walitza)
- The National Natural Science Foundation of China (82071518)
- Else Kröner Fresenius Stiftung (2017_A101)
- ENIGMA World Aging Center, NIA Award No. R01AG058854; ENIGMA Parkinson's Initiative: A Global Initiative for Parkinson's Disease, NINDS award RO1NS107513
- the Obsessive-Compulsive Foundation to Dan J. Stein
- Dutch Organization for Scientific Research (NWO/ZonMW) VENI grant (916-86-038) and Brain & Behavior Research Foundation (NARSAD grant), Netherlands Brain Foundation (2010(1)-50)
- Netherlands Organization for Scientific Research (NWO/ZonMW Vidi Grant No. 165.610.002, 016.156.318, and 917.15.318 G.A. van Wingen)
Collapse
Affiliation(s)
- Willem B Bruin
- Amsterdam UMC location University of Amsterdam, Department of Psychiatry, Meibergdreef 9, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Amsterdam, The Netherlands.
| | - Yoshinari Abe
- Department of Psychiatry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Pino Alonso
- Department of Psychiatry, Bellvitge University Hospital, Barcelona, Spain
- Department of Clinical Science, Faculty of Medicine, University of Barcelona, Barcelona, Spain
- IDIBELL, Bellvitge University Hospital, Barcelona, Spain
- CIBERSAM, Instituto de Salud Carlos III, Madrid, Spain
| | - Alan Anticevic
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Lea L Backhausen
- Department of Child and Adolescent Psychiatry, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Srinivas Balachander
- Department of Psychiatry, National Institute of Mental Health And Neurosciences (NIMHANS), Bangalore, India
| | - Nuria Bargallo
- CIBERSAM, Instituto de Salud Carlos III, Madrid, Spain
- Radiology Service, Diagnosis Image Center, Hospital Clinic de Barcelona, Barcelona, Spain
- Magnetic Resonance Image Core Facility, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Marcelo C Batistuzzo
- Department of Psychiatry, University of Sao Paulo School of Medicine, Sao Paulo, Brazil
- Department of Methods and Techniques in Psychology, Pontifical Catholic University, Sao Paulo, Brazil
| | - Francesco Benedetti
- Vita-Salute San Raffaele University, Milano, Italy
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Sara Bertolin Triquell
- Bellvitge Biomedical Research Insitute-IDIBELL, Bellvitge University Hospital, Barcelona, Spain
| | - Silvia Brem
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of Psychiatry, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Federico Calesella
- Vita-Salute San Raffaele University, Milano, Italy
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Beatriz Couto
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Clinical Academic Center-Braga, Braga, Portugal
| | - Damiaan A J P Denys
- Amsterdam UMC location University of Amsterdam, Department of Psychiatry, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Marco A N Echevarria
- Department of Psychiatry, University of Sao Paulo School of Medicine, Sao Paulo, Brazil
| | - Goi Khia Eng
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA
- Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Sónia Ferreira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Clinical Academic Center-Braga, Braga, Portugal
| | - Jamie D Feusner
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- General Adult Psychiatry & Health Systems, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | | | - Patricia Gruner
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Joyce Y Guo
- University of California, San Diego, CA, USA
| | - Kristen Hagen
- Molde Hospital, Møre og Romsdal Hospital Trust, Molde, Norway
- Bergen Center for Brain Plasticity, Haukeland University Hospital, Bergen, Norway
- Department of Mental Health, Norwegian University of Science and Technology, Trondheim, Norway
| | - Bjarne Hansen
- Bergen Center for Brain Plasticity, Haukeland University Hospital, Bergen, Norway
- Center for Crisis Psychology, University of Bergen, Bergen, Norway
| | - Yoshiyuki Hirano
- Research Center for Child Mental Development, Chiba University, Chiba, Japan
| | - Marcelo Q Hoexter
- Department of Psychiatry, University of Sao Paulo School of Medicine, Sao Paulo, Brazil
| | - Neda Jahanshad
- Imaging Genetics Center, Stevens Neuroimaging & Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Fern Jaspers-Fayer
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| | - Selina Kasprzak
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Department of Psychiatry, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Department of Anatomy and Neurosciences, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Minah Kim
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kathrin Koch
- Department of Neuroradiology, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Yoo Bin Kwak
- Department of Brain and Cognitive Sciences, Seoul National University College of Natural Sciences, Seoul, Republic of Korea
| | - Jun Soo Kwon
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Brain and Cognitive Sciences, Seoul National University College of Natural Sciences, Seoul, Republic of Korea
| | - Luisa Lazaro
- CIBERSAM, Instituto de Salud Carlos III, Madrid, Spain
- Magnetic Resonance Image Core Facility, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Department of Child and Adolescent Psychiatry and Psychology, Hospital Clinic of Barcelona, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
| | | | - Christine Lochner
- SA MRC Unit on Risk and Resilience in Mental Disorders, Department of Psychiatry, Stellenbosch University, Stellenbosch, South Africa
| | - Rachel Marsh
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Ignacio Martínez-Zalacaín
- Bellvitge Biomedical Research Insitute-IDIBELL, Bellvitge University Hospital, Barcelona, Spain
- Department of Clinical Sciences, University of Barcelona, Barcelona, Spain
| | - Jose M Menchon
- CIBERSAM, Instituto de Salud Carlos III, Madrid, Spain
- Bellvitge Biomedical Research Insitute-IDIBELL, Bellvitge University Hospital, Barcelona, Spain
- Department of Clinical Sciences, University of Barcelona, Barcelona, Spain
| | - Pedro S Moreira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Psychological Neuroscience Lab, CIPsi, School of Psychology, University of Minho, Braga, Portugal
| | - Pedro Morgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Clinical Academic Center-Braga, Braga, Portugal
| | - Akiko Nakagawa
- Research Center for Child Mental Development, Chiba University, Chiba, Japan
| | - Tomohiro Nakao
- Graduate School of Medical Sciences, Kyushu University, Fukuoka-shi, Japan
| | - Janardhanan C Narayanaswamy
- National Institute of Mental Health And Neurosciences (NIMHANS), Bangalore, India
- GVAMHS, Goulburn Valley Health, Shepparton, VIC, Australia
| | - Erika L Nurmi
- Department of Psychiatry and Biobehavioral Sciences, University of California at Los Angeles, Los Angeles, CA, USA
| | - Jose C Pariente Zorrilla
- Magnetic Resonance Image Core Facility, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - John Piacentini
- Division of Child and Adolescent Psychiatry, UCLA Semel Institute for Neuroscience, Los Angeles, CA, USA
| | - Maria Picó-Pérez
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Departamento de Psicología Básica, Clínica y Psicobiología, Universitat Jaume I, Castelló de la Plana, Spain
| | - Fabrizio Piras
- Laboratory of Neuropsychiatry, Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Federica Piras
- Laboratory of Neuropsychiatry, Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, Rome, Italy
| | | | - Janardhan Y C Reddy
- Department of Psychiatry, National Institute of Mental Health And Neurosciences (NIMHANS), Bangalore, India
| | - Daniela Rodriguez-Manrique
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Technical University of Munich, Munich, Germany
- TUM-Neuroimaging Center (TUM-NIC) of Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- Graduate School of Systemic Neurosciences (GSN), Ludwig-Maximilians-Universität, Munich, Germany
| | - Yuki Sakai
- Department of Psychiatry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
- ATR Brain Information Communication Research Laboratory Group, Kyoto, Japan
| | - Eiji Shimizu
- Research Center for Child Mental Development, Chiba University, Chiba, Japan
- United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Fukui, Japan
- Department of Cognitive Behavioral Physiology Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Venkataram Shivakumar
- Department of Integrative Medicine, National Institute of Mental Health And Neurosciences (NIMHANS), Bangalore, India
| | - Blair H Simpson
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Carles Soriano-Mas
- CIBERSAM, Instituto de Salud Carlos III, Madrid, Spain
- Bellvitge Biomedical Research Insitute-IDIBELL, Bellvitge University Hospital, Barcelona, Spain
- Department of Social Psychology and Quantitative Psychology, Universitat de Barcelona-UB, Barcelona, Spain
| | - Nuno Sousa
- ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Clinical Academic Center-Braga, Braga, Portugal
| | - Gianfranco Spalletta
- Laboratory of Neuropsychiatry, Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Emily R Stern
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA
- Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - S Evelyn Stewart
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
- British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
- British Columbia Mental Health and Substance Use Services Research Institute, Vancouver, BC, Canada
| | - Philip R Szeszko
- Department of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research, Education and Clinical Center (MIRECC), James J. Peters VA Medical Center, Bronx, NY, USA
| | - Jinsong Tang
- Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Sophia I Thomopoulos
- Imaging Genetics Center, Stevens Neuroimaging & Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Anders L Thorsen
- Bergen Center for Brain Plasticity, Haukeland University Hospital, Bergen, Norway
- Center for Crisis Psychology, University of Bergen, Bergen, Norway
| | - Tokiko Yoshida
- Research Center for Child Mental Development, Chiba University, Chiba, Japan
| | - Hirofumi Tomiyama
- Graduate School of Medical Sciences, Kyushu University, Fukuoka-shi, Japan
| | - Benedetta Vai
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Ilya M Veer
- Department of Developmental Psychology, University of Amsterdam, Amsterdam, The Netherlands
| | - Ganesan Venkatasubramanian
- Department of Psychiatry, National Institute of Mental Health And Neurosciences (NIMHANS), Bangalore, India
| | - Nora C Vetter
- Department of Child and Adolescent Psychiatry, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Department of Psychology, Faculty of Natural Sciences, MSB Medical School Berlin, Berlin, Germany
| | - Chris Vriend
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Department of Psychiatry, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Department of Anatomy and Neurosciences, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Compulsivity, Impulsivity & Attention program, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain Imaging program, Amsterdam, The Netherlands
| | - Susanne Walitza
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of Psychiatry, University of Zurich, Zurich, Switzerland
| | - Lea Waller
- Department of Psychiatry and Neurosciences CCM, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Zhen Wang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao, China
| | - Anri Watanabe
- Department of Psychiatry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Nicole Wolff
- Department of Child and Adolescent Psychiatry, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Je-Yeon Yun
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
- Yeongeon Student Support Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Qing Zhao
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao, China
| | - Wieke A van Leeuwen
- Amsterdam UMC location University of Amsterdam, Department of Psychiatry, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Hein J F van Marle
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Department of Psychiatry, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Mood Anxiety Psychosis Stress Sleep, Amsterdam, The Netherlands
| | - Laurens A van de Mortel
- Amsterdam UMC location University of Amsterdam, Department of Psychiatry, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Anouk van der Straten
- Amsterdam UMC location University of Amsterdam, Department of Psychiatry, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Ysbrand D van der Werf
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Department of Anatomy and Neurosciences, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Compulsivity, Impulsivity & Attention program, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain Imaging program, Amsterdam, The Netherlands
| | - Paul M Thompson
- Imaging Genetics Center, Stevens Neuroimaging & Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Dan J Stein
- SA MRC Unit on Risk and Resilience in Mental Disorders, Department of Psychiatry, Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Odile A van den Heuvel
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Department of Psychiatry, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Department of Anatomy and Neurosciences, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Compulsivity, Impulsivity & Attention program, Amsterdam, The Netherlands
| | - Guido A van Wingen
- Amsterdam UMC location University of Amsterdam, Department of Psychiatry, Meibergdreef 9, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Amsterdam, The Netherlands.
| |
Collapse
|
9
|
Higuchi Y, Arakawa H. Serotonergic mediation of the brain-wide neurogenesis: Region-dependent and receptor-type specific roles on neurogenic cellular transformation. CURRENT RESEARCH IN NEUROBIOLOGY 2023; 5:100102. [PMID: 37638344 PMCID: PMC10458724 DOI: 10.1016/j.crneur.2023.100102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 06/18/2023] [Accepted: 07/15/2023] [Indexed: 08/29/2023] Open
Abstract
Brain serotonin (5-hydroxytryptamine, 5-HT) is a key molecule for the mediation of depression-related brain states, but the neural mechanisms underlying 5-HT mediation need further investigation. A possible mechanism of the therapeutic antidepressant effects is neurogenic cell production, as stimulated by 5-HT signaling. Neurogenesis, the proliferation of neural stem cells (NSCs), and cell differentiation and maturation occur across brain regions, particularly the hippocampal dentate gyrus and the subventricular zone, throughout one's lifespan. 5-HT plays a major role in the mediation of neurogenic processes, which in turn leads to the therapeutic effect on depression-related states. In this review article, we aim to identify how the neuronal 5-HT system mediates the process of neurogenesis, including cell proliferation, cell-type differentiation and maturation. First, we will provide an overview of the neurogenic cell transformation that occurs in brain regions containing or lacking NSCs. Second, we will review brain region-specific mechanisms of 5-HT-mediated neurogenesis by comparing regions localized to NSCs, i.e., the hippocampus and subventricular zone, with those not containing NSCs. Highlighting these 5-HT mechanisms that mediate neurogenic cell production processes in a brain-region-specific manner would provide unique insights into the role of 5-HT in neurogenesis and its associated effects on depression.
Collapse
Affiliation(s)
- Yuki Higuchi
- Department of Systems Physiology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Hiroyuki Arakawa
- Department of Systems Physiology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| |
Collapse
|
10
|
Pinto L, Macedo J, Araújo B, Anjo S, Silveira-Rosa T, Patrício P, Teixeira F, Manadas B, Rodrigues AJ, Lepore A, Salgado A, Gomes E. Glial-Restricted Precursors stimulate endogenous cytogenesis and effectively recover emotional deficits in a model of cytogenesis ablation. RESEARCH SQUARE 2023:rs.3.rs-2747462. [PMID: 37034743 PMCID: PMC10081440 DOI: 10.21203/rs.3.rs-2747462/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
Abstract
Adult cytogenesis, the continuous generation of newly-born neurons (neurogenesis) and glial cells (gliogenesis) throughout life, is highly impaired in several neuropsychiatric disorders, such as Major Depressive Disorder (MDD), impacting negatively on cognitive and emotional domains. Despite playing a critical role in brain homeostasis, the importance of gliogenesis has been overlooked, both in healthy and diseased states. To examine the role of newly formed glia, we transplanted Glial Restricted Precursors (GRPs) into the adult hippocampal dentate gyrus (DG), or injected their secreted factors (secretome), into a previously validated transgenic GFAP-tk rat line, in which cytogenesis is transiently compromised. We explored the long-term effects of both treatments on physiological and behavioral outcomes. Grafted GRPs reversed anxiety-like and depressive-like deficits, while the secretome promoted recovery of only anxiety-like behavior. Furthermore, GRPs elicited a recovery of neurogenic and gliogenic levels in the ventral DG, highlighting the unique involvement of these cells in the regulation of brain cytogenesis. Both GRPs and their secretome induced significant alterations in the DG proteome, directly influencing proteins and pathways related to cytogenesis, regulation of neural plasticity and neuronal development. With this work, we demonstrate a valuable and specific contribution of glial progenitors to normalizing gliogenic levels, rescueing neurogenesis and, importantly, promoting recovery of emotional deficits characteristic of disorders such as MDD.
Collapse
Affiliation(s)
| | | | | | - Sandra Anjo
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Khan MM, Parikh V. Prospects for Neurotrophic Factor-Based Early Intervention in Schizophrenia: Lessons Learned from the Effects of Antipsychotic Drugs on Cognition, Neurogenesis, and Neurotrophic Factors. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2023; 22:289-303. [PMID: 35366786 DOI: 10.2174/1871527321666220401124151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 02/13/2022] [Accepted: 02/18/2022] [Indexed: 12/16/2022]
Abstract
Although reducing psychotic symptoms in schizophrenia has been a major focus of therapeutic interventions for decades, improving cognition is considered a better predictor of functional outcomes. However, the most commonly prescribed antipsychotic drugs (APDs) show only marginal beneficial effects on cognition in patients with schizophrenia. The neural mechanisms underlying cognitive disturbances in schizophrenia remain unknown that making drug development efforts very challenging. Since neurotrophic factors are the primary architects of neurogenesis, synaptic plasticity, learning, and memory, the findings from preclinical and clinical studies that assess changes in neurogenesis and neurotrophic factors and their relationship to cognitive performance in schizophrenia, and how these mechanisms might be impacted by APD treatment, may provide valuable clues in developing therapies to combat cognitive deficit in schizophrenia. Numerous evidence produced over the years suggests a deficit in a wide spectrum of neurotrophic factors in schizophrenia. Since schizophrenia is considered a neurodevelopmental disorder, early intervention with neurotrophic factors may be more effective in ameliorating the cognitive deficits and psychopathological symptoms associated with this pathology. In this context, results from initial clinical trials with neurotrophic factors and their future potential to improve cognition and psychosocial functioning in schizophrenia are discussed.
Collapse
Affiliation(s)
- Mohammad M Khan
- Laboratory of Translational Neurology and Molecular Psychiatry, Department of Biotechnology, Era\'s Lucknow Medical College and Hospital, and Faculty of Science, Era University, Lucknow, UP, India
| | - Vinay Parikh
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, USA
| |
Collapse
|
12
|
Shu S, Xu SY, Ye L, Liu Y, Cao X, Jia JQ, Bian HJ, Liu Y, Zhu XL, Xu Y. Prefrontal parvalbumin interneurons deficits mediate early emotional dysfunction in Alzheimer's disease. Neuropsychopharmacology 2023; 48:391-401. [PMID: 36229597 PMCID: PMC9750960 DOI: 10.1038/s41386-022-01435-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 07/19/2022] [Accepted: 08/16/2022] [Indexed: 12/26/2022]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease and has an insidious onset. Exploring the characteristics and mechanism of the early symptoms of AD plays a critical role in the early diagnosis and intervention of AD. Here we found that depressive-like behavior and short-term spatial memory dysfunction appeared in APPswe/PS1dE9 mice (AD mice) as early as 9-11 weeks of age. Electrophysiological analysis revealed excitatory/inhibitory (E/I) imbalance in the prefrontal cortex (PFC). This E/I imbalance was induced by significant reduction in the number and activity of parvalbumin interneurons (PV+ INs) in this region. Furthermore, optogenetic and chemogenetic activation of residual PV+ INs effectively ameliorated depressive-like behavior and rescued short-term spatial memory in AD mice. These results suggest the PFC is selectively vulnerable in the early stage of AD and prefrontal PV+ INs deficits play a key role in the occurrence and development of early symptoms of AD.
Collapse
Affiliation(s)
- Shu Shu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, Jiangsu, PR China
- Institute of Brain Sciences, Nanjing University, Nanjing, 210093, Jiangsu, PR China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, Jiangsu, PR China
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, 210008, Jiangsu, PR China
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, 210008, Jiangsu, PR China
| | - Si-Yi Xu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, Jiangsu, PR China
- Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Zhenjiang, 212013, Jiangsu, PR China
| | - Lei Ye
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, Jiangsu, PR China
| | - Yi Liu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, Jiangsu, PR China
- Institute of Brain Sciences, Nanjing University, Nanjing, 210093, Jiangsu, PR China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, Jiangsu, PR China
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, 210008, Jiangsu, PR China
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, 210008, Jiangsu, PR China
| | - Xiang Cao
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, Jiangsu, PR China
- Institute of Brain Sciences, Nanjing University, Nanjing, 210093, Jiangsu, PR China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, Jiangsu, PR China
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, 210008, Jiangsu, PR China
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, 210008, Jiangsu, PR China
| | - Jun-Qiu Jia
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, Jiangsu, PR China
| | - Hui-Jie Bian
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, Jiangsu, PR China
- Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China
| | - Ying Liu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, Jiangsu, PR China
| | - Xiao-Lei Zhu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, Jiangsu, PR China
- Institute of Brain Sciences, Nanjing University, Nanjing, 210093, Jiangsu, PR China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, Jiangsu, PR China
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, 210008, Jiangsu, PR China
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, 210008, Jiangsu, PR China
| | - Yun Xu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, Jiangsu, PR China.
- Institute of Brain Sciences, Nanjing University, Nanjing, 210093, Jiangsu, PR China.
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, Jiangsu, PR China.
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, 210008, Jiangsu, PR China.
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, 210008, Jiangsu, PR China.
| |
Collapse
|
13
|
Kukuia KK, Appiah F, Dugbartey GJ, Takyi YF, Amoateng P, Amponsah SK, Adi-Dako O, Koomson AE, Ayertey F, Adutwum-Ofosu KK. Extract of Mallotus oppositifolius (Geiseler) Müll. Arg. increased prefrontal cortex dendritic spine density and serotonin and attenuated para-chlorophenylalanine-aggravated aggressive and depressive behaviors in mice. Front Pharmacol 2022; 13:962549. [PMID: 36386158 PMCID: PMC9649488 DOI: 10.3389/fphar.2022.962549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 10/07/2022] [Indexed: 11/30/2022] Open
Abstract
Background/Aim: Depression-related aggression is linked to serotonin (5-HT) and dendritic spine alterations. Although Mallotus oppositifolius extract (MOE) has potential for reducing this effect, its specific role remains uncertain. Herein, we evaluated this potential and associated alterations in the brain. Methods: A standard resident-intruder model of para-chlorophenylalanine (pCPA)-induced depression-associated aggression in male ICR mice was used. The resident mice received pCPA (300 mg/kg, i. p.) for 3 consecutive days while saline-treated mice served as negative control. The pCPA aggressive mice were subsequently treated orally with either MOE (30, 100, 300 mg/kg), fluoxetine (20 mg/kg), tryptophan (20 mg/kg) or saline (untreated pCPA group) for 28 days. Locomotor activity was assessed using open field test. Serotonin (5-HT) levels in mice brain and phytochemical fingerprint of MOE were determined by high performance liquid chromatography (HPLC) while gas chromatography-mass spectrometry (GC-MS) was used to identify constituents of MOE. Dendritic spine density and morphology were evaluated using Golgi-Cox staining technique and analyzed with ImageJ and Reconstruct software. Results: Administration of pCPA induced aggressive behavior in mice, evidenced by increased attack behaviors (increased number and duration of attacks), which positively correlated with squeaking and tail rattling. MOE treatment significantly reduced these characteristics of aggression in comparison with vehicle (non-aggressive) and untreated pCPA groups (p < 0.001), and also reduced social exploration behavior. Although the behavioral effects of MOE were comparable to those of fluoxetine and tryptophan, these effects were quicker compared to fluoxetine and tryptophan. Additionally, MOE also markedly increased 5-HT concentration and dendritic spine density in the prefrontal cortex relative to vehicle and untreated pCPA groups (p < 0.05). Interestingly, these behavioral effects were produced without compromising locomotor activity. GC-MS analysis of the MOE identified 17 known compounds from different chemical classes with anti-inflammatory, antioxidant, neuroprotective and antidepressant activities, which may have contributed to its anti-aggressive effect. Conclusion: MOE decreased depression-associated aggressive behavior in mice via increased 5-HT concentration and dendritic spine density in the prefrontal cortex. The MOE-mediated effects were faster than those of fluoxetine and tryptophan. Our finding suggests that MOE may have clinical promise in decreasing aggressive and depressive behaviors.
Collapse
Affiliation(s)
- Kennedy K.E. Kukuia
- Department of Medical Pharmacology, University of Ghana Medical School, College of Health Sciences, University of Ghana, Accra, Ghana
- *Correspondence: Kennedy K.E. Kukuia, ; Kevin K. Adutwum-Ofosu,
| | - Frimpong Appiah
- Department of Community Health and Medicine, School of Food and Health Sciences, Anglican University College of Technology, Nkoranza, Ghana
| | - George J. Dugbartey
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Yaw F. Takyi
- Department of Medical Pharmacology, University of Ghana Medical School, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Patrick Amoateng
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Seth K. Amponsah
- Department of Medical Pharmacology, University of Ghana Medical School, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Ofosua Adi-Dako
- Department of Pharmaceutics and Microbiology, School of Pharmacy, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Awo E. Koomson
- Department of Medical Pharmacology, University of Ghana Medical School, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Frederick Ayertey
- Department of Phytochemistry, Center for Plant Medicine Research, Mampong-Akuapem, Ghana
| | - Kevin K. Adutwum-Ofosu
- Department of Anatomy, University of Ghana Medical School, College of Health Sciences, University of Ghana, Accra, Ghana
- *Correspondence: Kennedy K.E. Kukuia, ; Kevin K. Adutwum-Ofosu,
| |
Collapse
|
14
|
Jones KL, Zhou M, Jhaveri DJ. Dissecting the role of adult hippocampal neurogenesis towards resilience versus susceptibility to stress-related mood disorders. NPJ SCIENCE OF LEARNING 2022; 7:16. [PMID: 35842419 PMCID: PMC9288448 DOI: 10.1038/s41539-022-00133-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 07/01/2022] [Indexed: 05/13/2023]
Abstract
Adult hippocampal neurogenesis in the developmental process of generating and integrating new neurons in the hippocampus during adulthood and is a unique form of structural plasticity with enormous potential to modulate neural circuit function and behaviour. Dysregulation of this process is strongly linked to stress-related neuropsychiatric conditions such as anxiety and depression, and efforts have focused on unravelling the contribution of adult-born neurons in regulating stress response and recovery. Chronic stress has been shown to impair this process, whereas treatment with clinical antidepressants was found to enhance the production of new neurons in the hippocampus. However, the precise role of adult hippocampal neurogenesis in mediating the behavioural response to chronic stress is not clear and whether these adult-born neurons buffer or increase susceptibility to stress-induced mood-related maladaptation remains one of the controversial issues. In this review, we appraise evidence probing the causal role of adult hippocampal neurogenesis in the regulation of emotional behaviour in rodents. We find that the relationship between adult-born hippocampal neurons and stress-related mood disorders is not linear, and that simple subtraction or addition of these neurons alone is not sufficient to lead to anxiety/depression or have antidepressant-like effects. We propose that future studies examining how stress affects unique properties of adult-born neurons, such as the excitability and the pattern of connectivity during their critical period of maturation will provide a deeper understanding of the mechanisms by which these neurons contribute to functional outcomes in stress-related mood disorders.
Collapse
Affiliation(s)
- Katherine L Jones
- Queensland Brain Institute, University of Queensland, Brisbane, QLD 4072, Australia
| | - Mei Zhou
- Queensland Brain Institute, University of Queensland, Brisbane, QLD 4072, Australia
- Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Dhanisha J Jhaveri
- Queensland Brain Institute, University of Queensland, Brisbane, QLD 4072, Australia.
- Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia.
| |
Collapse
|
15
|
Osacka J, Kiss A, Bacova Z, Tillinger A. Effect of Haloperidol and Olanzapine on Hippocampal Cells’ Proliferation in Animal Model of Schizophrenia. Int J Mol Sci 2022; 23:ijms23147711. [PMID: 35887056 PMCID: PMC9323809 DOI: 10.3390/ijms23147711] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/07/2022] [Accepted: 07/12/2022] [Indexed: 12/07/2022] Open
Abstract
Aberrant neurogenesis in the subventricular zone (SVZ) and hippocampus (HIP) contributes to schizophrenia pathogenesis. Haloperidol (HAL) and olanzapine (OLA), commonly prescribed antipsychotics for schizophrenia treatment, affect neurogenesis too. The effect of HAL and OLA on an mHippoE-2 cell line was studied in vitro where we measured the cell number and projection length. In vivo, we studied the gene expression of DCX, Sox2, BDNF, and NeuN in the SVZ and HIP in an MK-801-induced animal schizophrenia model. Cells were incubated with HAL, OLA, and MK-801 for 24, 48, and 72 h. Animals were injected for 6 days with saline or MK801 (0.5 mg/kg), and from the 7th day with either vehicle HAL (1 mg/kg) or OLA (2 mg/kg), for the next 7 days. In vitro, HAL and OLA dose/time-dependently suppressed cells’ proliferation and shortened their projection length. HAL/OLA co-treatment with MK-801 for 24 h reversed HAL’s/OLA’s inhibitory effect. In vivo, HAL and OLA suppressed DCX and NeuN genes’ expression in the HIP and SVZ. MK-801 decreased DCX and NeuN genes’ expression in the HIP and OLA prevented this effect. The data suggest that subchronic HAL/OLA treatment can inhibit DCX and NeuN expression. In an MK-801 schizophrenia model, OLA reversed the MK-801 inhibitory effect on DCX and NeuN and HAL reversed the effect on DCX expression; however, only in the HIP.
Collapse
|
16
|
Waszczuk K, Kucharska-Mazur J, Tyburski E, Rek-Owodziń K, Plichta P, Rudkowski K, Podwalski P, Grąźlewski T, Mak M, Misiak B, Michalczyk A, Tarnowski M, Sielatycka K, Szczęśniak A, Łuczkowska K, Dołęgowska B, Budkowska M, Ratajczak MZ, Samochowiec J. Psychopathology and Stem Cell Mobilization in Ultra-High Risk of Psychosis and First-Episode Psychosis Patients. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19106001. [PMID: 35627537 PMCID: PMC9141672 DOI: 10.3390/ijerph19106001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/07/2022] [Accepted: 05/13/2022] [Indexed: 02/04/2023]
Abstract
Although regenerative and inflammatory processes are involved in the etiopathogenesis of many psychiatric disorders, their roles are poorly understood. We investigate the potential role of stem cells (SC) and factors influencing the trafficking thereof, such as complement cascade (CC) components, phospholipid substrates, and chemokines, in the etiology of schizophrenia. We measured sphingosine-1-phosphate (S1P), stromal-derived factor 1 (SDF-1), and CC cleavage fragments (C3a, C5a, and C5b-C9; also known as the membrane attack complex) in the peripheral blood of 49 unrelated patients: 9 patients with ultra-high risk of psychosis (UHR), 22 patients with first-episode psychosis (FEP), and 18 healthy controls (HC). When compared with the HC group, the UHR and FEP groups had higher levels of C3a. We found no significant differences in hematopoietic SC, very small embryonic-like stem cell (VSEL), C5a, S1P, or SDF-1 levels in the UHR and FEP groups. However, among FEP patients, there was a significant positive correlation between VSELs (CD133+) and negative symptoms. These preliminary findings support the role of the immune system and regenerative processes in the etiology of schizophrenia. To establish the relevance of SC and other factors affecting the trafficking thereof as potential biomarkers of schizophrenia, more studies on larger groups of individuals from across the disease spectrum are needed.
Collapse
Affiliation(s)
- Katarzyna Waszczuk
- Department of Psychiatry, Pomeranian Medical University in Szczecin, Broniewskiego 26, 71-460 Szczecin, Poland; (J.K.-M.); (K.R.); (P.P.); (T.G.); (A.M.); (J.S.)
- Correspondence: ; Tel./Fax: +48-91-35-11-322
| | - Jolanta Kucharska-Mazur
- Department of Psychiatry, Pomeranian Medical University in Szczecin, Broniewskiego 26, 71-460 Szczecin, Poland; (J.K.-M.); (K.R.); (P.P.); (T.G.); (A.M.); (J.S.)
| | - Ernest Tyburski
- Department of Health Psychology, Pomeranian Medical University in Szczecin, Broniewskiego 26, 71-460 Szczecin, Poland; (E.T.); (K.R.-O.); (P.P.); (M.M.)
| | - Katarzyna Rek-Owodziń
- Department of Health Psychology, Pomeranian Medical University in Szczecin, Broniewskiego 26, 71-460 Szczecin, Poland; (E.T.); (K.R.-O.); (P.P.); (M.M.)
| | - Piotr Plichta
- Department of Health Psychology, Pomeranian Medical University in Szczecin, Broniewskiego 26, 71-460 Szczecin, Poland; (E.T.); (K.R.-O.); (P.P.); (M.M.)
| | - Krzysztof Rudkowski
- Department of Psychiatry, Pomeranian Medical University in Szczecin, Broniewskiego 26, 71-460 Szczecin, Poland; (J.K.-M.); (K.R.); (P.P.); (T.G.); (A.M.); (J.S.)
| | - Piotr Podwalski
- Department of Psychiatry, Pomeranian Medical University in Szczecin, Broniewskiego 26, 71-460 Szczecin, Poland; (J.K.-M.); (K.R.); (P.P.); (T.G.); (A.M.); (J.S.)
| | - Tomasz Grąźlewski
- Department of Psychiatry, Pomeranian Medical University in Szczecin, Broniewskiego 26, 71-460 Szczecin, Poland; (J.K.-M.); (K.R.); (P.P.); (T.G.); (A.M.); (J.S.)
| | - Monika Mak
- Department of Health Psychology, Pomeranian Medical University in Szczecin, Broniewskiego 26, 71-460 Szczecin, Poland; (E.T.); (K.R.-O.); (P.P.); (M.M.)
| | - Błażej Misiak
- Department of Psychiatry, Division of Consultation Psychiatry and Neuroscience, Wroclaw Medical University, 50-367 Wroclaw, Poland;
| | - Anna Michalczyk
- Department of Psychiatry, Pomeranian Medical University in Szczecin, Broniewskiego 26, 71-460 Szczecin, Poland; (J.K.-M.); (K.R.); (P.P.); (T.G.); (A.M.); (J.S.)
| | - Maciej Tarnowski
- Department of Physiology, Pomeranian University of Medicine, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland;
| | - Katarzyna Sielatycka
- Institute of Biology, Faculty of Exact and Natural Sciences, University of Szczecin, Felczaka 3c, 71-415 Szczecin, Poland;
| | - Angelika Szczęśniak
- Department of Laboratory Medicine, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland; (A.S.); (B.D.)
| | - Karolina Łuczkowska
- Department of General Pathology, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland;
| | - Barbara Dołęgowska
- Department of Laboratory Medicine, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland; (A.S.); (B.D.)
| | - Marta Budkowska
- Department of Medical Analytics, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland;
| | - Mariusz Z. Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40292, USA;
| | - Jerzy Samochowiec
- Department of Psychiatry, Pomeranian Medical University in Szczecin, Broniewskiego 26, 71-460 Szczecin, Poland; (J.K.-M.); (K.R.); (P.P.); (T.G.); (A.M.); (J.S.)
| |
Collapse
|
17
|
Effect of dimethyl fumarate on the changes in the medial prefrontal cortex structure and behavior in the poly(I:C)-induced maternal immune activation model of schizophrenia in the male mice. Behav Brain Res 2022; 417:113581. [PMID: 34530042 DOI: 10.1016/j.bbr.2021.113581] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/31/2021] [Accepted: 09/09/2021] [Indexed: 11/21/2022]
Abstract
BACKGROUND The link between maternal immune activation (MIA) and the risk of developing schizophrenia (SCZ) later in life has been of major focus in recent years. This link could be bridged by activated inflammatory pathways and excessive cytokine release resulting in adverse effects on behavior, histology, and cytoarchitecture. The down-regulatory effects of immunomodulatory agents on the activated glial cells and their therapeutic effects on schizophrenic patients are consistent with this hypothesis. OBJECTIVE We investigated whether treatment with the anti-inflammatory drug dimethyl fumarate (DMF) could rescue impacts of prenatal exposure to polyinosinic:polycytidylic acid [poly (I:C)]. METHODS Pregnant dams were administered poly(I:C) at gestational day 9.5. Offspring born from these mothers were treated with DMF for fourteen consecutive days from postnatal day 80 and were assessed behaviorally before and after treatment. The brains were then stained with Cresyl Violet or Golgi-Cox. In addition to the estimation of stereological parameters, cytoarchitectural changes were also evaluated in the medial prefrontal cortex. RESULTS MIA caused some abnormalities in behavior, as well as changes in the number of neurons and non-neurons. These alterations were also extended to pyramidal layer III neurons with a significant decrease in dendritic complexity and spine density which DMF treatment could prevent these changes. Furthermore, DMF treatment was also effective against abnormal exploratory and depression-related behavior, but not the changes in the number of cells. CONCLUSION These findings support the idea of using anti-inflammatory agents as adjunctive therapy in patients with SCZ.
Collapse
|
18
|
Glial Cell Abnormalities in Major Psychiatric Diseases: A Systematic Review of Postmortem Brain Studies. Mol Neurobiol 2022; 59:1665-1692. [PMID: 35013935 DOI: 10.1007/s12035-021-02672-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 11/25/2021] [Indexed: 10/19/2022]
Abstract
There have been a large number of reports about glial cell dysfunction being related to major psychiatric diseases such as schizophrenia (SCZ), bipolar disorder (BD), and major depressive disorder (MDD). In this review, we provide an overview of postmortem studies analyzing the structural changes of glial cells in these three major psychiatric diseases, including the density, number and size of glial cells, and the expression of related markers. Up to May 1, 2021, 108 articles that met the inclusion criteria were identified by searching PubMed and Web of Science. Although most studies evaluating total glial cells did not show abnormalities in the brains of postmortem patients, astrocytes, microglial cells, and oligodendrocytes seem to have specific patterns of changes in each disease. For example, out of 20 studies that evaluated astrocyte markers in MDD, 11 studies found decreased astrocyte marker expression in MDD patients. Similarly, out of 25 studies evaluating oligodendrocyte markers in SCZ, 15 studies showed decreased expression of oligodendrocyte markers in different brain regions of SCZ patients. In addition, activated microglial cells were observed in patients with SCZ, BD, and MDD, but suicide may be a confounding factor for the observed effects. Although the data from the included studies were heterogeneous and this cannot be fully explained at present, it is likely that there are a variety of contributing factors, including the measured brain regions, methods of measurement, gender, age at time of death, and medications.
Collapse
|
19
|
Present and future antipsychotic drugs: a systematic review of the putative mechanisms of action for efficacy and a critical appraisal under a translational perspective. Pharmacol Res 2022; 176:106078. [PMID: 35026403 DOI: 10.1016/j.phrs.2022.106078] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/23/2021] [Accepted: 01/07/2022] [Indexed: 01/10/2023]
Abstract
Antipsychotics represent the mainstay of schizophrenia pharmacological therapy, and their role has been expanded in the last years to mood disorders treatment. Although introduced in 1952, many years of research were required before an accurate picture of how antipsychotics work began to emerge. Despite the well-recognized characterization of antipsychotics in typical and atypical based on their liability to induce motor adverse events, their main action at dopamine D2R to elicit the "anti-psychotic" effect, as well as the multimodal action at other classes of receptors, their effects on intracellular mechanisms starting with receptor occupancy is still not completely understood. Significant lines of evidence converge on the impact of these compounds on multiple molecular signaling pathways implicated in the regulation of early genes and growth factors, dendritic spine shape, brain inflammation, and immune response, tuning overall the function and architecture of the synapse. Here we present, based on PRISMA approach, a comprehensive and systematic review of the above mechanisms under a translational perspective to disentangle those intracellular actions and signaling that may underline clinically relevant effects and represent potential targets for further innovative strategies in antipsychotic therapy.
Collapse
|
20
|
Reduced adult neurogenesis is associated with increased macrophages in the subependymal zone in schizophrenia. Mol Psychiatry 2021; 26:6880-6895. [PMID: 34059796 DOI: 10.1038/s41380-021-01149-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 04/17/2021] [Accepted: 04/26/2021] [Indexed: 02/06/2023]
Abstract
Neural stem cells in the human subependymal zone (SEZ) generate neuronal progenitor cells that can differentiate and integrate as inhibitory interneurons into cortical and subcortical brain regions; yet the extent of adult neurogenesis remains unexplored in schizophrenia and bipolar disorder. We verified the existence of neurogenesis across the lifespan by chartering transcriptional alterations (2 days-103 years, n = 70) and identifying cells indicative of different stages of neurogenesis in the human SEZ. Expression of most neural stem and neuronal progenitor cell markers decreased during the first postnatal years and remained stable from childhood into ageing. We next discovered reduced neural stem and neuronal progenitor cell marker expression in the adult SEZ in schizophrenia and bipolar disorder compared to controls (n = 29-32 per group). RNA sequencing identified increased expression of the macrophage marker CD163 as the most significant molecular change in schizophrenia. CD163+ macrophages, which were localised along blood vessels and in the parenchyma within 10 µm of neural stem and progenitor cells, had increased density in schizophrenia but not in bipolar disorder. Macrophage marker expression negatively correlated with neuronal progenitor marker expression in schizophrenia but not in controls or bipolar disorder. Reduced neurogenesis and increased macrophage marker expression were also associated with polygenic risk for schizophrenia. Our results support that the human SEZ retains the capacity to generate neuronal progenitor cells throughout life, although this capacity is limited in schizophrenia and bipolar disorder. The increase in macrophages in schizophrenia but not in bipolar disorder indicates that immune cells may impair neurogenesis in the adult SEZ in a disease-specific manner.
Collapse
|
21
|
Kimura M, Oda Y, Hirose Y, Kimura H, Yoshino K, Niitsu T, Kanahara N, Shirayama Y, Hashimoto K, Iyo M. Upregulation of heat-shock protein HSP-70 and glutamate transporter-1/glutamine synthetase in the striatum and hippocampus in haloperidol-induced dopamine-supersensitivity-state rats. Pharmacol Biochem Behav 2021; 211:173288. [PMID: 34653399 DOI: 10.1016/j.pbb.2021.173288] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 10/07/2021] [Accepted: 10/07/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND The excessive blockade of dopamine D2 receptors (DRD2s) with long-term antipsychotic treatment is known to induce a dopamine supersensitivity state (DSS). The mechanism of DSS is speculated to be a compensatory up-regulation of DRD2s, but an excess blockade of DRD2s can also cause glutamatergic neuronal damage. Herein, we investigated whether antipsychotic-induced neuronal damage plays a role in the development of DSS. METHODS Haloperidol (HAL; 0.75 mg/kg/day for 14 days) or vehicle was administered to rats via an osmotic mini-pump. Haloperidol-treated rats were divided into groups of DSS rats and non-DSS rats based on their voluntary locomotion data. We then determined the tissue levels of glutamate transporter-1 (GLT-1)/glutamine synthetase (GS) and heat shock protein-70 (HSP-70) in the rats' brain regions. RESULTS The levels of HSP-70 in the striatum and CA-3 region of the DSS rats were significantly higher than those of the control and non-DSS rats, whereas the dentate gyrus HSP-70 levels in both the DSS and non-DSS rats were increased versus the controls. The levels of GLT-1/GS in the CA-3 and nucleus accumbens were increased in the DSS rats. CONCLUSIONS These results suggest that the DSS rats experienced striatal neuronal damage and indicate that a HAL-induced upregulation of HSP-70 and the GLT-1/GS system in the CA3 may be involved in the development of DSS. It remains unknown why the non-DSS rats did not suffer neuronal damage. In view of the need for therapeutic strategies for treatment-resistant schizophrenia, dopamine supersensitivity psychosis, and tardive dyskinesia, further investigations of our findings are warranted.
Collapse
Affiliation(s)
- Makoto Kimura
- Department of Psychiatry, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuou-ku, Chiba, Chiba 260-8670, Japan
| | - Yasunori Oda
- Department of Psychiatry, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuou-ku, Chiba, Chiba 260-8670, Japan.
| | - Yuki Hirose
- Department of Psychiatry, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuou-ku, Chiba, Chiba 260-8670, Japan
| | - Hiroshi Kimura
- Department of Psychiatry, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuou-ku, Chiba, Chiba 260-8670, Japan; Department of Psychiatry, School of Medicine, International University of Health and Welfare, 4-3 Kozunomori, Narita, Chiba 286-8686, Japan
| | - Kouhei Yoshino
- Department of Psychiatry, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuou-ku, Chiba, Chiba 260-8670, Japan
| | - Tomihisa Niitsu
- Department of Psychiatry, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuou-ku, Chiba, Chiba 260-8670, Japan
| | - Nobuhisa Kanahara
- Division of Medical Treatment and Rehabilitation, Chiba University Center for Forensic Mental Health, 1-8-1 Inohana, Chuou-ku, Chiba, Chiba 260-8670, Japan
| | - Yukihiko Shirayama
- Department of Psychiatry, Teikyo University Chiba Medical Center, 3426-3 Anesaki, Ichihara, Chiba 290-0111, Japan
| | - Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, 1-8-1 Inohana, Chuou-ku, Chiba, Chiba 260-8670, Japan
| | - Masaomi Iyo
- Department of Psychiatry, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuou-ku, Chiba, Chiba 260-8670, Japan
| |
Collapse
|
22
|
Martins-Macedo J, Salgado AJ, Gomes ED, Pinto L. Adult brain cytogenesis in the context of mood disorders: From neurogenesis to the emergent role of gliogenesis. Neurosci Biobehav Rev 2021; 131:411-428. [PMID: 34555383 DOI: 10.1016/j.neubiorev.2021.09.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 09/06/2021] [Accepted: 09/16/2021] [Indexed: 12/18/2022]
Abstract
Psychiatric disorders severely impact patients' lives. Motivational, cognitive and emotional deficits are the most common symptoms observed in these patients and no effective treatment is still available, either due to the adverse side effects or the low rate of efficacy of currently available drugs. Neurogenesis recovery has been one important focus in the treatment of psychiatric disorders, which undeniably contributes to the therapeutic action of antidepressants. However, glial plasticity is emerging as a new strategy to explore the deficits observed in mood disorders and the efficacy of therapeutic interventions. Thus, it is crucial to understand the mechanisms behind glio- and neurogenesis to better define treatments and preventive therapies, once adult cytogenesis is of pivotal importance to cognitive and emotional components of behavior, both in healthy and pathological contexts, including in psychiatric disorders. Here, we review the concepts and history of neuro- and gliogenesis, providing as well a reflection on the functional importance of cytogenesis in the context of disease.
Collapse
Affiliation(s)
- Joana Martins-Macedo
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Eduardo D Gomes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Luísa Pinto
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
23
|
Rodrigues RS, Paulo SL, Moreira JB, Tanqueiro SR, Sebastião AM, Diógenes MJ, Xapelli S. Adult Neural Stem Cells as Promising Targets in Psychiatric Disorders. Stem Cells Dev 2021; 29:1099-1117. [PMID: 32723008 DOI: 10.1089/scd.2020.0100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The development of new therapies for psychiatric disorders is of utmost importance, given the enormous toll these disorders pose to society nowadays. This should be based on the identification of neural substrates and mechanisms that underlie disease etiopathophysiology. Adult neural stem cells (NSCs) have been emerging as a promising platform to counteract brain damage. In this perspective article, we put forth a detailed view of how NSCs operate in the adult brain and influence brain homeostasis, having profound implications at both behavioral and functional levels. We appraise evidence suggesting that adult NSCs play important roles in regulating several forms of brain plasticity, particularly emotional and cognitive flexibility, and that NSC dynamics are altered upon brain pathology. Furthermore, we discuss the potential therapeutic value of utilizing adult endogenous NSCs as vessels for regeneration, highlighting their importance as targets for the treatment of multiple mental illnesses, such as affective disorders, schizophrenia, and addiction. Finally, we speculate on strategies to surpass current challenges in neuropsychiatric disease modeling and brain repair.
Collapse
Affiliation(s)
- Rui S Rodrigues
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Sara L Paulo
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - João B Moreira
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Sara R Tanqueiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Maria J Diógenes
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Sara Xapelli
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
24
|
Ouyang SH, Zhai YJ, Wu YP, Xie G, Wang GE, Mao ZF, Hu HH, Luo XH, Sun WY, Liang L, Duan WJ, Kurihara H, Li YF, He RR. Theacrine, a Potent Antidepressant Purine Alkaloid from a Special Chinese Tea, Promotes Adult Hippocampal Neurogenesis in Stressed Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:7016-7027. [PMID: 34060828 DOI: 10.1021/acs.jafc.1c01514] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Daily intake of tea has been known to relate to a low risk of depression. In this study, we report that a special variety of tea in China, Camellia assamica var. kucha (kucha), possesses antidepressant effects but with less adverse effects as compared to traditional tea Camellia sinensis. This action of kucha is related to its high amount of theacrine, a purine alkaloid structurally similar to caffeine. We investigated the antidepressant-like effects and mechanisms of theacrine in chronic water immersion restraint stress and chronic unpredictable mild stress mice models. PC12 cells and primary hippocampal neural stem cells were treated with stress hormone corticosterone (CORT) to reveal the potential antidepression mechanism of theacrine from the perspective of adult hippocampus neurogenesis. Results of behavioral and neurotransmitter analysis showed that intragastric administration of theacrine significantly counteracted chronic stress-induced depression-like disorders and abnormal 5-hydroxytryptamine (5-HT) metabolism with less central excitability. Further investigation from both in vivo and in vitro experiments indicated that the antidepressant mechanism of theacrine was associated with promoting adult hippocampal neurogenesis, via the modulation of the phosphodiesterase-4 (PDE4)/cyclic adenosine monophosphate (cAMP)/cAMP response-element binding (CREB)/brain-derived neurotrophic factor (BDNF)/tropomyosin-related kinase B (TrkB) pathway. Collectively, our findings could promote the prevalence of kucha as a common beverage with uses for health care and contribute to the development of theacrine as a potential novel antidepressant medicine.
Collapse
Affiliation(s)
- Shu-Hua Ouyang
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou 510632, China
| | - Yu-Jia Zhai
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
- Department of Pharmacy, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Yan-Ping Wu
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou 510632, China
| | - Guo Xie
- Zhongshan Institute, University of Electronic Science and Technology of China, Zhongshan 528402, China
| | - Guo-En Wang
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Zhong-Fu Mao
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Hui-Hua Hu
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Xue-Hua Luo
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Wan-Yang Sun
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Lei Liang
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Wen-Jun Duan
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Hiroshi Kurihara
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Yi-Fang Li
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Rong-Rong He
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| |
Collapse
|
25
|
Pourzinal D, Yang JHJ, Bakker A, McMahon KL, Byrne GJ, Pontone GM, Mari Z, Dissanayaka NN. Hippocampal correlates of episodic memory in Parkinson's disease: A systematic review of magnetic resonance imaging studies. J Neurosci Res 2021; 99:2097-2116. [PMID: 34075634 DOI: 10.1002/jnr.24863] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 05/09/2021] [Accepted: 05/11/2021] [Indexed: 12/15/2022]
Abstract
The present review asks whether magnetic resonance imaging (MRI) studies are able to define neural correlates of episodic memory within the hippocampus in Parkinson's disease (PD). Systematic searches were performed in PubMed, Web of Science, Medline, CINAHL, and EMBASE using search terms related to structural and functional MRI (fMRI), the hippocampus, episodic memory, and PD. Risk of bias was assessed for each study using the Newtown-Ottawa Scale. Thirty-nine studies met inclusion criteria; eight fMRI, seven diffusion MRI (dMRI), and 24 structural MRI (14 exploring whole hippocampus and 10 exploring hippocampal subfields). Critical analysis of the literature revealed mixed evidence from functional and dMRI, but stronger evidence from sMRI of the hippocampus as a biomarker for episodic memory impairment in PD. Hippocampal subfield studies most often implicated CA1, CA3/4, and subiculum volume in episodic memory and cognitive decline in PD. Despite differences in imaging methodology, study design, and sample characteristics, MRI studies have helped elucidate an important neural correlate of episodic memory impairment in PD with both clinical and theoretical implications. Natural progression of this work encourages future research on hippocampal subfield function as a potential biomarker of, or therapeutic target for, episodic memory dysfunction in PD.
Collapse
Affiliation(s)
- Dana Pourzinal
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Royal Brisbane & Women's Hospital, Brisbane, QLD, Australia
| | - Ji Hyun J Yang
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Royal Brisbane & Women's Hospital, Brisbane, QLD, Australia
| | - Arnold Bakker
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, USA.,Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Katie L McMahon
- School of Clinical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
| | - Gerard J Byrne
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Royal Brisbane & Women's Hospital, Brisbane, QLD, Australia.,Mental Health Service, Royal Brisbane & Women's Hospital, Brisbane, QLD, Australia
| | - Gregory M Pontone
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, USA.,Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Zoltan Mari
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| | - Nadeeka N Dissanayaka
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Royal Brisbane & Women's Hospital, Brisbane, QLD, Australia.,Department of Neurology, Royal Brisbane & Women's Hospital, Brisbane, QLD, Australia.,School of Psychology, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
26
|
Skałbania J, Pałasz A, Błaszczyk I, Suszka-Świtek A, Krzystanek M, Tulcanaz KP, Worthington JJ, Mordecka-Chamera K. Chlorpromazine affects the numbers of Sox-2, Musashi1 and DCX-expressing cells in the rat brain subventricular zone. Pharmacol Rep 2021; 73:1164-1169. [PMID: 33843023 PMCID: PMC8413197 DOI: 10.1007/s43440-021-00259-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 03/28/2021] [Accepted: 03/31/2021] [Indexed: 12/05/2022]
Abstract
Background Adult neurogenesis observed both in the subventricular zone (SVZ) and hippocampus may be regulated and modulated by several endogenous factors, xenobiotics and medications. Classical and atypical antipsychotic drugs are able to affect neuronal and glial cell proliferation in the rat brain. The main purpose of this structural study was to determine whether chronic chlorpromazine treatment affects adult neurogenesis in the canonical sites of the rat brain. At present, the clinical application of chlorpromazine is rather limited; however, it may still represent an important model in basic neuropharmacological and toxicological studies. Methods The number of neural progenitors and immature neurons was enumerated using immunofluorescent detection of Sox2, Musashi1 and doublecortin (DCX) expression within SVZ. Results Chlorpromazine has a depressive effect on the early phase of adult neurogenesis in the rat subventricular zone (SVZ), as the mean number of Sox-2 immunoexpressing cells decreased following treatment. Conclusion Collectively, these results may suggest that long-term treatment with chlorpromazine may decrease neurogenic stem/progenitor cell formation in the rat SVZ and may affect rostral migratory stream formation. Supplementary Information The online version contains supplementary material available at 10.1007/s43440-021-00259-7.
Collapse
Affiliation(s)
- Jakub Skałbania
- Department of Histology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, ul. Medyków 18, 40-752, Katowice, Poland
| | - Artur Pałasz
- Department of Histology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, ul. Medyków 18, 40-752, Katowice, Poland.
| | - Iwona Błaszczyk
- Department of Histology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, ul. Medyków 18, 40-752, Katowice, Poland
| | - Aleksandra Suszka-Świtek
- Department of Histology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, ul. Medyków 18, 40-752, Katowice, Poland
| | - Marek Krzystanek
- Clinic of Psychiatric Rehabilitation, Department of Psychiatry and Psychotherapy, Faculty of Medical Sciences in Katowice, Medical University of Silesia, ul. Ziołowa 45/47, 40-635, Katowice, Poland
| | - Karina Paola Tulcanaz
- Faculty of Medicine, Pontifical Catholic University of Ecuador, Av. 12 de Octubre 1076, 170143, Quito, Ecuador
| | - John J Worthington
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, LA1 4YQ, UK
| | - Kinga Mordecka-Chamera
- Department of Histology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, ul. Medyków 18, 40-752, Katowice, Poland
| |
Collapse
|
27
|
Chen MX, Cheng S, Lei L, Zhang XF, Liu Q, Lin A, Wallis CU, Lukowicz MJ, Sham PC, Li Q, Ao LJ. The effects of maternal SSRI exposure on the serotonin system, prefrontal protein expression and behavioral development in male and female offspring rats. Neurochem Int 2021; 146:105041. [PMID: 33836218 DOI: 10.1016/j.neuint.2021.105041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 03/21/2021] [Accepted: 03/31/2021] [Indexed: 11/24/2022]
Abstract
Fluoxetine (FLX), a commonly used selective serotonin reuptake inhibitor, is often used to treat depression during pregnancy. However, prenatal exposure to FLX has been associated with a series of neuropsychiatric illnesses. The use of a rodent model can provide a clear indication as to whether prenatal exposure to SSRIs, independent of maternal psychiatric disorders or genetic syndromes, can cause long-term behavioral abnormalities in offspring. Thus, the present study aimed to explore whether prenatal FLX exposure causes long-term neurobehavioral effects, and identify the underlying mechanism between FLX and abnormal behaviors. In our study, 12/mg/kg/day of FLX or equal normal saline (NS) was administered to pregnant Sprague-Dawley (SD) rats (FLX = 30, NS = 27) on gestation day 11 till birth. We assessed the physical development and behavior of offspring, and in vivo magnetic resonance spectroscopy (MRS) was conducted to quantify biochemical alterations in the prefrontal cortex (PFC). Ex vivo measurements of brain serotonin level and a proteomic analysis were also undertaken. Our results showed that the offspring (male offspring in particular) of fluoxetine exposed mothers showed delayed physical development, increased anxiety-like behavior, and impaired social interaction. Moreover, down-regulation of 5-HT and SERT expression were identified in the PFC. We also found that prenatal FLX exposure significantly decreased NAA/tCr with 1H-MRS in the PFC of offspring. Finally, a proteomic study revealed sex-dependent differential protein expression. These findings may have translational importance suggesting that using SSRI medication alone in pregnant mothers may result in developmental delay in their offspring. Our results also help guide the choice of outcome measures in identifying of molecular and developmental mechanisms.
Collapse
Affiliation(s)
- Mo Xian Chen
- School of Rehabilitation, Kunming Medical University, Kunming, China
| | - Shu Cheng
- Department of Rehabilitation, China Resources & WISCO General Hospital, Wuhan, China
| | - Lei Lei
- Rehabilitation Medicine Department, The Affiliated Hospital of Southwest Medical University, Tai Ping Road, Luzhou, Sichuan, China
| | - Xiao Fan Zhang
- Department of Psychiatry, Tongji Hospital of Huazhong University of Science and Technology (HUST), China
| | - Qiang Liu
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Aijin Lin
- School of Rehabilitation, Kunming Medical University, Kunming, China
| | | | | | - Pak C Sham
- Department of Psychiatry, The University of Hong Kong, Hong Kong, SAR, China; State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, SAR, China; Centre for Genomic Sciences, The University of Hong Kong, Hong Kong, SAR, China
| | - Qi Li
- Department of Psychiatry, The University of Hong Kong, Hong Kong, SAR, China; State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, SAR, China.
| | - Li Juan Ao
- School of Rehabilitation, Kunming Medical University, Kunming, China.
| |
Collapse
|
28
|
Jin B, Alam M, Tierno A, Zhong H, Roy RR, Gerasimenko Y, Lu DC, Edgerton VR. Serotonergic Facilitation of Forelimb Functional Recovery in Rats with Cervical Spinal Cord Injury. Neurotherapeutics 2021; 18:1226-1243. [PMID: 33420588 PMCID: PMC8423890 DOI: 10.1007/s13311-020-00974-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2020] [Indexed: 10/22/2022] Open
Abstract
Serotonergic agents can improve the recovery of motor ability after a spinal cord injury. Herein, we compare the effects of buspirone, a 5-HT1A receptor partial agonist, to fluoxetine, a selective serotonin reuptake inhibitor, on forelimb motor function recovery after a C4 bilateral dorsal funiculi crush in adult female rats. After injury, single pellet reaching performance and forelimb muscle activity decreased in all rats. From 1 to 6 weeks after injury, rats were tested on these tasks with and without buspirone (1-2 mg/kg) or fluoxetine (1-5 mg/kg). Reaching and grasping success rates of buspirone-treated rats improved rapidly within 2 weeks after injury and plateaued over the next 4 weeks of testing. Electromyography (EMG) from selected muscles in the dominant forelimb showed that buspirone-treated animals used new reaching strategies to achieve success after the injury. However, forelimb performance dramatically decreased within 2 weeks of buspirone withdrawal. In contrast, fluoxetine treatment resulted in a more progressive rate of improvement in forelimb performance over 8 weeks after injury. Neither buspirone nor fluoxetine significantly improved quadrupedal locomotion on the horizontal ladder test. The improved accuracy of reaching and grasping, patterns of muscle activity, and increased excitability of spinal motor-evoked potentials after buspirone administration reflect extensive reorganization of connectivity within and between supraspinal and spinal sensory-motor netxcopy works. Thus, both serotonergic drugs, buspirone and fluoxetine, neuromodulated these networks to physiological states that enabled markedly improved forelimb function after cervical spinal cord injury.
Collapse
Affiliation(s)
- Benita Jin
- Department of Integrative Biology and Physiology, University of California, Los Angeles, 610 Charles E. Young Drive, Los Angeles, CA, 90095-1527, USA
| | - Monzurul Alam
- Department of Integrative Biology and Physiology, University of California, Los Angeles, 610 Charles E. Young Drive, Los Angeles, CA, 90095-1527, USA
| | - Alexa Tierno
- Department of Integrative Biology and Physiology, University of California, Los Angeles, 610 Charles E. Young Drive, Los Angeles, CA, 90095-1527, USA
| | - Hui Zhong
- Department of Integrative Biology and Physiology, University of California, Los Angeles, 610 Charles E. Young Drive, Los Angeles, CA, 90095-1527, USA
| | - Roland R Roy
- Department of Integrative Biology and Physiology, University of California, Los Angeles, 610 Charles E. Young Drive, Los Angeles, CA, 90095-1527, USA
- Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Yury Gerasimenko
- Department of Integrative Biology and Physiology, University of California, Los Angeles, 610 Charles E. Young Drive, Los Angeles, CA, 90095-1527, USA
- Pavlov Institute of Physiology, St. Petersburg, 199034, Russia
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, 420006, Russia
| | - Daniel C Lu
- Department of Neurosurgery, University of California, Los Angeles, CA, 90095, USA
| | - V Reggie Edgerton
- Department of Integrative Biology and Physiology, University of California, Los Angeles, 610 Charles E. Young Drive, Los Angeles, CA, 90095-1527, USA.
- Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Neurosurgery, University of California, Los Angeles, CA, 90095, USA.
- Department of Neurobiology, University of California, Los Angeles, CA, 90095, USA.
- Faculty of Science, The Centre for Neuroscience and Regenerative Medicine, University of Technology Sydney, Ultimo, NSW, Australia.
- Institut Guttmann, Hospital de Neurorehabilitació, Institut Universitari adscript a la Universitat Autònoma de Barcelona, 08916, Badalona, Spain.
| |
Collapse
|
29
|
Abstract
The adult brain is the result of a multistages complex neurodevelopmental process involving genetic, molecular and microenvironmental factors as well as diverse patterns of electrical activity. In the postnatal life, immature neuronal circuits undergo an experience-dependent maturation during critical periods of plasticity, but the brain still retains plasticity during adult life. In all these stages, the neurotransmitter GABA plays a pivotal role. In this chapter, we will describe the interaction of 5-HT with GABA in regulating neurodevelopment and plasticity.
Collapse
|
30
|
Chao FL, Zhang Y, Zhang L, Jiang L, Zhou CN, Tang J, Liang X, Fan JH, Dou XY, Tang Y. Fluoxetine Promotes Hippocampal Oligodendrocyte Maturation and Delays Learning and Memory Decline in APP/PS1 Mice. Front Aging Neurosci 2021; 12:627362. [PMID: 33519426 PMCID: PMC7838348 DOI: 10.3389/fnagi.2020.627362] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022] Open
Abstract
Oligodendrogenesis dysfunction impairs memory consolidation in adult mice, and an oligodendrocyte abnormality is an important change occurring in Alzheimer's disease (AD). While fluoxetine (FLX) is known to delay memory decline in AD models, its effects on hippocampal oligodendrogenesis are unclear. Here, we subjected 8-month-old male amyloid precursor protein (APP)/presenilin 1 (PS1) mice to the FLX intervention for 2 months. Their exploratory behaviors and general activities in a novel environment, spatial learning and memory and working and reference memory were assessed using the open-field test, Morris water maze, and Y maze. Furthermore, changes in hippocampal oligodendrogenesis were investigated using stereology, immunohistochemistry, immunofluorescence staining, and Western blotting techniques. FLX delayed declines in the spatial learning and memory, as well as the working and reference memory of APP/PS1 mice. In addition, APP/PS1 mice exhibited immature hippocampal oligodendrogenesis, and FLX increased the numbers of 2'3'cyclic nucleotide 3'-phosphodiesterase (CNPase)+ and newborn CNPase+ oligodendrocytes in the hippocampi of APP/PS1 mice. Moreover, FLX increased the density of SRY-related HMG-box 10 protein (SOX10)+ cells and reduced the percentage of oligodendrocyte lineage cells displaying the senescence phenotype (CDKN2A/p16INK4a) in the hippocampus of APP/PS1 mice. Moreover, FLX had no effect on the serotonin (5-HT) 1A receptor (5-HT1AR) content or number of 5-HT1AR+ oligodendrocytes, but it reduced the content and activity of glycogen synthase kinase 3β (GSK3β) in the hippocampus of APP/PS1 transgenic mice. Taken together, FLX delays the senescence of oligodendrocyte lineage cells and promotes oligodendrocyte maturation in the hippocampus of APP/PS1 mice. FLX may regulate GSK3β through a mechanism other than 5-HT1AR and then inhibit the negative effect of GSK3β on oligodendrocyte maturation in the hippocampus of an AD mouse model.
Collapse
Affiliation(s)
- Feng-lei Chao
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, China
- Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, China
| | - Yi Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lei Zhang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, China
- Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, China
| | - Lin Jiang
- Experimental Teaching Management Center, Chongqing Medical University, Chongqing, China
| | - Chun-ni Zhou
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, China
- Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, China
| | - Jing Tang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, China
- Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, China
| | - Xin Liang
- Department of Physiology, Chongqing Medical University, Chongqing, China
| | - Jin-hua Fan
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, China
- Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, China
| | - Xiao-yun Dou
- Academy of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Yong Tang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, China
- Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, China
| |
Collapse
|
31
|
Martins-Macedo J, Lepore AC, Domingues HS, Salgado AJ, Gomes ED, Pinto L. Glial restricted precursor cells in central nervous system disorders: Current applications and future perspectives. Glia 2020; 69:513-531. [PMID: 33052610 DOI: 10.1002/glia.23922] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 10/01/2020] [Accepted: 10/02/2020] [Indexed: 12/27/2022]
Abstract
The crosstalk between glial cells and neurons represents an exceptional feature for maintaining the normal function of the central nervous system (CNS). Increasing evidence has revealed the importance of glial progenitor cells in adult neurogenesis, reestablishment of cellular pools, neuroregeneration, and axonal (re)myelination. Several types of glial progenitors have been described, as well as their potentialities for recovering the CNS from certain traumas or pathologies. Among these precursors, glial-restricted precursor cells (GRPs) are considered the earliest glial progenitors and exhibit tripotency for both Type I/II astrocytes and oligodendrocytes. GRPs have been derived from embryos and embryonic stem cells in animal models and have maintained their capacity for self-renewal. Despite the relatively limited knowledge regarding the isolation, characterization, and function of these progenitors, GRPs are promising candidates for transplantation therapy and reestablishment/repair of CNS functions in neurodegenerative and neuropsychiatric disorders, as well as in traumatic injuries. Herein, we review the definition, isolation, characterization and potentialities of GRPs as cell-based therapies in different neurological conditions. We briefly discuss the implications of using GRPs in CNS regenerative medicine and their possible application in a clinical setting. MAIN POINTS: GRPs are progenitors present in the CNS with differentiation potential restricted to the glial lineage. These cells have been employed in the treatment of a myriad of neurodegenerative and traumatic pathologies, accompanied by promising results, herein reviewed.
Collapse
Affiliation(s)
- Joana Martins-Macedo
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Angelo C Lepore
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Helena S Domingues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Eduardo D Gomes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Luísa Pinto
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
32
|
Bruin WB, Taylor L, Thomas RM, Shock JP, Zhutovsky P, Abe Y, Alonso P, Ameis SH, Anticevic A, Arnold PD, Assogna F, Benedetti F, Beucke JC, Boedhoe PSW, Bollettini I, Bose A, Brem S, Brennan BP, Buitelaar JK, Calvo R, Cheng Y, Cho KIK, Dallaspezia S, Denys D, Ely BA, Feusner JD, Fitzgerald KD, Fouche JP, Fridgeirsson EA, Gruner P, Gürsel DA, Hauser TU, Hirano Y, Hoexter MQ, Hu H, Huyser C, Ivanov I, James A, Jaspers-Fayer F, Kathmann N, Kaufmann C, Koch K, Kuno M, Kvale G, Kwon JS, Liu Y, Lochner C, Lázaro L, Marques P, Marsh R, Martínez-Zalacaín I, Mataix-Cols D, Menchón JM, Minuzzi L, Moreira PS, Morer A, Morgado P, Nakagawa A, Nakamae T, Nakao T, Narayanaswamy JC, Nurmi EL, O'Neill J, Pariente JC, Perriello C, Piacentini J, Piras F, Piras F, Reddy YCJ, Rus-Oswald OG, Sakai Y, Sato JR, Schmaal L, Shimizu E, Simpson HB, Soreni N, Soriano-Mas C, Spalletta G, Stern ER, Stevens MC, Stewart SE, Szeszko PR, Tolin DF, Venkatasubramanian G, Wang Z, Yun JY, van Rooij D, Thompson PM, van den Heuvel OA, Stein DJ, van Wingen GA. Structural neuroimaging biomarkers for obsessive-compulsive disorder in the ENIGMA-OCD consortium: medication matters. Transl Psychiatry 2020; 10:342. [PMID: 33033241 PMCID: PMC7598942 DOI: 10.1038/s41398-020-01013-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 09/09/2020] [Accepted: 09/14/2020] [Indexed: 11/08/2022] Open
Abstract
No diagnostic biomarkers are available for obsessive-compulsive disorder (OCD). Here, we aimed to identify magnetic resonance imaging (MRI) biomarkers for OCD, using 46 data sets with 2304 OCD patients and 2068 healthy controls from the ENIGMA consortium. We performed machine learning analysis of regional measures of cortical thickness, surface area and subcortical volume and tested classification performance using cross-validation. Classification performance for OCD vs. controls using the complete sample with different classifiers and cross-validation strategies was poor. When models were validated on data from other sites, model performance did not exceed chance-level. In contrast, fair classification performance was achieved when patients were grouped according to their medication status. These results indicate that medication use is associated with substantial differences in brain anatomy that are widely distributed, and indicate that clinical heterogeneity contributes to the poor performance of structural MRI as a disease marker.
Collapse
Affiliation(s)
- Willem B Bruin
- Amsterdam UMC, University of Amsterdam, Department of Psychiatry, Amsterdam Neuroscience, Amsterdam, Netherlands.
| | - Luke Taylor
- Department of Physiology, Anatomy and Genetics, Oxford, UK
| | - Rajat M Thomas
- Amsterdam UMC, University of Amsterdam, Department of Psychiatry, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Jonathan P Shock
- Department of mathematics and applied mathematics, University of Cape Town, Cape Town, South Africa
| | - Paul Zhutovsky
- Amsterdam UMC, University of Amsterdam, Department of Psychiatry, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Yoshinari Abe
- Department of Psychiatry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Pino Alonso
- Department of Psychiatry, Bellvitge University Hospital, Bellvitge Biomedical Research Institute-IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
- Centro de Investigación Biomèdica en Red de Salud Mental-CIBERSAM, Barcelona, Spain
- Department of Clinical Sciences, University of Barcelona, Barcelona, Spain
| | - Stephanie H Ameis
- The Margaret and Wallace McCain Centre for Child, Youth and Family Mental Health, Campbell Family Mental Health Research Institute, The Centre for Addiction and Mental Health, Department of Psychiatry, Faculty of Medicine, University of Toronto, Toronto, Canada
- Centre for Brain and Mental Health, The Hospital for Sick Children, Toronto, Canada
| | - Alan Anticevic
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Paul D Arnold
- Mathison Centre for Mental Health Research and Education, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 1N4, Canada
| | - Francesca Assogna
- Laboratory of Neuropsychiatry, Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Francesco Benedetti
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Jan C Beucke
- Department of Psychology, Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Clinical Neuroscience, Centre for Psychiatric Research and Education, Karolinska Institutet, Stockholm, Sweden
| | - Premika S W Boedhoe
- Amsterdam UMC, Vrije Universteit Amsterdam, Department of Psychiatry, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Anatomy and Neurosciences, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Irene Bollettini
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Anushree Bose
- Obsessive-Compulsive Disorder (OCD) Clinic Department of Psychiatry National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Silvia Brem
- Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric Hospital, University of Zurich, Zurich, Switzerland
- University of Zurich and ETH Zurich, Neuroscience Center Zurich, Zurich, Switzerland
| | - Brian P Brennan
- McLean Hospital, Harvard Medical School, Belmont, MA, 02115, USA
| | - Jan K Buitelaar
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, Netherlands
- Karakter Child and Adolescent Psychiatry University Center, Nijmegen, The Netherlands
| | - Rosa Calvo
- Department of Child and Adolescent Psychiatry and Psychology, Institute of Neurosciences, Hospital Clínic Universitari, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en red de Salud Mental (CIBERSAM), Barcelona, Spain
| | - Yuqi Cheng
- Department of Psychiatry, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Kang Ik K Cho
- Institute of Human Behavioral Medicine, SNU-MRC, Seoul, Republic of Korea
| | - Sara Dallaspezia
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Damiaan Denys
- Amsterdam UMC, University of Amsterdam, Department of Psychiatry, Amsterdam Neuroscience, Amsterdam, Netherlands
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Benjamin A Ely
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jamie D Feusner
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, 94612, USA
| | - Kate D Fitzgerald
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jean-Paul Fouche
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| | - Egill A Fridgeirsson
- Amsterdam UMC, University of Amsterdam, Department of Psychiatry, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Patricia Gruner
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Deniz A Gürsel
- Department of Neuroradiology, Klinikum rechts der Isar, Technische Universität München, München, Germany
- TUM-Neuroimaging Center (TUM-NIC) of Klinikum rechts der Isar, Technische Universität München, München, Germany
| | - Tobias U Hauser
- Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric Hospital, University of Zurich, Zurich, Switzerland
- Max Planck UCL Centre for Computational Psychiatry and Ageing Research, London, UK
- Wellcome Centre for Human Neuroimaging, University College London, London, UK
| | - Yoshiyuki Hirano
- Research Center for Child Mental Development, Chiba University, Chiba, Japan
| | - Marcelo Q Hoexter
- Departamento e Instituto de Psiquiatria do Hospital das Clinicas, IPQ HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brasil
| | - Hao Hu
- Shanghai Mental Health Center Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chaim Huyser
- De Bascule, Academic Center for Child and Adolescent Psychiatry, Amsterdam, The Netherlands
- Department of child and adolescent psychiatry Amsterdam UMC, Amsterdam, The Netherlands
| | - Iliyan Ivanov
- Division of Child and Adolescent Psychiatry, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Anthony James
- Department of Psychiatry, , Oxford University, Oxford, UK
| | | | - Norbert Kathmann
- Department of Psychology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Christian Kaufmann
- Department of Psychology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Kathrin Koch
- Department of Neuroradiology, Klinikum rechts der Isar, Technische Universität München, München, Germany
- TUM-Neuroimaging Center (TUM-NIC) of Klinikum rechts der Isar, Technische Universität München, München, Germany
| | - Masaru Kuno
- Research Center for Child Mental Development, Chiba University, Chiba, Japan
| | - Gerd Kvale
- Bergen Center for Brain Plasticity, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Psychology, University of Bergen, Bergen, Norway
| | - Jun Soo Kwon
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Brain and Cognitive Sciences, Seoul National University College of Natural Sciences, Seoul, Korea
| | - Yanni Liu
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Christine Lochner
- SAMRC Unit on Risk and Resilience in Mental Disorders, Department of Psychiatry, Stellenbosch University, Stellenbosch, South Africa
| | - Luisa Lázaro
- Department of Child and Adolescent Psychiatry and Psychology, Institute of Neurosciences, Hospital Clínic Universitari, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en red de Salud Mental (CIBERSAM), Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Paulo Marques
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Clinical Academic Center-Braga, Braga, Portugal
| | - Rachel Marsh
- Columbia University Irving Medical Center, Columbia University, New York, NY, 10027, USA
- The Division of Child and Adolescent Psychiatry, New York State Psychiatric Institute, Columbia University, New York, NY, 10027, USA
| | - Ignacio Martínez-Zalacaín
- Department of Psychiatry, Bellvitge University Hospital, Bellvitge Biomedical Research Institute-IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
- Department of Clinical Sciences, University of Barcelona, Barcelona, Spain
| | - David Mataix-Cols
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet, Stockholm, Sweden
| | - José M Menchón
- Department of Psychiatry, Bellvitge University Hospital, Bellvitge Biomedical Research Institute-IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
- Centro de Investigación Biomèdica en Red de Salud Mental-CIBERSAM, Barcelona, Spain
- Department of Clinical Sciences, University of Barcelona, Barcelona, Spain
| | - Luciano Minuzzi
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, L8S 4L8, Canada
| | - Pedro S Moreira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Clinical Academic Center-Braga, Braga, Portugal
| | - Astrid Morer
- Department of Child and Adolescent Psychiatry and Psychology, Institute of Neurosciences, Hospital Clínic Universitari, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en red de Salud Mental (CIBERSAM), Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Pedro Morgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Clinical Academic Center-Braga, Braga, Portugal
| | - Akiko Nakagawa
- Research Center for Child Mental Development, Chiba University, Chiba, Japan
| | - Takashi Nakamae
- Department of Psychiatry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tomohiro Nakao
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Janardhanan C Narayanaswamy
- Obsessive-Compulsive Disorder (OCD) Clinic Department of Psychiatry National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Erika L Nurmi
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, 94612, USA
| | - Joseph O'Neill
- Division of Child and Adolescent Psychiatry, Jane and Terry Semel Institute For Neurosciences, University of California, Los Angeles, CA, 94612, USA
| | - Jose C Pariente
- Magnetic Resonance Image Core Facility, IDIBAPS (Institut d'Investigacions Biomèdiques August Pi i Sunyer), Barcelona, Spain
| | - Chris Perriello
- McLean Hospital, Harvard Medical School, Belmont, MA, 02115, USA
- University of Illinois at Urbana-Champaign, Champaign, IL, 61820, USA
| | - John Piacentini
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, 94612, USA
| | - Fabrizio Piras
- Laboratory of Neuropsychiatry, Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Federica Piras
- Laboratory of Neuropsychiatry, Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Y C Janardhan Reddy
- Obsessive-Compulsive Disorder (OCD) Clinic Department of Psychiatry National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Oana G Rus-Oswald
- University of Zürich, University Hospital Zürich, Dept. Neuroradiology, Zürich, Switzerland
- University Department of Geriatric Medicine Felix Platter, Basel, Switzerland
| | - Yuki Sakai
- Department of Psychiatry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
- ATR Brain Information Communication Research Laboratory Group, Kyoto, Japan
| | - João R Sato
- Center of Mathematics, Computing and Cognition, Universidade Federal do ABC, Santo Andre, Brazil
| | - Lianne Schmaal
- Orygen, Parkville, VIC, Australia
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Eiji Shimizu
- Research Center for Child Mental Development, Chiba University, Chiba, Japan
- Department of Cognitive Behavioral Physiology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - H Blair Simpson
- Columbia University Irving Medical Center, Columbia University, New York, NY, 10027, USA
- Center for OCD and Related Disorders, New York State Psychiatric Institute, New York, NY, 10032, USA
| | - Noam Soreni
- Pediatric OCD Consultation service, Anxiety Treatment and Research Center, St. Joseph's HealthCare, Hamilton, ON, L9C 0E3, Canada
- Offord Child Center, McMaster University, Hamilton, ON, L8S 4L8, Canada
| | - Carles Soriano-Mas
- Department of Psychiatry, Bellvitge University Hospital, Bellvitge Biomedical Research Institute-IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
- Centro de Investigación Biomèdica en Red de Salud Mental-CIBERSAM, Barcelona, Spain
- Department of Psychobiology and Methodology of Health Sciences, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Gianfranco Spalletta
- Laboratory of Neuropsychiatry, Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, Rome, Italy
- Beth K. and Stuart C. Yudofsky Division of Neuropsychiatry, Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Emily R Stern
- Department of Psychiatry, New York University Langone School of Medicine, New York, NY, 10016, USA
- Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, 10962, USA
| | - Michael C Stevens
- Olin Neuropsychiatry Research Center, Hartford Hospital, Hartford, CT, 06106, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, 06510, USA
| | - S Evelyn Stewart
- University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
- British Columbia Children's Hospital, Vancouver, BC, V6H 3N1, Canada
- British Columbia Mental Health and Addictions Research Institute, Vancouver, BC, V6H 3N1, Canada
| | - Philip R Szeszko
- Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- James J. Peters VA Medical Center, Bronx, New York, NY, 10468, USA
| | - David F Tolin
- Institute of Living/Hartford Hospital, Hartford, CT, 06119, USA
- Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Ganesan Venkatasubramanian
- Obsessive-Compulsive Disorder (OCD) Clinic Department of Psychiatry National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Zhen Wang
- Shanghai Mental Health Center Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai, China
| | - Je-Yeon Yun
- Seoul National University Hospital, Seoul, Republic of Korea
- Yeongeon Student Support Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Daan van Rooij
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Donders Centre for Cognitive Neuroimaging, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Paul M Thompson
- Imaging Genetics Center, Stevens Institute for Neuroimaging and Informatics, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90007, USA
| | - Odile A van den Heuvel
- Amsterdam UMC, Vrije Universteit Amsterdam, Department of Psychiatry, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Anatomy and Neurosciences, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Dan J Stein
- SAMRC Unit on Risk and Resilience in Mental Disorders, Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Guido A van Wingen
- Amsterdam UMC, University of Amsterdam, Department of Psychiatry, Amsterdam Neuroscience, Amsterdam, Netherlands.
| |
Collapse
|
33
|
Schönfeld LM, Schäble S, Zech MP, Kalenscher T. 5-HT 1A receptor agonism in the basolateral amygdala increases mutual-reward choices in rats. Sci Rep 2020; 10:16622. [PMID: 33024202 PMCID: PMC7538979 DOI: 10.1038/s41598-020-73829-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 09/23/2020] [Indexed: 12/19/2022] Open
Abstract
Rats show mutual-reward preferences, i.e., they prefer options that result in a reward for both themselves and a conspecific partner to options that result in a reward for themselves, but not for the partner. In a previous study, we have shown that lesions of the basolateral amygdala (BLA) reduced choices for mutual rewards. Here, we aimed to explore the role of 5-HT1A receptors within the BLA in mutual-reward choices. Rats received daily injections of either 50 or 25 ng of the 5-HT1A receptor agonist 8-OH-DPAT or a vehicle solution into the BLA and mutual-reward choices were measured in a rodent prosocial choice task. Compared to vehicle injections, 8-OH-DPAT significantly increased mutual-reward choices when a conspecific was present. By contrast, mutual-reward choices were significantly reduced by 8-OH-DPAT injections in the presence of a toy rat. The effect of 8-OH-DPAT injections was statistically significant during the expression, but not during learning of mutual-reward behavior, although an influence of 8-OH-DPAT injections on learning could not be excluded. There were no differences between 8-OH-DPAT-treated and vehicle-treated rats in general reward learning, behavioral flexibility, locomotion or anxiety. In this study, we have shown that repeated injections of the 5-HT1A receptor agonist 8-OH-DPAT have the potential to increase mutual-reward choices in a social setting without affecting other behavioral parameters.
Collapse
Affiliation(s)
- Lisa-Maria Schönfeld
- Comparative Psychology, Institute of Experimental Psychology, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany.
| | - Sandra Schäble
- Comparative Psychology, Institute of Experimental Psychology, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Maurice-Philipp Zech
- Comparative Psychology, Institute of Experimental Psychology, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Tobias Kalenscher
- Comparative Psychology, Institute of Experimental Psychology, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| |
Collapse
|
34
|
Brimson JM, Brimson S, Chomchoei C, Tencomnao T. Using sigma-ligands as part of a multi-receptor approach to target diseases of the brain. Expert Opin Ther Targets 2020; 24:1009-1028. [PMID: 32746649 DOI: 10.1080/14728222.2020.1805435] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION The sigma receptors are found abundantly in the central nervous system and are targets for the treatment of various diseases, including Alzheimer's (AD), Parkinson's (PD), Huntington's disease (HD), depression, amyotrophic lateral sclerosis (ALS), and multiple sclerosis (MS). However, for many of these diseases, other receptors and targets have been the focus of the most, such as acetylcholine esterase inhibitors in Alzheimer's and dopamine replacement in Parkinson's. The currently available drugs for these diseases have limited success resulting in the requirement of an alternative approach to their treatment. AREAS COVERED In this review, we discuss the potential role of the sigma receptors and their ligands as part of a multi receptor approach in the treatment of the diseases mentioned above. The literature reviewed was obtained through searches in databases, including PubMed, Web of Science, Google Scholar, and Scopus. EXPERT OPINION Given sigma receptor agonists provide neuroprotection along with other benefits such as potentiating the effects of other receptors, further development of multi-receptor targeting ligands, and or the development of multi-drug combinations to target multiple receptors may prove beneficial in the future treatment of degenerative diseases of the CNS, especially when coupled with better diagnostic techniques.
Collapse
Affiliation(s)
- James Michael Brimson
- Age-related Inflammation and Degeneration Research Unit, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University , Bangkok, Thailand
| | - Sirikalaya Brimson
- Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University , Bangkok, Thailand
| | - Chanichon Chomchoei
- Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University , Bangkok, Thailand
| | - Tewin Tencomnao
- Age-related Inflammation and Degeneration Research Unit, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University , Bangkok, Thailand
| |
Collapse
|
35
|
Ceccarelli M, D’Andrea G, Micheli L, Tirone F. Interaction Between Neurogenic Stimuli and the Gene Network Controlling the Activation of Stem Cells of the Adult Neurogenic Niches, in Physiological and Pathological Conditions. Front Cell Dev Biol 2020; 8:211. [PMID: 32318568 PMCID: PMC7154047 DOI: 10.3389/fcell.2020.00211] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 03/11/2020] [Indexed: 12/26/2022] Open
Abstract
In the adult mammalian brain new neurons are continuously generated throughout life in two niches, the dentate gyrus of the hippocampus and the subventricular zone. This process, called adult neurogenesis, starts from stem cells, which are activated and enter the cell cycle. The proliferative capability of stem cells progressively decreases during aging. The population of stem cells is generally quiescent, and it is not clear whether the potential for stem cells to expand is limited, or whether they can expand and then return to quiescence, remaining available for further activation. Certain conditions may deregulate stem cells quiescence and self-renewal. In fact we discuss the possibility of activation of stem cells by neurogenic stimuli as a function of the intensity of the stimulus (i.e., whether this is physiological or pathological), and of the deregulation of the system (i.e., whether the model is aged or carrying genetic mutations in the gene network controlling quiescence). It appears that when the system is aged and/or carrying mutations of quiescence-maintaining genes, preservation of the quiescent state of stem cells is more critical and stem cells can be activated by a neurogenic stimulus which is ineffective in normal conditions. Moreover, when a neurogenic stimulus is in itself a cause of brain damage (e.g., kainic acid treatment) the activation of stem cells occurs bypassing any inhibitory control. Plausibly, with strong neurogenic stimuli, such as kainic acid injected into the dentate gyrus, the self-renewal capacity of stem cells may undergo rapid exhaustion. However, the self-renewal capability of stem cells persists when normal stimuli are elicited in the presence of a mutation of one of the quiescence-maintaining genes, such as p16Ink4a, p21Cip1 or Btg1. In this case, stem cells become promptly activated by a neurogenic stimulus even during aging. This indicates that stem cells retain a high proliferative capability and plasticity, and suggests that stem cells are protected against the response to stimulus and are resilient to exhaustion. It will be interesting to assess at which functional degree of deregulation of the quiescence-maintaining system, stem cells will remain responsive to repeated neurogenic stimuli without undergoing exhaustion of their pool.
Collapse
Affiliation(s)
| | | | | | - Felice Tirone
- Institute of Biochemistry and Cell Biology, National Research Council (IBBC-CNR), Rome, Italy
| |
Collapse
|
36
|
Chen F, Danladi J, Wegener G, Madsen TM, Nyengaard JR. Sustained Ultrastructural Changes in Rat Hippocampal Formation After Repeated Electroconvulsive Seizures. Int J Neuropsychopharmacol 2020; 23:446-458. [PMID: 32215561 PMCID: PMC7387769 DOI: 10.1093/ijnp/pyaa021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 03/03/2020] [Accepted: 03/20/2020] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Electroconvulsive therapy (ECT) is a highly effective and fast-acting treatment for depression used in the clinic. Its mechanism of therapeutic action remains uncertain. Previous studies have focused on documenting neuroplasticity in the early phase following electroconvulsive seizures (ECS), an animal model of ECT. Here, we investigate whether changes in synaptic plasticity and nonneuronal plasticity (vascular and mitochondria) are sustained 3 months after repeated ECS trials. METHODS ECS or sham treatment was given daily for 1 day or 10 days to a genetic animal model of depression: the Flinders Sensitive and Resistant Line rats. Stereological principles were employed to quantify numbers of synapses and mitochondria as well as length of microvessels in the hippocampus 24 hours after a single ECS. Three months after 10 ECS treatments (1 per day for 10 days) and sham-treatment, brain-derived neurotrophic factor and vascular endothelial growth factor protein levels were quantified with immunohistochemistry. RESULTS A single ECS treatment significantly increased the volume of hippocampal CA1-stratum radiatum, the total length of microvessels, mitochondria number, and synapse number. Observed changes were sustained as shown in the multiple ECS treatment group analyzed 3 months after the last of 10 ECS treatments. CONCLUSION A single ECS caused rapid effects of synaptic plasticity and nonneuronal plasticity, while repeated ECS induced long-lasting changes in the efficacy of synaptic plasticity and nonneuronal plasticity at least up to 3 months after ECS.
Collapse
Affiliation(s)
- Fenghua Chen
- Core Center for Molecular Morphology, Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark,Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark,Correspondence: Fenghua Chen, MD, PhD, Department of Clinical Medicine - Translational Neuropsychiatry Unit, Nørrebrogade 44, Building 2B, 8000 Aarhus C, Denmark ()
| | - Jibrin Danladi
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Gregers Wegener
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark,Center of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa,AUGUST Centre, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Torsten M Madsen
- Core Center for Molecular Morphology, Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jens R Nyengaard
- Core Center for Molecular Morphology, Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark,Centre for Stochastic Geometry and Advanced Bioimaging, Aarhus University, Aarhus, Denmark
| |
Collapse
|
37
|
Mohammed A, El-Bakly WM, Ali A, El-Demerdash E. Rosuvastatin improves olanzapine's effects on behavioral impairment and hippocampal, hepatic and metabolic damages in isolated reared male rats. Behav Brain Res 2020; 378:112305. [PMID: 31634496 DOI: 10.1016/j.bbr.2019.112305] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 09/29/2019] [Accepted: 10/14/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIM Schizophrenia is a chronic, disabling neurological illness. This study investigated the effect of rosuvastatin (RSU) addition to the antipsychotic drug: olanzapine (OLZ) in treatment of post-weaning isolation rearing (IR) damaging effect and assessed behavioral impairment, metabolic and hepatic abnormalities, oxidative stress, and inflammatory markers. METHODS Treatment with OLZ (6 mg/kg, P.O.) and/or RSU (10 mg/kg, I.P.) have been started 6 weeks after isolation. We assessed behavioral tests, serum cortisol level, and hippocampal content of neurotransmitters. In addition, we assessed histopathology, inflammatory and oxidative stress markers of hippocampus, liver and adipose tissue RESULTS: Treatment of IR animals with OLZ, and/or RSU significantly counteracted the changes in hippocampus, liver and adipose tissue induced by post-weaning IR. Co-treatment of IR rats with both OLZ and RSU showed additive effects in some areas like improving both tumor necrosis factor alpha (TNFα) in both hippocampus and liver, histopathology of liver, oxidative stress markers of adipose tissue, β3 adrenergic receptors (ADRβ3), serum cortisol and total cholesterol. In addition, RSU alone alleviated the damage of IR rats by the same efficacy as OLZ with more benefit in cognition and exploration. CONCLUSION post-weaning IR as a model has behavioral, hippocampal, hepatic and marked metabolic changes more relevant to schizophrenia than drug-induced models. These effects were ameliorated by RSU and/or OLZ that are explained by their antioxidant, anti-inflammatory, anti-stress and anti-hyperlipidemic properties. Interestingly, co-treatment with both drugs showed a better effect.
Collapse
Affiliation(s)
- Aya Mohammed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Egypt
| | - Wesam M El-Bakly
- Department of Pharmacology, Faculty of Medicine, Ain Shams University, Egypt
| | - Azza Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Egypt
| | - Ebtehal El-Demerdash
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Egypt.
| |
Collapse
|
38
|
Pardo JV, Sheikh SA, Schwindt G, Lee JT, Adson DE, Rittberg B, Abuzzahab FS. A preliminary study of resting brain metabolism in treatment-resistant depression before and after treatment with olanzapine-fluoxetine combination. PLoS One 2020; 15:e0226486. [PMID: 31931515 PMCID: PMC6957341 DOI: 10.1371/journal.pone.0226486] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 11/26/2019] [Indexed: 12/28/2022] Open
Abstract
Treatment-resistant depression (TRD) occurs in many patients and causes high morbidity and mortality. Because TRD subjects are particularly difficult to study especially longitudinally, biological data remain very limited. In a preliminary study to judge feasibility and power, 25 TRD patients were referred from specialty psychiatric practices. All were severely and chronically depressed and mostly had comorbid psychiatric disorders as is typical in TRD. Nine patients were able to complete all required components of the protocol that included diagnostic interview; rating scales; clinical magnetic resonance imaging; medication washout; treatment with maximally tolerated olanzapine-fluoxetine combination for 8 weeks; and pre- and post-treatment fluorodeoxyglucose positron emission tomography. This drug combination is an accepted standard of treatment for TRD. Dropouts arose from worsening depression, insomnia, and anxiety. One patient remitted; three responded. A priori regions of interest included the amygdala and subgenual cingulate cortex (sgACC; Brodmann area BA25). Responders showed decreased metabolism with treatment in the right amygdala that correlated with clinical response; no significant changes in BA25; better response to treatment the higher the baseline BA25 metabolism; and decreased right ventromedial prefrontal metabolism (VMPFC; broader than BA25) with treatment which did not correlate with depression scores. The baseline metabolism of all individuals showed heterogeneous patterns when compared to a normative metabolic database. Although preliminary given the sample size, this study highlights several issues important for future work: marked dropout rate in this study design; need for large sample size for adequate power; baseline metabolic heterogeneity of TRD requiring careful subject characterization for future studies of interventions; relationship of amygdala activity decreases with response; and the relationship between baseline sgACC and VMPFC activity with response. Successful treatment of TRD with olanzapine-fluoxetine combination shows changes in cerebral metabolism like those seen in treatment-responsive major depression.
Collapse
Affiliation(s)
- José V. Pardo
- Cognitive Neuroimaging Unit, Mental Health PSL, Minneapolis VA Health Care System, Minneapolis, Minnesota, United States of America
- Department of Psychiatry, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Sohail A. Sheikh
- Cognitive Neuroimaging Unit, Mental Health PSL, Minneapolis VA Health Care System, Minneapolis, Minnesota, United States of America
- Department of Psychiatry, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Graeme Schwindt
- Cognitive Neuroimaging Unit, Mental Health PSL, Minneapolis VA Health Care System, Minneapolis, Minnesota, United States of America
| | - Joel T. Lee
- Cognitive Neuroimaging Unit, Mental Health PSL, Minneapolis VA Health Care System, Minneapolis, Minnesota, United States of America
| | - David E. Adson
- Department of Psychiatry, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Barry Rittberg
- Department of Psychiatry, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Faruk S. Abuzzahab
- Department of Psychiatry, University of Minnesota, Minneapolis, Minnesota, United States of America
| |
Collapse
|
39
|
Boda E. Myelin and oligodendrocyte lineage cell dysfunctions: New players in the etiology and treatment of depression and stress‐related disorders. Eur J Neurosci 2019; 53:281-297. [DOI: 10.1111/ejn.14621] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 10/06/2019] [Accepted: 11/12/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Enrica Boda
- Department of Neuroscience Rita Levi‐Montalcini University of Turin Turin Italy
- Neuroscience Institute Cavalieri Ottolenghi (NICO) University of Turin Turin Italy
| |
Collapse
|
40
|
Running exercise protects oligodendrocytes in the medial prefrontal cortex in chronic unpredictable stress rat model. Transl Psychiatry 2019; 9:322. [PMID: 31780641 PMCID: PMC6882819 DOI: 10.1038/s41398-019-0662-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 11/05/2019] [Accepted: 11/08/2019] [Indexed: 12/28/2022] Open
Abstract
Previous postmortem and animal studies have shown decreases in the prefrontal cortex (PFC) volume and the number of glial cells in the PFC of depression. Running exercise has been shown to alleviate depressive symptoms. However, the effects of running exercise on the medial prefrontal cortex (mPFC) volume and oligodendrocytes in the mPFC of depressed patients and animals have not been investigated. To address these issues, adult male rats were subjected to chronic unpredictable stress (CUS) for 5 weeks, followed by treadmill running for 6 weeks. Then, the mPFC volume and the mPFC oligodendrocytes were investigated using stereology, immunohistochemistry, immunofluorescence and western blotting. Using a CUS paradigm that allowed for the analysis of anhedonia, we found that running exercise alleviated the deficits in sucrose preference, as well as the decrease in the mPFC volume. Meanwhile, we found that running exercise significantly increased the number of CNPase+ oligodendrocytes and Olig2+ oligodendrocytes, reduced the ratio between Olig2+/NG2+ oligodendrocytes and Olig2+ oligodendrocytes and increased myelin basic protein (MBP), CNPase and Olig2 protein expression in the mPFC of the CUS rat model. However, running exercise did not change NG2+ oligodendrocyte number in the mPFC in these rats. These results indicated that running exercise promoted the differentiation of oligodendrocytes and myelin-forming ability in the mPFC in the context of depression. These findings suggest that the beneficial effects of running exercise on mPFC volume and oligodendrocytes in mPFC might be an important structural basis for the antidepressant effects of running exercise.
Collapse
|
41
|
Kim W, Hahn KR, Jung HY, Kwon HJ, Nam SM, Kim JW, Park JH, Yoo DY, Kim DW, Won MH, Yoon YS, Hwang IK. Melatonin ameliorates cuprizone-induced reduction of hippocampal neurogenesis, brain-derived neurotrophic factor, and phosphorylation of cyclic AMP response element-binding protein in the mouse dentate gyrus. Brain Behav 2019; 9:e01388. [PMID: 31429533 PMCID: PMC6749490 DOI: 10.1002/brb3.1388] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 07/01/2019] [Accepted: 07/28/2019] [Indexed: 01/03/2023] Open
Abstract
INTRODUCTION The aim of this study was to investigate the effects of cuprizone on adult hippocampal neurogenesis in naïve mice. Additionally, we also studied how melatonin affects the neuronal degeneration induced by cuprizone. METHODS Eight-week-old male C57BL/6J mice were randomly divided into three groups: (a) the control group, (b) the group treated with cuprizone only, and (c) the group treated with both cuprizone and melatonin. Cuprizone was administered with food at 0.2% ad libitum for 6 weeks. Melatonin was also administered with tap water at 6 g/L ad libitum for 6 weeks; the animals were then euthanized for immunohistochemistry with Ki67, doublecortin (DCX), glucose transporter 3 (GLUT3), and phosphorylation of cyclic adenosine monophosphate (AMP) response element binding (pCREB); double immunofluorescence of neuronal nuclei (NeuN) and myelin basic protein (MBP); and Western blot analysis of brain-derived neurotrophic factor (BDNF) expression to reveal the effects of cuprizone and melatonin on cell damage and hippocampal neurogenesis. RESULTS Administration of cuprizone significantly decreased the number of differentiating (DCX-positive) neuroblasts and proliferating (Ki67-positive) cells in the dentate gyrus. Moreover, cuprizone administration decreased glucose utilization (GLUT3-positive cells) and cell transcription (pCREB-positive cells and BDNF protein expression) in the dentate gyrus. Administration of melatonin ameliorated the cuprizone-induced reduction of differentiating neuroblasts and proliferating cells, glucose utilization, and cell transcription. CONCLUSION The results of the study suggest that cuprizone treatment disrupts hippocampal neurogenesis in the dentate gyrus by reducing BDNF levels and decreasing the phosphorylation of CREB. These effects were ameliorated by melatonin treatment.
Collapse
Affiliation(s)
- Woosuk Kim
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
| | - Kyu Ri Hahn
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
| | - Hyo Young Jung
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
| | - Hyun Jung Kwon
- Department of Biochemistry and Molecular Biology, College of Dentistry, Research Institute of Oral Sciences, Gangneung-Wonju National University, Gangneung, South Korea
| | - Sung Min Nam
- Department of Anatomy, College of Veterinary Medicine, Konkuk University, Seoul, South Korea
| | - Jong Whi Kim
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
| | - Joon Ha Park
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, Korea
| | - Dae Young Yoo
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan, South Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, College of Dentistry, Research Institute of Oral Sciences, Gangneung-Wonju National University, Gangneung, South Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Korea
| | - Yeo Sung Yoon
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
| |
Collapse
|
42
|
Fluoxetine-induced dematuration of hippocampal neurons and adult cortical neurogenesis in the common marmoset. Mol Brain 2019; 12:69. [PMID: 31383032 PMCID: PMC6683334 DOI: 10.1186/s13041-019-0489-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 07/24/2019] [Indexed: 12/22/2022] Open
Abstract
The selective serotonin reuptake inhibitor fluoxetine (FLX) is widely used to treat depression and anxiety disorders. Chronic FLX treatment reportedly induces cellular responses in the brain, including increased adult hippocampal and cortical neurogenesis and reversal of neuron maturation in the hippocampus, amygdala, and cortex. However, because most previous studies have used rodent models, it remains unclear whether these FLX-induced changes occur in the primate brain. To evaluate the effects of FLX in the primate brain, we used immunohistological methods to assess neurogenesis and the expression of neuronal maturity markers following chronic FLX treatment (3 mg/kg/day for 4 weeks) in adult marmosets (n = 3 per group). We found increased expression of doublecortin and calretinin, markers of immature neurons, in the hippocampal dentate gyrus of FLX-treated marmosets. Further, FLX treatment reduced parvalbumin expression and the number of neurons with perineuronal nets, which indicate mature fast-spiking interneurons, in the hippocampus, but not in the amygdala or cerebral cortex. We also found that FLX treatment increased the generation of cortical interneurons; however, significant up-regulation of adult hippocampal neurogenesis was not observed in FLX-treated marmosets. These results suggest that dematuration of hippocampal neurons and increased cortical neurogenesis may play roles in FLX-induced effects and/or side effects. Our results are consistent with those of previous studies showing hippocampal dematuration and increased cortical neurogenesis in FLX-treated rodents. In contrast, FLX did not affect hippocampal neurogenesis or dematuration of interneurons in the amygdala and cerebral cortex.
Collapse
|
43
|
Abstract
A theoretical framework is proposed to gain insight into the pathogenesis of major depressive disorder (MDD). Despite being a relatively weak argument, the neurogenesis theory is suggested to compensate for the limitations of the monoamine theory. In the adult hippocampus, neurogenesis is functionally related to regulation of the hypothalamic-pituitary-adrenal (HPA) axis, inflammatory processes, cognitive functions and other aspects that contribute to etiological factors that lead to MDD and promote recovery from MDD. Despite a lack of investigation into neurogenesis and antidepressant action, it is proposed that chronic administration of antidepressant(s) can induce the recruitment and integration of newborn neurons into the dentate gyrus and, ultimately, lead to the remission of MDD. The extant body of literature indicates that the suppression of neurogenesis per se may be associated with an impaired response to antidepressant treatment rather than with the induction of depressive-like behaviors. Moreover, recent studies have shown that increasing the survival rate and incorporation of new neurons can alleviate depressive-like behaviors and promote stress resilience. According to the neurogenic reserve hypothesis, hippocampal neurogenesis supports specific cortical functions, including executive functions, pattern separation and contextual information processing, control over the HPA axis and behavioral coping mechanisms in response to stressful situations. Therefore, hippocampal neurogenesis may be a promising biological indicator of stress resilience and antidepressant response in patients with MDD.
Collapse
Affiliation(s)
- Seon-Cheol Park
- Department of Psychiatry, Inje University Haeundae Paik Hospital, 875, Haeun-daero, Haeundae-gu, Busan, 48108, Republic of Korea.
| |
Collapse
|
44
|
Saylor RA, Hersey M, West A, Buchanan AM, Berger SN, Nijhout HF, Reed MC, Best J, Hashemi P. In vivo Hippocampal Serotonin Dynamics in Male and Female Mice: Determining Effects of Acute Escitalopram Using Fast Scan Cyclic Voltammetry. Front Neurosci 2019; 13:362. [PMID: 31110471 PMCID: PMC6499219 DOI: 10.3389/fnins.2019.00362] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 03/29/2019] [Indexed: 01/07/2023] Open
Abstract
Depression is a highly prevalent psychiatric disorder, impacting females at a rate roughly twice that of males. This disparity has become the focus of many studies which are working to determine if there are environmental or biological underpinnings to depression pathology. The biology of depression is not well understood, but experts agree that a key neurotransmitter of interest is serotonin. Most research on basic serotonin neurochemistry, by us and others, has predominantly focused on male models. Thus, it is now critical to include female models to decipher possible fundamental differences between the sexes that may underlie this disorder. In this paper, we seek to determine any such differences using fast-scan cyclic voltammetry (FSCV) and fast-scan controlled adsorption voltammetry. These techniques allow us to probe the serotonergic system via measurement of evoked and ambient serotonin at carbon fiber microelectrodes (CFMs). Our data reveal no statistical differences, in the hippocampus, in female serotonin chemistry during the different stages of the estrous cycle compared to the mean female response. Furthermore, no difference was observed in evoked serotonin release and reuptake, nor ambient extracellular serotonin levels between male and female mice. We applied a previously developed mathematical model that fits our serotonin signals as a function of several synaptic processes that control the extracellular levels of this transmitter. We used the model to study potential system differences between males and females. One hypothesis brought fourth, that female mice exhibit tighter autoreceptor control of serotonin, is validated via literature and methiothepin challenge. We postulate that this tight regulation may act as a control mechanism against changes in the serotonin signal mediated by estrogen spikes. Importantly, this safety mechanism has no consequence for acutely administered escitalopram’s (ESCIT’s) ability to increase extracellular serotonin between the sexes. This work demonstrates little fundamental differences in in vivo hippocampal serotonin between the sexes, bar control mechanisms in female mice that can be observed under extraneous circumstances. We thus highlight the importance of considering sex as a biological factor in determining pharmacodynamics for personalized medical treatments that involve targeting serotonin receptors.
Collapse
Affiliation(s)
- Rachel A Saylor
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, United States
| | - Melinda Hersey
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, United States.,Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Alyssa West
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, United States
| | - Anna Marie Buchanan
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, United States.,Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Shane N Berger
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, United States
| | | | - Michael C Reed
- Department of Mathematics, Duke University, Durham, NC, United States
| | - Janet Best
- Department of Mathematics, The Ohio State University, Columbus, OH, United States
| | - Parastoo Hashemi
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, United States
| |
Collapse
|
45
|
Song T, Wu H, Li R, Xu H, Rao X, Gao L, Zou Y, Lei H. Repeated fluoxetine treatment induces long-lasting neurotrophic changes in the medial prefrontal cortex of adult rats. Behav Brain Res 2019; 365:114-124. [PMID: 30849415 DOI: 10.1016/j.bbr.2019.03.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 03/02/2019] [Accepted: 03/04/2019] [Indexed: 01/23/2023]
Abstract
Fluoxetine (Flx), a selective serotonin reuptake inhibitor, is extensively used to treat mood and anxiety disorders. Previous animal studies have shown that early-life exposure to Flx results in long-lasting behavioral alterations and neuroplasticity in the hippocampus and cortex, which may persist into adulthood. It remains unclear whether repeated Flx treatment in normal adult animals can induce lasting neuroplasticity and behavioral alterations persisting long beyond the treatment period. In this study, young adult rats (about 9 weeks old) were treated with Flx (10 mg/kg body weight, twice daily) for 15 consecutive days, and the effects of Flx on medial prefrontal cortex (mPFC) neuroplasticity and mPFC-related behaviors were assessed 20 days after the last injection. It was observed that the mPFC of Flx-treated rats had significant increases in the number of 5-bromodeoxyuridine-positive (BrdU+) cells, dendritic complexity/spine density in layer II/III pyramidal neurons, and brain-derived neurotrophic factor (BDNF)/tropomyosin receptor kinase B (TrkB) expression levels, as well as a significant decrease in the number of parvalbumin-positive (PV+) interneurons. The Flx-treated rats exhibited higher motivation to explore new environments, evidenced by a significantly increased number of entries into the novel arm in the Y-maze test. However, they did not show any significant changes in the anhedonia and anxiety levels measured by sucrose preference and elevated plus maze tests respectively. In conclusion, repeated Flx treatment, with the paradigm used, induces long-lasting neuroplastic changes in the mPFC of normal adult rats; such changes and related behavioral manifestations may persist up to 20 days after the last dose.
Collapse
Affiliation(s)
- Tao Song
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430074, PR China; National Center for Magnetic Resonance in Wuhan, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, PR China
| | - Hao Wu
- Department of Radiology, Institute of Surgery Research, the Third Affiliated Hospital, Army Medical University, Chongqing, 400042, PR China
| | - Ronghui Li
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430074, PR China; National Center for Magnetic Resonance in Wuhan, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, PR China
| | - Hui Xu
- National Center for Magnetic Resonance in Wuhan, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, PR China
| | - Xiaoping Rao
- Center for Brain Science, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, PR China
| | - Lifeng Gao
- National Center for Magnetic Resonance in Wuhan, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, PR China; Department of Medical Imaging, School of Medicine, Jianghan University, Wuhan 430056, PR China
| | - Yijuan Zou
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430074, PR China; National Center for Magnetic Resonance in Wuhan, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, PR China
| | - Hao Lei
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430074, PR China; National Center for Magnetic Resonance in Wuhan, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, PR China.
| |
Collapse
|
46
|
Murata Y, Matsuda H, Mikami Y, Hirose S, Mori M, Ohe K, Mine K, Enjoji M. Chronic administration of quetiapine stimulates dorsal hippocampal proliferation and immature neurons of male rats, but does not reverse psychosocial stress-induced hyponeophagic behavior. Psychiatry Res 2019; 272:411-418. [PMID: 30611957 DOI: 10.1016/j.psychres.2018.12.137] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 12/25/2018] [Accepted: 12/26/2018] [Indexed: 12/14/2022]
Abstract
Quetiapine, an atypical antipsychotic, has been used for the treatment of several neuropsychiatric disorders. However, the underlying mechanism of the broad therapeutic range of quetiapine remains unknown. We previously reported that several aversive conditions affect dorsal/ventral hippocampal neurogenesis differentially. This study was aimed to elucidate the positive effects of chronic treatment with quetiapine on regional differences in hippocampal proliferation and immature neurons and behavioral changes under psychosocial stress using the Resident-Intruder paradigm. Twenty-three male Sprague-Dawley rats were intraperitoneally administered a vehicle or quetiapine (10 mg/kg) once daily for 28 days. Two weeks after starting the injections, animals were exposed to intermittent social defeat (four times over two weeks). The behavioral effects of stress and quetiapine were evaluated by the Novelty-Suppressed Feeding (NSF) test. The stereological quantification of hippocampal neurogenesis was estimated using immunostaining with Ki-67 and doublecortin (DCX). Chronic quetiapine treatment stimulated the Ki-67- and DCX-positive cells in the dorsal hippocampus, but not in the ventral subregion. The stress-induced changes in neurogenesis and hyponeophagic behavior were not reversed by repeated administration of quetiapine. Future study with additional behavioral tests is needed to elucidate the functional significance of the quetiapine-induced increase in dorsal hippocampal neurogenesis.
Collapse
Affiliation(s)
- Yusuke Murata
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1, Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| | - Hiroko Matsuda
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1, Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Yoko Mikami
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1, Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Shiori Hirose
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1, Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Masayoshi Mori
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1, Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Kenji Ohe
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1, Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Kazunori Mine
- Faculty of Neurology and Psychiatry, Mito Hospital, 4-1-1, Shime-Higashi, Shime-Machi, Kasuya-Gun, Fukuoka 811-2243, Japan
| | - Munechika Enjoji
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1, Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| |
Collapse
|
47
|
Gao Y, Yao Y, Liang X, Tang J, Ma J, Qi YQ, Huang CX, Zhang Y, Chen LM, Chao FL, Zhang L, Luo YM, Xiao Q, Du L, Xiao Q, Wang SR, Tang Y. Changes in white matter and the effects of fluoxetine on such changes in the CUS rat model of depression. Neurosci Lett 2019; 694:104-110. [DOI: 10.1016/j.neulet.2018.11.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 11/02/2018] [Accepted: 11/07/2018] [Indexed: 01/05/2023]
|
48
|
Cannabinoid signalling in embryonic and adult neurogenesis: possible implications for psychiatric and neurological disorders. Acta Neuropsychiatr 2019; 31:1-16. [PMID: 29764526 DOI: 10.1017/neu.2018.11] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cannabinoid signalling modulates several aspects of brain function, including the generation and survival of neurons during embryonic and adult periods. The present review intended to summarise evidence supporting a role for the endocannabinoid system on the control of neurogenesis and neurogenesis-dependent functions. Studies reporting participation of cannabinoids on the regulation of any step of neurogenesis and the effects of cannabinoid compounds on animal models possessing neurogenesis-dependent features were selected from Medline. Qualitative evaluation of the selected studies indicated that activation of cannabinoid receptors may change neurogenesis in embryonic or adult nervous systems alongside rescue of phenotypes in animal models of different psychiatric and neurological disorders. The text offers an overview on the effects of cannabinoids on central nervous system development and the possible links with psychiatric and neurological disorders such as anxiety, depression, schizophrenia, brain ischaemia/stroke and Alzheimer's disease. An understanding of the mechanisms by which cannabinoid signalling influences developmental and adult neurogenesis will help foster the development of new therapeutic strategies for neurodevelopmental, psychiatric and neurological disorders.
Collapse
|
49
|
Abstract
Adult neurogenesis continues to captivate the curiosity of the scientific community; and researchers seem to have a particular interest in identifying the functional implications of such plasticity. While the majority of research focuses on the association between adult neurogenesis and learning and memory (including spatial learning associated with hippocampal neurogenesis and olfactory discrimination associated with neurogenesis in the olfactory system), the following review will explore the link to motivated behaviors. In particular, goal-directed behaviors such as sociosexual, parental, aggressive, as well as depression- and anxiety-like behaviors and their reciprocal association to adult neurogenesis will be evaluated. The review will detail research in humans and other mammalian species. Furthermore, the potential mechanisms underlying these neurogenic alterations will be highlighted. Lastly, the review will conclude with a discussion on the functional significance of these newly generated cells in mediating goal-directed behaviors.
Collapse
Affiliation(s)
- Claudia Jorgensen
- Behavioral Science Department, Utah Valley University, Orem, Utah, USA
| |
Collapse
|
50
|
Famitafreshi H, Karimian M. Modulation of catalase, copper and zinc in the hippocampus and the prefrontal cortex in social isolation-induced depression in male rats. Acta Neurobiol Exp (Wars) 2019. [DOI: 10.21307/ane-2019-016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|