1
|
Palmisano A, Pandit S, Smeralda CL, Demchenko I, Rossi S, Battelli L, Rivolta D, Bhat V, Santarnecchi E. The Pathophysiological Underpinnings of Gamma-Band Alterations in Psychiatric Disorders. Life (Basel) 2024; 14:578. [PMID: 38792599 PMCID: PMC11122172 DOI: 10.3390/life14050578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/04/2024] [Accepted: 04/06/2024] [Indexed: 05/26/2024] Open
Abstract
Investigating the biophysiological substrates of psychiatric illnesses is of great interest to our understanding of disorders' etiology, the identification of reliable biomarkers, and potential new therapeutic avenues. Schizophrenia represents a consolidated model of γ alterations arising from the aberrant activity of parvalbumin-positive GABAergic interneurons, whose dysfunction is associated with perineuronal net impairment and neuroinflammation. This model of pathogenesis is supported by molecular, cellular, and functional evidence. Proof for alterations of γ oscillations and their underlying mechanisms has also been reported in bipolar disorder and represents an emerging topic for major depressive disorder. Although evidence from animal models needs to be further elucidated in humans, the pathophysiology of γ-band alteration represents a common denominator for different neuropsychiatric disorders. The purpose of this narrative review is to outline a framework of converging results in psychiatric conditions characterized by γ abnormality, from neurochemical dysfunction to alterations in brain rhythms.
Collapse
Affiliation(s)
- Annalisa Palmisano
- Chair of Lifespan Developmental Neuroscience, Faculty of Psychology, TUD Dresden University of Technology, 01069 Dresden, Germany
- Precision Neuroscience and Neuromodulation Program, Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA (E.S.)
- Department of Education, Psychology, and Communication, University of Bari Aldo Moro, 70121 Bari, Italy;
| | - Siddhartha Pandit
- Precision Neuroscience and Neuromodulation Program, Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA (E.S.)
| | - Carmelo L. Smeralda
- Precision Neuroscience and Neuromodulation Program, Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA (E.S.)
- Siena Brain Investigation and Neuromodulation (SI-BIN) Laboratory, Department of Medicine, Surgery and Neuroscience, Neurology and Clinical Neurophysiology Section, University of Siena, 53100 Siena, Italy;
| | - Ilya Demchenko
- Interventional Psychiatry Program, St. Michael’s Hospital—Unity Health Toronto, Toronto, ON M5B 1W8, Canada; (I.D.)
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Simone Rossi
- Siena Brain Investigation and Neuromodulation (SI-BIN) Laboratory, Department of Medicine, Surgery and Neuroscience, Neurology and Clinical Neurophysiology Section, University of Siena, 53100 Siena, Italy;
| | - Lorella Battelli
- Berenson-Allen Center for Noninvasive Brain Stimulation, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Center for Neuroscience and Cognitive Systems@UniTn, Istituto Italiano di Tecnologia, 38068 Rovereto, Italy
| | - Davide Rivolta
- Department of Education, Psychology, and Communication, University of Bari Aldo Moro, 70121 Bari, Italy;
| | - Venkat Bhat
- Interventional Psychiatry Program, St. Michael’s Hospital—Unity Health Toronto, Toronto, ON M5B 1W8, Canada; (I.D.)
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Emiliano Santarnecchi
- Precision Neuroscience and Neuromodulation Program, Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA (E.S.)
- Department of Neurology and Radiology, Massachusetts General Hospital, Boston, MA 02114, USA
| |
Collapse
|
2
|
Rakotobe M, Fjerdingstad N, Ruiz-Reig N, Lamonerie T, D'Autréaux F. Central role of the habenulo-interpeduncular system in the neurodevelopmental basis of susceptibility and resilience to anxiety in mice. Neurobiol Dis 2024; 191:106392. [PMID: 38145853 DOI: 10.1016/j.nbd.2023.106392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 12/27/2023] Open
Abstract
Having experienced stress during sensitive periods of brain development strongly influences how individuals cope with later stress. Some are prone to develop anxiety or depression, while others appear resilient. The as-yet-unknown mechanisms underlying these differences may lie in how genes and environmental stress interact to shape the circuits that control emotions. Here, we investigated the role of the habenulo-interpeduncular system (HIPS), a critical node in reward circuits, in early stress-induced anxiety in mice. We found that habenular and IPN components characterized by the expression of Otx2 are synaptically connected and particularly sensitive to chronic stress (CS) during the peripubertal period. Stress-induced peripubertal activation of this HIPS subcircuit elicits both HIPS hypersensitivity to later stress and susceptibility to develop anxiety. We also show that HIPS silencing through conditional Otx2 knockout counteracts these effects of stress. Together, these results demonstrate that a genetic factor, Otx2, and stress interact during the peripubertal period to shape the stress sensitivity of the HIPS, which is shown to be a key modulator of susceptibility or resilience to develop anxiety.
Collapse
Affiliation(s)
- Malalaniaina Rakotobe
- Université Côte d'Azur, CNRS, Inserm, iBV, Institut de Biologie Valrose, 06108 Nice, France
| | - Niels Fjerdingstad
- Université Côte d'Azur, CNRS, Inserm, iBV, Institut de Biologie Valrose, 06108 Nice, France
| | - Nuria Ruiz-Reig
- Université Côte d'Azur, CNRS, Inserm, iBV, Institut de Biologie Valrose, 06108 Nice, France
| | - Thomas Lamonerie
- Université Côte d'Azur, CNRS, Inserm, iBV, Institut de Biologie Valrose, 06108 Nice, France.
| | - Fabien D'Autréaux
- Université Côte d'Azur, CNRS, Inserm, iBV, Institut de Biologie Valrose, 06108 Nice, France. Fabien.D'
| |
Collapse
|
3
|
Larsen B, Sydnor VJ, Keller AS, Yeo BTT, Satterthwaite TD. A critical period plasticity framework for the sensorimotor-association axis of cortical neurodevelopment. Trends Neurosci 2023; 46:847-862. [PMID: 37643932 PMCID: PMC10530452 DOI: 10.1016/j.tins.2023.07.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/23/2023] [Accepted: 07/25/2023] [Indexed: 08/31/2023]
Abstract
To understand human brain development it is necessary to describe not only the spatiotemporal patterns of neurodevelopment but also the neurobiological mechanisms that underlie them. Human neuroimaging studies have provided evidence for a hierarchical sensorimotor-to-association (S-A) axis of cortical neurodevelopment. Understanding the biological mechanisms that underlie this program of development using traditional neuroimaging approaches has been challenging. Animal models have been used to identify periods of enhanced experience-dependent plasticity - 'critical periods' - that progress along cortical hierarchies and are governed by a conserved set of neurobiological mechanisms that promote and then restrict plasticity. In this review we hypothesize that the S-A axis of cortical development in humans is partly driven by the cascading maturation of critical period plasticity mechanisms. We then describe how recent advances in in vivo neuroimaging approaches provide a promising path toward testing this hypothesis by linking signals derived from non-invasive imaging to critical period mechanisms.
Collapse
Affiliation(s)
- Bart Larsen
- Penn Lifespan Informatics and Neuroimaging Center (PennLINC), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Penn-CHOP Lifespan Brain Institute, Perelman School of Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Valerie J Sydnor
- Penn Lifespan Informatics and Neuroimaging Center (PennLINC), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Penn-CHOP Lifespan Brain Institute, Perelman School of Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Arielle S Keller
- Penn Lifespan Informatics and Neuroimaging Center (PennLINC), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Penn-CHOP Lifespan Brain Institute, Perelman School of Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - B T Thomas Yeo
- Centre for Sleep and Cognition (CSC), and Centre for Translational Magnetic Resonance Research (TMR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Electrical and Computer Engineering, National University of Singapore, Singapore; N.1 Institute for Health and Institute for Digital Medicine (WisDM), National University of Singapore, Singapore; Integrative Sciences and Engineering Programme (ISEP), National University of Singapore, Singapore
| | - Theodore D Satterthwaite
- Penn Lifespan Informatics and Neuroimaging Center (PennLINC), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Penn-CHOP Lifespan Brain Institute, Perelman School of Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
4
|
Vincent C, Gilabert-Juan J, Gibel-Russo R, Alvarez-Fischer D, Krebs MO, Le Pen G, Prochiantz A, Di Nardo AA. Non-cell-autonomous OTX2 transcription factor regulates anxiety-related behavior in the mouse. Mol Psychiatry 2021; 26:6469-6480. [PMID: 33963285 PMCID: PMC8760049 DOI: 10.1038/s41380-021-01132-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/06/2021] [Accepted: 04/14/2021] [Indexed: 02/03/2023]
Abstract
The OTX2 homeoprotein transcription factor is expressed in the dopaminergic neurons of the ventral tegmental area, which projects to limbic structures controlling complex behaviors. OTX2 is also produced in choroid plexus epithelium, from which it is secreted into cerebrospinal fluid and transferred to limbic structure parvalbumin interneurons. Previously, adult male mice subjected to early-life stress were found susceptible to anxiety-like behaviors, with accompanying OTX2 expression changes in ventral tegmental area or choroid plexus. Here, we investigated the consequences of reduced OTX2 levels in Otx2 heterozygote mice, as well as in Otx2+/AA and scFvOtx2tg/0 mouse models for decreasing OTX2 transfer from choroid plexus to parvalbumin interneurons. Both male and female adult mice show anxiolysis-like phenotypes in all three models. In Otx2 heterozygote mice, we observed no changes in dopaminergic neuron numbers and morphology in ventral tegmental area, nor in their metabolic output and projections to target structures. However, we found reduced expression of parvalbumin in medial prefrontal cortex, which could be rescued in part by adult overexpression of Otx2 specifically in choroid plexus, resulting in increased anxiety-like behavior. Taken together, OTX2 synthesis by the choroid plexus followed by its secretion into the cerebrospinal fluid is an important regulator of anxiety-related phenotypes in the mouse.
Collapse
Affiliation(s)
- Clémentine Vincent
- Centre for Interdisciplinary Research in Biology (CIRB), CNRS UMR 7241, INSERM U1050, Labex MemoLife, PSL Research University, Collège de France, Paris, France
- Institut NeuroMyoGène, CNRS UMR 5310, INSERM U1217, Université Claude Bernard Lyon 1, Lyon, France
| | - Javier Gilabert-Juan
- Centre for Interdisciplinary Research in Biology (CIRB), CNRS UMR 7241, INSERM U1050, Labex MemoLife, PSL Research University, Collège de France, Paris, France
- Department of Anatomy, Histology and Neuroscience, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Rachel Gibel-Russo
- Centre for Interdisciplinary Research in Biology (CIRB), CNRS UMR 7241, INSERM U1050, Labex MemoLife, PSL Research University, Collège de France, Paris, France
| | | | - Marie-Odile Krebs
- Laboratoire de Physiopathologie des Maladies Psychiatriques, INSERM U1266, Institut de Psychiatrie et Neurosciences de Paris, Université de Paris, Paris, France
- Institut de Psychiatrie, CNRS GDR 3557, Paris, France
- Faculté de Médecine, Université de Paris, Pôle Hospitalo-Universitaire Evaluation Prévention et Innovation Thérapeutique, GHU Paris Psychiatrie et Neurosciences site Sainte-Anne, Paris, France
| | - Gwenaëlle Le Pen
- Laboratoire de Physiopathologie des Maladies Psychiatriques, INSERM U1266, Institut de Psychiatrie et Neurosciences de Paris, Université de Paris, Paris, France
| | - Alain Prochiantz
- Centre for Interdisciplinary Research in Biology (CIRB), CNRS UMR 7241, INSERM U1050, Labex MemoLife, PSL Research University, Collège de France, Paris, France.
| | - Ariel A Di Nardo
- Centre for Interdisciplinary Research in Biology (CIRB), CNRS UMR 7241, INSERM U1050, Labex MemoLife, PSL Research University, Collège de France, Paris, France.
| |
Collapse
|
5
|
Kamata S, Hashiyama R, Hana-Ika H, Ohkubo I, Saito R, Honda A, Anan Y, Akahoshi N, Noguchi K, Kanda Y, Ishii I. Cytotoxicity comparison of 35 developmental neurotoxicants in human induced pluripotent stem cells (iPSC), iPSC-derived neural progenitor cells, and transformed cell lines. Toxicol In Vitro 2020; 69:104999. [PMID: 32949729 DOI: 10.1016/j.tiv.2020.104999] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 08/05/2020] [Accepted: 09/09/2020] [Indexed: 11/27/2022]
Abstract
The Organization for Economic Co-operation and Development (OECD) test guideline 426 for developmental neurotoxicity (DNT) of industrial/environmental chemicals depends primarily on animal experimentation. This requirement raises various critical issues, such as high cost, long duration, the sacrifice of large numbers of animals, and interspecies differences. This study demonstrates an alternative protocol that is simple, quick, less expensive, and standardized to evaluate DNT of many chemicals using human induced pluripotent stem cells (iPSC) and their differentiation to neural progenitor cells (NPC). Initially, concentration-dependent cytotoxicity of 35 DNT chemicals, including industrial materials, insecticides, and clinical drugs, were compared among iPSC, NPC, and two transformed cells, Cos-7 and HepG2, using tetrazolium dye (MTS)-reducing colorimetric and ATP luciferase assays, and IC50 values were calculated. Next, inhibitory effects of the 14 representative chemicals (mainly insecticides) on iPSC differentiation to NPC were evaluated by measuring altered expression of neural differentiation and undifferentiation marker genes. Results show that both iPSC and NPC were much more sensitive to most DNT chemicals than the transformed cells, and 14 chemicals induced differential patterns of marker gene expression, highlighting the validity and utility of the protocol for evaluation and classification of DNT chemicals and preclinical DNT tests for safety assessment.
Collapse
Affiliation(s)
- Shotaro Kamata
- Department of Health Chemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Reina Hashiyama
- Department of Health Chemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Hiroto Hana-Ika
- Department of Health Chemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Issei Ohkubo
- Department of Health Chemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Ryota Saito
- Department of Health Chemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Akihiro Honda
- Department of Health Chemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Yasumi Anan
- Department of Health Chemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Noriyuki Akahoshi
- Department of Health Chemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Kohji Noguchi
- Laboratory of Molecular Target Therapy, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba 278-8510, Japan
| | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences, Kawasaki, Kanagawa 210-9501, Japan.
| | - Isao Ishii
- Department of Health Chemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan.
| |
Collapse
|
6
|
Di Nardo AA, Fuchs J, Joshi RL, Moya KL, Prochiantz A. The Physiology of Homeoprotein Transduction. Physiol Rev 2019; 98:1943-1982. [PMID: 30067157 DOI: 10.1152/physrev.00018.2017] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The homeoprotein family comprises ~300 transcription factors and was long seen as primarily involved in developmental programs through cell autonomous regulation. However, recent evidence reveals that many of these factors are also expressed in the adult where they exert physiological functions not yet fully deciphered. Furthermore, the DNA-binding domain of most homeoproteins contains two signal sequences allowing their secretion and internalization, thus intercellular transfer. This review focuses on this new-found signaling in cell migration, axon guidance, and cerebral cortex physiological homeostasis and speculates on how it may play important roles in early arealization of the neuroepithelium. It also describes the use of homeoproteins as therapeutic proteins in mouse models of diseases affecting the central nervous system, in particular Parkinson disease and glaucoma.
Collapse
Affiliation(s)
- Ariel A Di Nardo
- Centre for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR 7241, INSERM U1050, Labex MemoLife, PSL Research University , Paris , France
| | - Julia Fuchs
- Centre for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR 7241, INSERM U1050, Labex MemoLife, PSL Research University , Paris , France
| | - Rajiv L Joshi
- Centre for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR 7241, INSERM U1050, Labex MemoLife, PSL Research University , Paris , France
| | - Kenneth L Moya
- Centre for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR 7241, INSERM U1050, Labex MemoLife, PSL Research University , Paris , France
| | - Alain Prochiantz
- Centre for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR 7241, INSERM U1050, Labex MemoLife, PSL Research University , Paris , France
| |
Collapse
|
7
|
Developmental Requirement of Homeoprotein Otx2 for Specific Habenulo-Interpeduncular Subcircuits. J Neurosci 2018; 39:1005-1019. [PMID: 30593496 DOI: 10.1523/jneurosci.1818-18.2018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 12/06/2018] [Accepted: 12/16/2018] [Indexed: 01/11/2023] Open
Abstract
The habenulo-interpeduncular system (HIPS) is now recognized as a critical circuit modulating aversion, reward, and social behavior. There is evidence that dysfunction of this circuit leads to psychiatric disorders. Because psychiatric diseases may originate in developmental abnormalities, it is crucial to investigate the developmental mechanisms controlling the formation of the HIPS. Thus far, this issue has been the focus of limited studies. Here, we explored the developmental processes underlying the formation of the medial habenula (MHb) and its unique output, the interpeduncular nucleus (IPN), in mice independently of their gender. We report that the Otx2 homeobox gene is essential for the proper development of both structures. We show that MHb and IPN neurons require Otx2 at different developmental stages and, in both cases, Otx2 deletion leads to disruption of HIPS subcircuits. Finally, we show that Otx2+ neurons tend to be preferentially interconnected. This study reveals that synaptically connected components of the HIPS, despite radically different developmental strategies, share high sensitivity to Otx2 expression.SIGNIFICANCE STATEMENT Brain reward circuits are highly complex and still poorly understood. In particular, it is important to understand how these circuits form as many psychiatric diseases may arise from their abnormal development. This work shows that Otx2, a critical evolutionary conserved gene implicated in brain development and a predisposing factor for psychiatric diseases, is required for the formation of the habenulo-interpeduncular system (HIPS), an important component of the reward circuit. Otx2 deletion affects multiple processes such as proliferation and migration of HIPS neurons. Furthermore, neurons expressing Otx2 are preferentially interconnected. Therefore, Otx2 expression may represent a code that specifies the connectivity of functional subunits of the HIPS. Importantly, the Otx2 conditional knock-out animals used in this study might represent a new genetic model of psychiatric diseases.
Collapse
|
8
|
Testa D, Prochiantz A, Di Nardo AA. Perineuronal nets in brain physiology and disease. Semin Cell Dev Biol 2018; 89:125-135. [PMID: 30273653 DOI: 10.1016/j.semcdb.2018.09.011] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 08/24/2018] [Accepted: 09/27/2018] [Indexed: 12/31/2022]
Abstract
Perineuronal nets (PNNs) in the brain are condensed glycosaminoglycan-rich extracellular matrix structures with heterogeneous composition yet specific organization. They typically assemble around a subset of fast-spiking interneurons that are implicated in learning and memory. Owing to their unique structural organization, PNNs have neuroprotective capacities but also participate in signal transduction and in controlling neuronal activity and plasticity. In this review, we define PNN structure in detail and describe its various biochemical and physiological functions. We further discuss the role of PNNs in brain disorders such as schizophrenia, bipolar disorder, Alzheimer disease and addictions. Lastly, we describe therapeutic approaches that target PNNs to alter brain physiology and counter brain dysfunction.
Collapse
Affiliation(s)
- Damien Testa
- Centre for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR 7241, INSERM U1050, PSL University, Labex MemoLife, 75005 Paris, France
| | - Alain Prochiantz
- Centre for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR 7241, INSERM U1050, PSL University, Labex MemoLife, 75005 Paris, France
| | - Ariel A Di Nardo
- Centre for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR 7241, INSERM U1050, PSL University, Labex MemoLife, 75005 Paris, France.
| |
Collapse
|
9
|
Proteolytic Remodeling of Perineuronal Nets: Effects on Synaptic Plasticity and Neuronal Population Dynamics. Neural Plast 2018. [PMID: 29531525 PMCID: PMC5817213 DOI: 10.1155/2018/5735789] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The perineuronal net (PNN) represents a lattice-like structure that is prominently expressed along the soma and proximal dendrites of parvalbumin- (PV-) positive interneurons in varied brain regions including the cortex and hippocampus. It is thus apposed to sites at which PV neurons receive synaptic input. Emerging evidence suggests that changes in PNN integrity may affect glutamatergic input to PV interneurons, a population that is critical for the expression of synchronous neuronal population discharges that occur with gamma oscillations and sharp-wave ripples. The present review is focused on the composition of PNNs, posttranslation modulation of PNN components by sulfation and proteolysis, PNN alterations in disease, and potential effects of PNN remodeling on neuronal plasticity at the single-cell and population level.
Collapse
|
10
|
Sakai A, Nakato R, Ling Y, Hou X, Hara N, Iijima T, Yanagawa Y, Kuwano R, Okuda S, Shirahige K, Sugiyama S. Genome-Wide Target Analyses of Otx2 Homeoprotein in Postnatal Cortex. Front Neurosci 2017; 11:307. [PMID: 28620275 PMCID: PMC5450002 DOI: 10.3389/fnins.2017.00307] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 05/16/2017] [Indexed: 11/13/2022] Open
Abstract
Juvenile brain has a unique time window, or critical period, in which neuronal circuits are remodeled by experience. Mounting evidence indicates the importance of neuronal circuit rewiring in various neurodevelopmental disorders of human cognition. We previously showed that Otx2 homeoprotein, essential for brain formation, is recaptured during postnatal maturation of parvalbumin-positive interneurons (PV cells) to activate the critical period in mouse visual cortex. Cortical Otx2 is the only interneuron-enriched transcription factor known to regulate the critical period, but its downstream targets remain unknown. Here, we used ChIP-seq (chromatin immunoprecipitation sequencing) to identify genome-wide binding sites of Otx2 in juvenile mouse cortex, and interneuron-specific RNA-seq to explore the Otx2-dependent transcriptome. Otx2-bound genes were associated with human diseases such as schizophrenia as well as critical periods. Of these genes, expression of neuronal factors involved in transcription, signal transduction and mitochondrial function was moderately and broadly affected in Otx2-deficient interneurons. In contrast to reported binding sites in the embryo, genes encoding potassium ion transporters such as KV3.1 had juvenile cortex-specific binding sites, suggesting that Otx2 is involved in regulating fast-spiking properties during PV cell maturation. Moreover, transcripts of oxidative resistance-1 (Oxr1), whose promoter has Otx2 binding sites, were markedly downregulated in Otx2-deficient interneurons. Therefore, an important role of Otx2 may be to protect the cells from the increased oxidative stress in fast-spiking PV cells. Our results suggest that coordinated expression of Otx2 targets promotes PV cell maturation and maintains its function in neuronal plasticity and disease.
Collapse
Affiliation(s)
- Akiko Sakai
- Laboratory of Neuronal Development, Graduate School of Medical and Dental Sciences, Niigata UniversityNiigata, Japan
| | - Ryuichiro Nakato
- Research Center for Epigenetic Disease, Institute of Molecular and Cellular Biosciences, University of TokyoTokyo, Japan
| | - Yiwei Ling
- Bioinformatics Laboratory, Graduate School of Medical and Dental Sciences, Niigata UniversityNiigata, Japan
| | - Xubin Hou
- Laboratory of Neuronal Development, Graduate School of Medical and Dental Sciences, Niigata UniversityNiigata, Japan
| | - Norikazu Hara
- Department of Molecular Genetics, Center for Bioresources, Brain Research Institute, Niigata UniversityNiigata, Japan
| | - Tomoya Iijima
- Laboratory of Neuronal Development, Graduate School of Medical and Dental Sciences, Niigata UniversityNiigata, Japan
| | - Yuchio Yanagawa
- Department of Genetic and Behavioral Neuroscience, Graduate School of Medicine, Gunma UniversityGunma, Japan
| | - Ryozo Kuwano
- Department of Molecular Genetics, Center for Bioresources, Brain Research Institute, Niigata UniversityNiigata, Japan
| | - Shujiro Okuda
- Bioinformatics Laboratory, Graduate School of Medical and Dental Sciences, Niigata UniversityNiigata, Japan
| | - Katsuhiko Shirahige
- Research Center for Epigenetic Disease, Institute of Molecular and Cellular Biosciences, University of TokyoTokyo, Japan
| | - Sayaka Sugiyama
- Laboratory of Neuronal Development, Graduate School of Medical and Dental Sciences, Niigata UniversityNiigata, Japan
| |
Collapse
|
11
|
Genetic Otx2 mis-localization delays critical period plasticity across brain regions. Mol Psychiatry 2017; 22:680-688. [PMID: 28194008 PMCID: PMC5400722 DOI: 10.1038/mp.2017.1] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 11/21/2016] [Accepted: 12/21/2016] [Indexed: 12/15/2022]
Abstract
Accumulation of non-cell autonomous Otx2 homeoprotein in postnatal mouse visual cortex (V1) has been implicated in both the onset and closure of critical period (CP) plasticity. Here, we show that a genetic point mutation in the glycosaminoglycan recognition motif of Otx2 broadly delays the maturation of pivotal parvalbumin-positive (PV+) interneurons not only in V1 but also in the primary auditory (A1) and medial prefrontal cortex (mPFC). Consequently, not only visual, but also auditory plasticity is delayed, including the experience-dependent expansion of tonotopic maps in A1 and the acquisition of acoustic preferences in mPFC, which mitigates anxious behavior. In addition, Otx2 mis-localization leads to dynamic turnover of selected perineuronal net (PNN) components well beyond the normal CP in V1 and mPFC. These findings reveal widespread actions of Otx2 signaling in the postnatal cortex controlling the maturational trajectory across modalities. Disrupted PV+ network function and deficits in PNN integrity are implicated in a variety of psychiatric illnesses, suggesting a potential global role for Otx2 function in establishing mental health.
Collapse
|
12
|
Kaut O, Schmitt I, Hofmann A, Hoffmann P, Schlaepfer TE, Wüllner U, Hurlemann R. Aberrant NMDA receptor DNA methylation detected by epigenome-wide analysis of hippocampus and prefrontal cortex in major depression. Eur Arch Psychiatry Clin Neurosci 2015; 265:331-41. [PMID: 25571874 DOI: 10.1007/s00406-014-0572-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 12/19/2014] [Indexed: 12/21/2022]
Abstract
Current perspectives on the molecular underpinnings of major depressive disorder (MDD) posit a mechanistic role of epigenetic DNA modifications in mediating the interaction between environmental risk factors and a genetic predisposition. However, conclusive evidence for differential methylation signatures in the brain's epigenome of MDD patients as compared to controls is still lacking. To address this issue, we conducted a pilot study including an epigenome-wide methylation analysis in six individuals diagnosed with recurrent MDD and six control subjects matched for age and gender, with a priori focus on the hippocampus and prefrontal cortex as pathophysiologically relevant candidate regions. Our analysis revealed differential methylation profiles of 11 genes in hippocampus and 20 genes in prefrontal cortex, five of which were selected for replication of the methylation status using pyrosequencing. Among these replicated targets, GRIN2A was found to be hypermethylated in both prefrontal cortex and hippocampus. This finding may be of particular functional relevance as GRIN2A encodes the glutamatergic N-methyl-D-aspartate receptor subunit epsilon-1 (NR2A) and is known to be involved in a plethora of synaptic plasticity-related regulatory processes probably disturbed in MDD.
Collapse
Affiliation(s)
- Oliver Kaut
- Department of Neurology, University of Bonn, Sigmund-Freud-Str. 25, 53105, Bonn, Germany,
| | | | | | | | | | | | | |
Collapse
|
13
|
Li WH, Zhou L, Li Z, Wang Y, Shi JT, Yang YJ, Gui JF. Zebrafish Lbh-like Is Required for Otx2-mediated Photoreceptor Differentiation. Int J Biol Sci 2015; 11:688-700. [PMID: 25999792 PMCID: PMC4440259 DOI: 10.7150/ijbs.11244] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Accepted: 04/11/2015] [Indexed: 12/12/2022] Open
Abstract
The homeobox transcription factor orthodenticle homolog 2 (otx2) is supposed as an organizer that orchestrates a transcription factor network during photoreceptor development. However, its regulation in the process remains unclear. In this study, we have identified a zebrafish limb bud and heart-like gene (lbh-like), which is expressed initially at 30 hours post fertilization (hpf) in the developing brain and eyes. Lbh-like knockdown by morpholinos specifically inhibits expression of multiple photoreceptor-specific genes, such as opsins, gnat1, gnat2 and irbp. Interestingly, otx2 expression in the morphants is not significantly reduced until 32 hpf when lbh-like begins to express, but its expression level in 72 hpf morphants is higher than that in wild type embryos. Co-injection of otx2 and its downstream target neuroD mRNAs can rescue the faults in eyes of Lbh-like morphants. Combined with the results of promoter-reporter assay, we suggest that lbh-like is a new regulator of photoreceptor differentiation directly through affecting otx2 expression in zebrafish. Furthermore, knockdown of lbh-like increases the activity of Notch pathway and perturbs the balance among proliferation, differentiation and survival of photoreceptor precursors.
Collapse
Affiliation(s)
- Wen-Hua Li
- 1. State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, ; 2. Graduate University of the Chinese Academy of Sciences, Wuhan 430072, China
| | - Li Zhou
- 1. State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences
| | - Zhi Li
- 1. State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences
| | - Yang Wang
- 1. State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences
| | - Jian-Tao Shi
- 1. State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences
| | - Yan-Jing Yang
- 1. State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, ; 2. Graduate University of the Chinese Academy of Sciences, Wuhan 430072, China
| | - Jian-Fang Gui
- 1. State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences
| |
Collapse
|
14
|
Jukic MM, Carrillo-Roa T, Bar M, Becker G, Jovanovic VM, Zega K, Binder EB, Brodski C. Abnormal development of monoaminergic neurons is implicated in mood fluctuations and bipolar disorder. Neuropsychopharmacology 2015; 40:839-48. [PMID: 25241801 PMCID: PMC4330498 DOI: 10.1038/npp.2014.244] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 07/29/2014] [Accepted: 08/26/2014] [Indexed: 01/11/2023]
Abstract
Subtle mood fluctuations are normal emotional experiences, whereas drastic mood swings can be a manifestation of bipolar disorder (BPD). Despite their importance for normal and pathological behavior, the mechanisms underlying endogenous mood instability are largely unknown. During embryogenesis, the transcription factor Otx2 orchestrates the genetic networks directing the specification of dopaminergic (DA) and serotonergic (5-HT) neurons. Here we behaviorally phenotyped mouse mutants overexpressing Otx2 in the hindbrain, resulting in an increased number of DA neurons and a decreased number of 5-HT neurons in both developing and mature animals. Over the course of 1 month, control animals exhibited stable locomotor activity in their home cages, whereas mutants showed extended periods of elevated or decreased activity relative to their individual average. Additional behavioral paradigms, testing for manic- and depressive-like behavior, demonstrated that mutants showed an increase in intra-individual fluctuations in locomotor activity, habituation, risk-taking behavioral parameters, social interaction, and hedonic-like behavior. Olanzapine, lithium, and carbamazepine ameliorated the behavioral alterations of the mutants, as did the mixed serotonin receptor agonist quipazine and the specific 5-HT2C receptor agonist CP-809101. Testing the relevance of the genetic networks specifying monoaminergic neurons for BPD in humans, we applied an interval-based enrichment analysis tool for genome-wide association studies. We observed that the genes specifying DA and 5-HT neurons exhibit a significant level of aggregated association with BPD but not with schizophrenia or major depressive disorder. The results of our translational study suggest that aberrant development of monoaminergic neurons leads to mood fluctuations and may be associated with BPD.
Collapse
Affiliation(s)
- Marin M Jukic
- Department of Physiology and Cell Biology, Zlotowski Center for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Tania Carrillo-Roa
- The Department of Translational Research in Psychiatry, Max-Planck Institute of Psychiatry, Munich, Germany,Faculty of Biology, Ludwig-Maximilian University of Munich, Munich, Germany
| | - Michal Bar
- Department of Physiology and Cell Biology, Zlotowski Center for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Gal Becker
- Department of Physiology and Cell Biology, Zlotowski Center for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Vukasin M Jovanovic
- Department of Physiology and Cell Biology, Zlotowski Center for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Ksenija Zega
- Department of Physiology and Cell Biology, Zlotowski Center for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Elisabeth B Binder
- The Department of Translational Research in Psychiatry, Max-Planck Institute of Psychiatry, Munich, Germany
| | - Claude Brodski
- Department of Physiology and Cell Biology, Zlotowski Center for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel,Department of Physiology and Cell Biology, Zlotowski Center for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva 84105, Israel, Tel: +972 8647 7320, Fax: +972 8647 7627, E-mail:
| |
Collapse
|
15
|
Boeckx C, Benítez-Burraco A. The shape of the human language-ready brain. Front Psychol 2014; 5:282. [PMID: 24772099 PMCID: PMC3983487 DOI: 10.3389/fpsyg.2014.00282] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 03/17/2014] [Indexed: 12/14/2022] Open
Abstract
Our core hypothesis is that the emergence of our species-specific language-ready brain ought to be understood in light of the developmental changes expressed at the levels of brain morphology and neural connectivity that occurred in our species after the split from Neanderthals–Denisovans and that gave us a more globular braincase configuration. In addition to changes at the cortical level, we hypothesize that the anatomical shift that led to globularity also entailed significant changes at the subcortical level. We claim that the functional consequences of such changes must also be taken into account to gain a fuller understanding of our linguistic capacity. Here we focus on the thalamus, which we argue is central to language and human cognition, as it modulates fronto-parietal activity. With this new neurobiological perspective in place, we examine its possible molecular basis. We construct a candidate gene set whose members are involved in the development and connectivity of the thalamus, in the evolution of the human head, and are known to give rise to language-associated cognitive disorders. We submit that the new gene candidate set opens up new windows into our understanding of the genetic basis of our linguistic capacity. Thus, our hypothesis aims at generating new testing grounds concerning core aspects of language ontogeny and phylogeny.
Collapse
Affiliation(s)
- Cedric Boeckx
- Catalan Institute for Advanced Studies and Research (ICREA) Barcelona, Spain ; Department of Linguistics, Universitat de Barcelona Barcelona, Spain
| | | |
Collapse
|
16
|
Laufer BI, Mantha K, Kleiber ML, Diehl EJ, Addison SMF, Singh SM. Long-lasting alterations to DNA methylation and ncRNAs could underlie the effects of fetal alcohol exposure in mice. Dis Model Mech 2013; 6:977-92. [PMID: 23580197 PMCID: PMC3701217 DOI: 10.1242/dmm.010975] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Fetal alcohol spectrum disorders (FASDs) are characterized by life-long changes in gene expression, neurodevelopment and behavior. What mechanisms initiate and maintain these changes are not known, but current research suggests a role for alcohol-induced epigenetic changes. In this study we assessed alterations to adult mouse brain tissue by assaying DNA cytosine methylation and small noncoding RNA (ncRNA) expression, specifically the microRNA (miRNA) and small nucleolar RNA (snoRNA) subtypes. We found long-lasting alterations in DNA methylation as a result of fetal alcohol exposure, specifically in the imprinted regions of the genome harboring ncRNAs and sequences interacting with regulatory proteins. A large number of major nodes from the identified networks, such as Pten signaling, contained transcriptional repressor CTCF-binding sites in their promoters, illustrating the functional consequences of alcohol-induced changes to DNA methylation. Next, we assessed ncRNA expression using two independent array platforms and quantitative PCR. The results identified 34 genes that are targeted by the deregulated miRNAs. Of these, four (Pten, Nmnat1, Slitrk2 and Otx2) were viewed as being crucial in the context of FASDs given their roles in the brain. Furthermore, ∼20% of the altered ncRNAs mapped to three imprinted regions (Snrpn-Ube3a, Dlk1-Dio3 and Sfmbt2) that showed differential methylation and have been previously implicated in neurodevelopmental disorders. The findings of this study help to expand on the mechanisms behind the long-lasting changes in the brain transcriptome of FASD individuals. The observed changes could contribute to the initiation and maintenance of the long-lasting effect of alcohol.
Collapse
Affiliation(s)
- Benjamin I Laufer
- Molecular Genetics Unit, Department of Biology, Western University, London, Ontario, N6A 5B7, Canada
| | | | | | | | | | | |
Collapse
|
17
|
Kleiber ML, Laufer BI, Wright E, Diehl EJ, Singh SM. Long-term alterations to the brain transcriptome in a maternal voluntary consumption model of fetal alcohol spectrum disorders. Brain Res 2012; 1458:18-33. [PMID: 22560501 DOI: 10.1016/j.brainres.2012.04.016] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Revised: 03/29/2012] [Accepted: 04/09/2012] [Indexed: 12/16/2022]
Abstract
Many women continue to consume low to moderate quantities of alcohol during pregnancy, which can result in the variable neurobehavioural effects in the absence of physiological abnormalities that characterize fetal alcohol spectrum disorders (FASD). Previously, we reported that a mouse model for FASD based on voluntary maternal ethanol consumption throughout gestation resulted in offspring that showed mild developmental delay, anxiety-related traits, and deficits in spatial learning. Here, we extend this model by evaluating the gene expression changes that occur in the adult brain of C57BL/6J mice prenatally exposed to ethanol via maternal preference drinking. The results of two independent expression array experiments indicate that ethanol induces subtle but consistent changes to global gene expression. Gene enrichment analysis showed over-represented gene ontology classifications of cellular, embryonic, and nervous system development. Molecular network analysis supported these classifications, with significant networks related to cellular and tissue development, free radical scavenging, and small molecule metabolism. Further, a number of genes identified have previously been implicated in FASD-relevant neurobehavioural phenotypes such as cognitive function (Ache, Bcl2, Cul4b, Dkc1, Ebp, Lcat, Nsdh1, Sstr3), anxiety (Bcl2), attention deficit hyperactivity disorder (Nsdh1), and mood disorders (Bcl2, Otx2, Sstr3). The results suggest a complex residual "footprint" of neurodevelopmental ethanol exposure that may provide a new perspective for identifying mechanisms that underlie the life-long persistence of FASD-related cognitive and behavioural alterations, including potential targets for treatment.
Collapse
Affiliation(s)
- Morgan L Kleiber
- Molecular Genetics Unit, Department of Biology, University of Western Ontario, London, Ontario, Canada N6A 5B7
| | | | | | | | | |
Collapse
|
18
|
Drago A, De Ronchi D, Serretti A. Incomplete coverage of candidate genes: a poorly considered bias. Curr Genomics 2011; 8:476-83. [PMID: 19412419 PMCID: PMC2647155 DOI: 10.2174/138920207783591681] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2007] [Revised: 10/22/2007] [Accepted: 11/01/2007] [Indexed: 12/31/2022] Open
Abstract
Current genetic investigations are performed both on the basis of a rational and biologically based choice of candidate genes and through genome wide scans. Nonetheless, lack of replication is a common problem in psychiatric genetics as well as in other genetic fields. There are a number of reasons for this inconsistency, among them a well known but poorly considered issue is gene coverage. The aim of the present paper is to focus on this well known and defectively deemed bias, especially when a candidate gene approach is chosen. The rational and the technical feasibility of this proposal are discussed as well as a survey of current investigations. The known consistent methodology to fix this bias is also discussed.
Collapse
Affiliation(s)
- Antonio Drago
- Institute of Psychiatry, University of Bologna, Italy
| | | | | |
Collapse
|
19
|
Tilleman H, Kofman O, Nashelsky L, Livneh U, Roz N, Sillaber I, Biegon A, Rehavi M, Brodski C. Critical role of the embryonic mid-hindbrain organizer in the behavioral response to amphetamine and methylphenidate. Neuroscience 2009; 163:1012-23. [PMID: 19635527 DOI: 10.1016/j.neuroscience.2009.07.040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2009] [Revised: 06/14/2009] [Accepted: 07/18/2009] [Indexed: 10/20/2022]
Abstract
The embryonic mid-hindbrain organizer, which is composed of a transient cell population in the brainstem, controls the development of dopaminergic and serotonergic neurons. Different genes determining the position and activity of this embryonic structure have been implicated in dopamine- and serotonin-associated disorders. Mouse mutants with a caudally shifted mid-hindbrain organizer, are hyperactive, show increased numbers of dopaminergic neurons and a reduction in serotonergic cells. In the present study we used these mutants to gain insights into the genetic and developmental mechanisms underlying motor activity and the response to psychostimulants. To this end, we studied the motor activity of these animals after exposure to methylphenidate and amphetamine and characterized their dopaminergic and serotonergic innervation. Saline-treated mutants showed increased locomotion, more stereotypic behavior and a decrease in rearing compared to wild-type mice. This baseline level of activity was similar to behaviors observed in wild-type animals treated with high doses of psychostimulants. In mutants methylphenidate (5 or 30 mg/kg) or amphetamine (2 or 4 mg/kg) did not further increase activity or even caused a decrease of locomotor activity, in contrast to wild-type mice. Fluoxetine (5 or 10 mg/kg) reduced hyperactivity of mutants to levels observed in wild-types. Transmitter measurements, dopamine and serotonin transporter binding assays and autoradiography, indicated a subtle increase in striatal dopaminergic innervation and a marked general decrease of serotonergic innervation in mutants. Taken together, our data suggest that mice with an aberrantly positioned mid-hindbrain organizer show altered sensitivity to psychostimulants and that an increase of serotonergic neurotransmission reverses their hyperactivity. We conclude that the mid-hindbrain organizer, by orchestrating the formation of dopaminergic and serotonergic neurons, is an essential developmental parameter of locomotor activity and psychostimulant response.
Collapse
Affiliation(s)
- H Tilleman
- Department of Morphology, Zlotowski Center for Neuroscience, Division of Basic Sciences, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Serretti A, Mandelli L. The genetics of bipolar disorder: genome 'hot regions,' genes, new potential candidates and future directions. Mol Psychiatry 2008; 13:742-71. [PMID: 18332878 DOI: 10.1038/mp.2008.29] [Citation(s) in RCA: 131] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Bipolar disorder (BP) is a complex disorder caused by a number of liability genes interacting with the environment. In recent years, a large number of linkage and association studies have been conducted producing an extremely large number of findings often not replicated or partially replicated. Further, results from linkage and association studies are not always easily comparable. Unfortunately, at present a comprehensive coverage of available evidence is still lacking. In the present paper, we summarized results obtained from both linkage and association studies in BP. Further, we indicated new potential interesting genes, located in genome 'hot regions' for BP and being expressed in the brain. We reviewed published studies on the subject till December 2007. We precisely localized regions where positive linkage has been found, by the NCBI Map viewer (http://www.ncbi.nlm.nih.gov/mapview/); further, we identified genes located in interesting areas and expressed in the brain, by the Entrez gene, Unigene databases (http://www.ncbi.nlm.nih.gov/entrez/) and Human Protein Reference Database (http://www.hprd.org); these genes could be of interest in future investigations. The review of association studies gave interesting results, as a number of genes seem to be definitively involved in BP, such as SLC6A4, TPH2, DRD4, SLC6A3, DAOA, DTNBP1, NRG1, DISC1 and BDNF. A number of promising genes, which received independent confirmations, and genes that have to be further investigated in BP, have been also systematically listed. In conclusion, the combination of linkage and association approaches provided a number of liability genes. Nevertheless, other approaches are required to disentangle conflicting findings, such as gene interaction analyses, interaction with psychosocial and environmental factors and, finally, endophenotype investigations.
Collapse
Affiliation(s)
- A Serretti
- Institute of Psychiatry, University of Bologna, Bologna, Italy.
| | | |
Collapse
|