1
|
Incontro S, Musella ML, Sammari M, Di Scala C, Fantini J, Debanne D. Lipids shape brain function through ion channel and receptor modulations: physiological mechanisms and clinical perspectives. Physiol Rev 2025; 105:137-207. [PMID: 38990068 DOI: 10.1152/physrev.00004.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/28/2024] [Accepted: 07/01/2024] [Indexed: 07/12/2024] Open
Abstract
Lipids represent the most abundant molecular type in the brain, with a fat content of ∼60% of the dry brain weight in humans. Despite this fact, little attention has been paid to circumscribe the dynamic role of lipids in brain function and disease. Membrane lipids such as cholesterol, phosphoinositide, sphingolipids, arachidonic acid, and endocannabinoids finely regulate both synaptic receptors and ion channels that ensure critical neural functions. After a brief introduction on brain lipids and their respective properties, we review here their role in regulating synaptic function and ion channel activity, action potential propagation, neuronal development, and functional plasticity and their contribution in the development of neurological and neuropsychiatric diseases. We also provide possible directions for future research on lipid function in brain plasticity and diseases.
Collapse
Affiliation(s)
| | | | - Malika Sammari
- UNIS, INSERM, Aix-Marseille Université, Marseille, France
| | | | | | | |
Collapse
|
2
|
Devine EA, Imami AS, Eby H, Sahay S, Hamoud AR, Golchin H, Ryan W, Shedroff EA, Arvay T, Joyce AW, Asah SM, Walss-Bass C, O'Donovan S, McCullumsmith RE. Neuronal alterations in AKT isotype expression in schizophrenia. Mol Psychiatry 2024:10.1038/s41380-024-02770-8. [PMID: 39424930 DOI: 10.1038/s41380-024-02770-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 09/05/2024] [Accepted: 09/25/2024] [Indexed: 10/21/2024]
Abstract
Schizophrenia is characterized by substantial alterations in brain function, and previous studies suggest insulin signaling pathways, particularly involving AKT, are implicated in the pathophysiology of the disorder. This study demonstrates elevated mRNA expression of AKT1-3 in neurons from schizophrenia subjects, contrary to unchanged or diminished total AKT protein expression reported in previous postmortem studies, suggesting a potential decoupling of transcript and protein levels. Sex-specific differential AKT activity was observed, indicating divergent roles in males and females with schizophrenia. Alongside AKT, upregulation of PDPK1, a critical component of the insulin signaling pathway, and several protein phosphatases known to regulate AKT were detected. Moreover, enhanced expression of the transcription factor FOXO1, a regulator of glucose metabolism, hints at possible compensatory mechanisms related to insulin signaling dysregulation. Findings were largely independent of antipsychotic medication use, suggesting inherent alterations in schizophrenia. These results highlight the significance of AKT and related signaling pathways in schizophrenia, proposing that these changes might represent a compensatory response to a primary defect of canonical insulin signaling pathways. This research underscores the need for a detailed understanding of these signaling pathways for the development of effective therapeutic strategies.
Collapse
Affiliation(s)
- Emily A Devine
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA.
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - Ali S Imami
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Hunter Eby
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Smita Sahay
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Abdul-Rizaq Hamoud
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Hasti Golchin
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - William Ryan
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Elizabeth A Shedroff
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Taylen Arvay
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Alex W Joyce
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Sophie M Asah
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Consuelo Walss-Bass
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Sinead O'Donovan
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Robert E McCullumsmith
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
- Department of Psychiatry, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
- Neurosciences Institute, ProMedica, Toledo, OH, USA
| |
Collapse
|
3
|
Bergstrom JJD, Fu MM. Dysregulation of myelination-related genes in schizophrenia. J Neurochem 2024; 168:2227-2242. [PMID: 39086020 PMCID: PMC11449665 DOI: 10.1111/jnc.16152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 08/02/2024]
Abstract
Schizophrenic individuals display disrupted myelination patterns, altered oligodendrocyte distribution, and abnormal oligodendrocyte morphology. Schizophrenia is linked with dysregulation of a variety of genes involved in oligodendrocyte function and myelin production. Single-nucleotide polymorphisms (SNPs) and rare mutations in myelination-related genes are observed in certain schizophrenic populations, representing potential genetic risk factors. Downregulation of myelination-related RNAs and proteins, particularly in frontal and limbic regions, is consistently associated with the disorder across multiple studies. These findings support the notion that disruptions in myelination may contribute to the cognitive and behavioral impairments experienced in schizophrenia, although further evidence of causation is needed.
Collapse
Affiliation(s)
| | - Meng-Meng Fu
- Department of Molecular and Cell Biology, UC Berkeley, Berkeley, California, USA
| |
Collapse
|
4
|
Chien WH, Chen CH, Cheng MC, Wu YY, Gau SSF. Neuregulin 2 Is a Candidate Gene for Autism Spectrum Disorder. Int J Mol Sci 2024; 25:5547. [PMID: 38791584 PMCID: PMC11121989 DOI: 10.3390/ijms25105547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/04/2024] [Accepted: 05/16/2024] [Indexed: 05/26/2024] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder with heterogeneous and complex genetic underpinnings. Our previous microarray gene expression profiling identified significantly different neuregulin-2 gene (NRG2) expression between ASD patients and controls. Thus, we aimed to clarify whether NRG2 is a candidate gene associated with ASD. The study consisted of two stages. First, we used real-time quantitative PCR in 20 ASDs and 20 controls to confirm the microarray gene expression profiling results. The average NRG2 gene expression level in patients with ASD (3.23 ± 2.80) was significantly lower than that in the controls (9.27 ± 4.78, p < 0.001). Next, we conducted resequencing of all the exons of NRG2 in a sample of 349 individuals with ASD, aiming to identify variants of the NRG2 associated with ASD. We identified three variants, including two single nucleotide variants (SNVs), IVS3 + 13A > G (rs889022) and IVS10 + 32T > A (rs182642591), and one small deletion at exon 11 of NRG2 (delGCCCGG, rs933769137). Using data from the Taiwan Biobank as the controls, we found no significant differences in allele frequencies of rs889022 and rs182642591 between two groups. However, there is a significant difference in the genotype and allele frequency distribution of rs933769137 between ASDs and controls (p < 0.0001). The small deletion is located in the EGF-like domain at the C-terminal of the NRG2 precursor protein. Our findings suggest that NRG2 might be a susceptibility gene for ASD.
Collapse
Affiliation(s)
- Wei-Hsien Chien
- Department of Occupational Therapy, College of Medicine, Fu Jen Catholic University, New Taipei City 242062, Taiwan
| | - Chia-Hsiang Chen
- Department of Psychiatry, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (C.-H.C.); (Y.-Y.W.)
| | - Min-Chih Cheng
- Department of Psychiatry, Yuli Branch, Taipei Veterans General Hospital, Hualien 981, Taiwan;
| | - Yu-Yu Wu
- Department of Psychiatry, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (C.-H.C.); (Y.-Y.W.)
| | - Susan Shur-Fen Gau
- Department of Psychiatry, National Taiwan University Hospital, Taipei 10002, Taiwan
- Graduate Institute of Brain and Mind Sciences and Graduate of Clinical Medicine, National Taiwan University, Taipei 10002, Taiwan
| |
Collapse
|
5
|
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
|
6
|
Rivera AD, Normanton JR, Butt AM, Azim K. The Genomic Intersection of Oligodendrocyte Dynamics in Schizophrenia and Aging Unravels Novel Pathological Mechanisms and Therapeutic Potentials. Int J Mol Sci 2024; 25:4452. [PMID: 38674040 PMCID: PMC11050044 DOI: 10.3390/ijms25084452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/28/2024] [Accepted: 03/30/2024] [Indexed: 04/28/2024] Open
Abstract
Schizophrenia is a significant worldwide health concern, affecting over 20 million individuals and contributing to a potential reduction in life expectancy by up to 14.5 years. Despite its profound impact, the precise pathological mechanisms underlying schizophrenia continue to remain enigmatic, with previous research yielding diverse and occasionally conflicting findings. Nonetheless, one consistently observed phenomenon in brain imaging studies of schizophrenia patients is the disruption of white matter, the bundles of myelinated axons that provide connectivity and rapid signalling between brain regions. Myelin is produced by specialised glial cells known as oligodendrocytes, which have been shown to be disrupted in post-mortem analyses of schizophrenia patients. Oligodendrocytes are generated throughout life by a major population of oligodendrocyte progenitor cells (OPC), which are essential for white matter health and plasticity. Notably, a decline in a specific subpopulation of OPC has been identified as a principal factor in oligodendrocyte disruption and white matter loss in the aging brain, suggesting this may also be a factor in schizophrenia. In this review, we analysed genomic databases to pinpoint intersections between aging and schizophrenia and identify shared mechanisms of white matter disruption and cognitive dysfunction.
Collapse
Affiliation(s)
- Andrea D. Rivera
- Department of Neuroscience, Institute of Human Anatomy, University of Padova, Via A. Gabelli 65, 35127 Padua, Italy;
| | - John R. Normanton
- GliaGenesis Limited, Orchard Lea, Horns Lane, Oxfordshire, Witney OX29 8NH, UK; (J.R.N.); (K.A.)
| | - Arthur M. Butt
- GliaGenesis Limited, Orchard Lea, Horns Lane, Oxfordshire, Witney OX29 8NH, UK; (J.R.N.); (K.A.)
- School of Pharmacy and Biomedical Science, University of Portsmouth, Hampshire PO1 2UP, UK
| | - Kasum Azim
- GliaGenesis Limited, Orchard Lea, Horns Lane, Oxfordshire, Witney OX29 8NH, UK; (J.R.N.); (K.A.)
- Independent Data Lab UG, Frauenmantelanger 31, 80937 Munich, Germany
| |
Collapse
|
7
|
Devine EA, Imami AS, Eby H, Hamoud AR, Golchin H, Ryan W, Sahay S, Shedroff EA, Arvay T, Joyce AW, Asah SM, Walss-Bass C, O'Donovan S, McCullumsmith RE. Neuronal alterations in AKT isotype expression in schizophrenia. RESEARCH SQUARE 2024:rs.3.rs-3940448. [PMID: 38559131 PMCID: PMC10980160 DOI: 10.21203/rs.3.rs-3940448/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Schizophrenia is characterized by substantial alterations in brain function, and previous studies suggest insulin signaling pathways, particularly involving AKT, are implicated in the pathophysiology of the disorder. This study demonstrates elevated mRNA expression of AKT1-3 in neurons from schizophrenia subjects, contrary to unchanged or diminished total AKT protein expression reported in previous postmortem studies, suggesting a potential decoupling of transcript and protein levels. Sex-specific differential AKT activity was observed, indicating divergent roles in males and females with schizophrenia. Alongside AKT, upregulation of PDPK1, a critical component of the insulin signaling pathway, and several protein phosphatases known to regulate AKT were detected. Moreover, enhanced expression of the transcription factor FOXO1, a regulator of glucose metabolism, hints at possible compensatory mechanisms related to insulin signaling dysregulation. Findings were largely independent of antipsychotic medication use, suggesting inherent alterations in schizophrenia. These results highlight the significance of AKT and related signaling pathways in schizophrenia, proposing that these changes might represent a compensatory response to a primary defect of conical insulin signaling pathways. This research underscores the need for a detailed understanding of these signaling pathways for the development of effective therapeutic strategies.
Collapse
Affiliation(s)
- Emily A Devine
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ali S Imami
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Hunter Eby
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Abdul-Rizaq Hamoud
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Hasti Golchin
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - William Ryan
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Smita Sahay
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Elizabeth A Shedroff
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Taylen Arvay
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Alex W Joyce
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Sophie M Asah
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Consuelo Walss-Bass
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Sinead O'Donovan
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Robert E McCullumsmith
- Department of Neuroscience, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
- Neurosciences Institute, ProMedica, Toledo, OH, USA
- Department of Psychiatry, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| |
Collapse
|
8
|
Yang Q, Zhang L, Li M, Xu Y, Chen X, Yuan R, Ou X, He M, Liao M, Zhang L, Dai H, Lv M, Xie X, Liang W, Chen X. Single-nucleus transcriptomic mapping uncovers targets for traumatic brain injury. Genome Res 2023; 33:1818-1832. [PMID: 37730437 PMCID: PMC10691476 DOI: 10.1101/gr.277881.123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 09/11/2023] [Indexed: 09/22/2023]
Abstract
The subventricular zone (SVZ) is a neurogenic niche that contributes to homeostasis and repair after brain injury. However, the effects of mild traumatic brain injury (mTBI) on the divergence of the regulatory DNA landscape within the SVZ and its link to functional alterations remain unexplored. In this study, we mapped the transcriptome atlas of murine SVZ and its responses to mTBI at the single-cell level. We observed cell-specific gene expression changes following mTBI and unveiled diverse cell-to-cell interaction networks that influence a wide array of cellular processes. Moreover, we report novel neurogenesis lineage trajectories and related key transcription factors, which we validate through loss-of-function experiments. Specifically, we validate the role of Tcf7l1, a cell cycle gene regulator, in promoting neural stem cell differentiation toward the neuronal lineage after mTBI, providing a potential target for regenerative medicine. Overall, our study profiles an SVZ transcriptome reference map, which underlies the differential cellular behavior in response to mTBI. The identified key genes and pathways that may ameliorate brain damage or facilitate neural repair serve as a comprehensive resource for drug discovery in the context of mTBI.
Collapse
Affiliation(s)
- Qiuyun Yang
- Department of Forensic Genetics, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610000, China
- West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Lingxuan Zhang
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610000, China
| | - Manrui Li
- Department of Forensic Genetics, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610000, China
| | - Yang Xu
- Department of Forensic Genetics, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610000, China
| | - Xiaogang Chen
- Department of Forensic Pathology and Forensic Clinical Medicine, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610000, China
| | - Ruixuan Yuan
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610000, China
| | - Xiaofeng Ou
- Department of Critical Care Medicine, Sichuan University, Chengdu 610000, China
| | - Min He
- Department of Critical Care Medicine, Sichuan University, Chengdu 610000, China
| | - Miao Liao
- Department of Forensic Genetics, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610000, China
| | - Lin Zhang
- Sichuan University, Chengdu 610041, China
| | - Hao Dai
- Department of Forensic Pathology and Forensic Clinical Medicine, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610000, China
| | - Meili Lv
- Department of Immunology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610000, China
| | - Xiaoqi Xie
- Department of Critical Care Medicine, Sichuan University, Chengdu 610000, China;
| | - Weibo Liang
- Department of Forensic Genetics, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610000, China;
| | - Xiameng Chen
- Department of Forensic Pathology and Forensic Clinical Medicine, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610000, China;
| |
Collapse
|
9
|
Synaptic plasticity in Schizophrenia pathophysiology. IBRO Neurosci Rep 2023. [DOI: 10.1016/j.ibneur.2023.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
10
|
Hampel H, Caruso G, Nisticò R, Piccioni G, Mercuri NB, Giorgi FS, Ferrarelli F, Lemercier P, Caraci F, Lista S, Vergallo A. Biological Mechanism-based Neurology and Psychiatry: A BACE1/2 and Downstream Pathway Model. Curr Neuropharmacol 2023; 21:31-53. [PMID: 34852743 PMCID: PMC10193755 DOI: 10.2174/1570159x19666211201095701] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 11/26/2021] [Accepted: 11/28/2021] [Indexed: 02/04/2023] Open
Abstract
In oncology, comprehensive omics and functional enrichment studies have led to an extensive profiling of (epi)genetic and neurobiological alterations that can be mapped onto a single tumor's clinical phenotype and divergent clinical phenotypes expressing common pathophysiological pathways. Consequently, molecular pathway-based therapeutic interventions for different cancer typologies, namely tumor type- and site-agnostic treatments, have been developed, encouraging the real-world implementation of a paradigm shift in medicine. Given the breakthrough nature of the new-generation translational research and drug development in oncology, there is an increasing rationale to transfertilize this blueprint to other medical fields, including psychiatry and neurology. In order to illustrate the emerging paradigm shift in neuroscience, we provide a state-of-the-art review of translational studies on the β-site amyloid precursor protein cleaving enzyme (BACE) and its most studied downstream effector, neuregulin, which are molecular orchestrators of distinct biological pathways involved in several neurological and psychiatric diseases. This body of data aligns with the evidence of a shared genetic/biological architecture among Alzheimer's disease, schizoaffective disorder, and autism spectrum disorders. To facilitate a forward-looking discussion about a potential first step towards the adoption of biological pathway-based, clinical symptom-agnostic, categorization models in clinical neurology and psychiatry for precision medicine solutions, we engage in a speculative intellectual exercise gravitating around BACE-related science, which is used as a paradigmatic case here. We draw a perspective whereby pathway-based therapeutic strategies could be catalyzed by highthroughput techniques embedded in systems-scaled biology, neuroscience, and pharmacology approaches that will help overcome the constraints of traditional descriptive clinical symptom and syndrome-focused constructs in neurology and psychiatry.
Collapse
Affiliation(s)
- Harald Hampel
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris, France
| | | | - Robert Nisticò
- Laboratory of Pharmacology of Synaptic Plasticity, EBRI Rita Levi-Montalcini Foundation, Rome, Italy
- School of Pharmacy, University of Rome “Tor Vergata”, Rome, Italy
| | - Gaia Piccioni
- Laboratory of Pharmacology of Synaptic Plasticity, EBRI Rita Levi-Montalcini Foundation, Rome, Italy
- Department of Physiology and Pharmacology “V.Erspamer”, Sapienza University of Rome, Rome, Italy
| | - Nicola B. Mercuri
- Department of Systems Medicine, University of Rome “Tor Vergata”, Rome, Italy
- IRCCS Santa Lucia Foundation, Rome, Italy
| | - Filippo Sean Giorgi
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | - Fabio Ferrarelli
- Department of Psychiatry, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Pablo Lemercier
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris, France
| | - Filippo Caraci
- Oasi Research Institute-IRCCS, Troina, Italy
- Department of Drug Sciences, University of Catania, Catania, Italy
| | - Simone Lista
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris, France
- Memory Resources and Research Center (CMRR), Neurology Department, Gui de Chauliac University Hospital, Montpellier, France
| | - Andrea Vergallo
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris, France
| |
Collapse
|
11
|
Mansour HM, El-Khatib AS. Repositioning of receptor tyrosine kinase inhibitors. RECEPTOR TYROSINE KINASES IN NEURODEGENERATIVE AND PSYCHIATRIC DISORDERS 2023:353-401. [DOI: 10.1016/b978-0-443-18677-6.00010-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
12
|
Zhang K, Liao P, Wen J, Hu Z. Synaptic plasticity in schizophrenia pathophysiology. IBRO Neurosci Rep 2022; 13:478-487. [PMID: 36590092 PMCID: PMC9795311 DOI: 10.1016/j.ibneur.2022.10.008] [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: 05/22/2022] [Accepted: 10/21/2022] [Indexed: 11/05/2022] Open
Abstract
Schizophrenia is a severe neuropsychiatric syndrome with psychotic behavioral abnormalities and marked cognitive deficits. It is widely accepted that genetic and environmental factors contribute to the onset of schizophrenia. However, the etiology and pathology of the disease remain largely unexplored. Recently, the synaptopathology and the dysregulated synaptic plasticity and function have emerging as intriguing and prominent biological mechanisms of schizophrenia pathogenesis. Synaptic plasticity is the ability of neurons to change the strength of their connections in response to internal or external stimuli, which is essential for brain development and function, learning and memory, and vast majority of behavior responses relevant to psychiatric diseases including schizophrenia. Here, we reviewed molecular and cellular mechanisms of the multiple forms synaptic plasticity, and the functional regulations of schizophrenia-risk factors including disease susceptible genes and environmental alterations on synaptic plasticity and animal behavior. Recent genome-wide association studies have provided fruitful findings of hundreds of risk gene variances associated with schizophrenia, thus further clarifying the role of these disease-risk genes in synaptic transmission and plasticity will be beneficial to advance our understanding of schizophrenia pathology, as well as the molecular mechanism of synaptic plasticity.
Collapse
Affiliation(s)
- Kexuan Zhang
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China,Center for Medical Genetics, School of Life Sciences, Central South University, Changsha 410008, Hunan, PR China
| | - Panlin Liao
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China,National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Jin Wen
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China,National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Zhonghua Hu
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China,Center for Medical Genetics, School of Life Sciences, Central South University, Changsha 410008, Hunan, PR China,National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China,Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China,Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Sciences, Central South University, Changsha 410008, Hunan, PR China,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha 410008, Hunan, PR China,Correspondence to: Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, 87 Xiangya Rd, Changsha, Hunan, PR China.
| |
Collapse
|
13
|
Disrupted in schizophrenia 1 regulates ectopic neurogenesis in the mouse hilus after pilocarpine-induced status epilepticus. Neuroscience 2022; 494:69-81. [DOI: 10.1016/j.neuroscience.2022.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 05/01/2022] [Accepted: 05/06/2022] [Indexed: 11/20/2022]
|
14
|
Furuta S, Aleksic B, Nawa Y, Kimura H, Kushima I, Ishizuka K, Kato H, Toyama M, Arioka Y, Mori D, Morikawa M, Inada T, Ozaki N. Investigation of OLIG2 as a candidate gene for schizophrenia and autism spectrum disorder. NAGOYA JOURNAL OF MEDICAL SCIENCE 2022; 84:260-268. [PMID: 35967956 PMCID: PMC9350582 DOI: 10.18999/nagjms.84.2.260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 07/21/2021] [Indexed: 11/28/2022]
Abstract
A number of genomic mutations that are thought to be strongly involved in the development of schizophrenia (SCZ) and autism spectrum disorder (ASD) have been identified. Abnormalities involving oligodendrocytes have been reported in SCZ, and as a related gene, oligodendrocyte lineage transcription factor 2 (OLIG2) has been reported to be strongly associated with SCZ. In this study, based on the common disease-rare variant hypothesis, target sequencing of candidate genes was performed to identify rare mutations with a high effect size and the possibility that the identified mutations may increase the risks of SCZ and ASD in the Japanese population. In this study, the exon region of OLIG2 was targeted; 370 patients with SCZ and 192 with ASD were subjected to next-generation sequencing. As a result, one rare missense mutation (A33T) was detected. We used the Sanger method to validate this missense mutation with a low frequency (<1%), and then carried out a genetic association analysis involving 3299 unrelated individuals (1447 with SCZ, 380 with ASD, and 1472 healthy controls) to clarify whether A33T was associated with SCZ or ASD. A33T was not found in either case group, and in only one control. We did not find evidence that p.A33T is involved in the onset of ASD or SCZ; however, associations with this variant need to be evaluated in larger samples to confirm our results.
Collapse
Affiliation(s)
- Sho Furuta
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Branko Aleksic
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshihiro Nawa
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroki Kimura
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Itaru Kushima
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
,Medical Genomics Center, Nagoya University Hospital, Nagoya, Japan
| | - Kanako Ishizuka
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hidekazu Kato
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Miho Toyama
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuko Arioka
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
,Institute for Advanced Research, Nagoya University, Nagoya, Japan
,Center for Advanced Medicine and Clinical Research, Nagoya University Hospital, Nagoya, Japan
| | - Daisuke Mori
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
,Brain and Mind Research Center, Nagoya University, Nagoya, Japan
| | - Mako Morikawa
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Toshiya Inada
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Norio Ozaki
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
15
|
Maly IV, Morales MJ, Pletnikov MV. Astrocyte Bioenergetics and Major Psychiatric Disorders. ADVANCES IN NEUROBIOLOGY 2021; 26:173-227. [PMID: 34888836 DOI: 10.1007/978-3-030-77375-5_9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Ongoing research continues to add new elements to the emerging picture of involvement of astrocyte energy metabolism in the pathophysiology of major psychiatric disorders, including schizophrenia, mood disorders, and addictions. This review outlines what is known about the energy metabolism in astrocytes, the most numerous cell type in the brain, and summarizes the recent work on how specific perturbations of astrocyte bioenergetics may contribute to the neuropsychiatric conditions. The role of astrocyte energy metabolism in mental health and disease is reviewed on the organism, organ, and cell level. Data arising from genomic, metabolomic, in vitro, and neurobehavioral studies is critically analyzed to suggest future directions in research and possible metabolism-focused therapeutic interventions.
Collapse
Affiliation(s)
- Ivan V Maly
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY, USA
| | - Michael J Morales
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY, USA
| | - Mikhail V Pletnikov
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY, USA.
| |
Collapse
|
16
|
Tsai SH, Tsao CY, Lee LJ. Altered White Matter and Layer VIb Neurons in Heterozygous Disc1 Mutant, a Mouse Model of Schizophrenia. Front Neuroanat 2020; 14:605029. [PMID: 33384588 PMCID: PMC7769951 DOI: 10.3389/fnana.2020.605029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 11/24/2020] [Indexed: 11/13/2022] Open
Abstract
Increased white matter neuron density has been associated with neuropsychiatric disorders including schizophrenia. However, the pathogenic features of these neurons are still largely unknown. Subplate neurons, the earliest generated neurons in the developing cortex have also been associated with schizophrenia and autism. The link between these neurons and mental disorders is also not well established. Since cortical layer VIb neurons are believed to be the remnant of subplate neurons in the adult rodent brain, in this study, we aimed to examine the cytoarchitecture of neurons in cortical layer VIb and the underlying white matter in heterozygous Disc1 mutant (Het) mice, a mouse model of schizophrenia. In the white matter, the number of NeuN-positive neurons was quite low in the external capsule; however, the density of these cells was found increased (54%) in Het mice compared with wildtype (WT) littermates. The density of PV-positive neurons was unchanged in the mutants. In the cortical layer VIb, the density of CTGF-positive neurons increased (21.5%) in Het mice, whereas the number of Cplx3-positive cells reduced (16.1%) in these mutants, compared with WT mice. Layer VIb neurons can be classified by their morphological characters. The morphology of Type I pyramidal neurons was comparable between genotypes while the dendritic length and complexity of Type II multipolar neurons were significantly reduced in Het mice. White matter neurons and layer VIb neurons receive synaptic inputs and modulate the process of sensory information and sleep/arousal pattern. Aberrances of these neurons in Disc1 mutants implies altered brain functions in these mice.
Collapse
Affiliation(s)
- Shin-Hwa Tsai
- School of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chih-Yu Tsao
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University, Taipei, Taiwan
| | - Li-Jen Lee
- School of Medicine, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University, Taipei, Taiwan
- Institute of Brain and Mind Sciences, National Taiwan University, Taipei, Taiwan
- Neurobiology and Cognitive Science Center, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
17
|
Shevelkin AV, Terrillion CE, Hasegawa Y, Mychko OA, Jouroukhin Y, Sawa A, Kamiya A, Pletnikov MV. Astrocyte DISC1 contributes to cognitive function in a brain region-dependent manner. Hum Mol Genet 2020; 29:2936-2950. [PMID: 32803234 PMCID: PMC8248941 DOI: 10.1093/hmg/ddaa180] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/11/2020] [Accepted: 06/30/2020] [Indexed: 12/13/2022] Open
Abstract
Our understanding of the contribution of genetic risk factors to neuropsychiatric diseases is limited to abnormal neurodevelopment and neuronal dysfunction. Much less is known about the mechanisms whereby risk variants could affect the physiology of glial cells. Our prior studies have shown that a mutant (dominant-negative) form of a rare but highly penetrant psychiatric risk factor, Disrupted-In-Schizophrenia-1 (DISC1), impairs metabolic functions of astrocytes and leads to cognitive dysfunction. In order to overcome the limitations of the mutant DISC1 model and understand the putative regional properties of astrocyte DISC1, we assessed whether knockdown of Disc1 (Disc1-KD) in mature mouse astrocytes of the prefrontal cortex (PFC) or the hippocampus would produce behavioral abnormalities that could be attributed to astrocyte bioenergetics. We found that Disc1-KD in the hippocampus but not PFC impaired trace fear conditioning in adult mice. Using the innovative deep learning approach and convolutional deep neural networks (cDNNs), ResNet50 or ResNet18, and single cell-based analysis, we found that Disc1-KD decreased the spatial density of astrocytes associated with abnormal levels and distribution of the mitochondrial markers and the glutamate transporter, GLAST. Disc1-KD in astrocytes also led to decreased expression of the glutamatergic and increased expression of the GABA-ergic synaptic markers, possibly via non-apoptotic activation of caspase 3 in neurons located within the individual territories of Disc1-KD astrocytes. Our results indicate that altered expression of DISC1 in astrocytes could impair astrocyte bioenergetics, leading to abnormalities in synaptic neurotransmission and cognitive function in a region-dependent fashion.
Collapse
Affiliation(s)
| | | | | | | | | | - Akira Sawa
- Departments of Psychiatry and Behavioral Sciences
- Solomon H. Snyder Department of Neuroscience
- Department of Biomedical Engineering
- Department of Genetic Medicine, Johns Hopkins University School of Medicine
- Department of Mental Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21287, USA
| | - Atsushi Kamiya
- Departments of Psychiatry and Behavioral Sciences
- Solomon H. Snyder Department of Neuroscience
| | - Mikhail V Pletnikov
- Departments of Psychiatry and Behavioral Sciences
- Solomon H. Snyder Department of Neuroscience
| |
Collapse
|
18
|
Kano SI, Hodgkinson CA, Jones-Brando L, Eastwood S, Ishizuka K, Niwa M, Choi EY, Chang DJ, Chen Y, Velivela SD, Leister F, Wood J, Chowdari K, Ducci F, Caycedo DA, Heinz E, Newman ER, Cascella N, Mortensen PB, Zandi PP, Dickerson F, Nimgaonkar V, Goldman D, Harrison PJ, Yolken RH, Sawa A. Host-parasite interaction associated with major mental illness. Mol Psychiatry 2020; 25:194-205. [PMID: 30127472 PMCID: PMC6382596 DOI: 10.1038/s41380-018-0217-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 04/16/2018] [Accepted: 06/20/2018] [Indexed: 11/23/2022]
Abstract
Clinical studies frequently report that patients with major mental illness such as schizophrenia and bipolar disorder have co-morbid physical conditions, suggesting that systemic alterations affecting both brain and peripheral tissues might underlie the disorders. Numerous studies have reported elevated levels of anti-Toxoplasma gondii (T. gondii) antibodies in patients with major mental illnesses, but the underlying mechanism was unclear. Using multidisciplinary epidemiological, cell biological, and gene expression profiling approaches, we report here multiple lines of evidence suggesting that a major mental illness-related susceptibility factor, Disrupted in schizophrenia (DISC1), is involved in host immune responses against T. gondii infection. Specifically, our cell biology and gene expression studies have revealed that DISC1 Leu607Phe variation, which changes DISC1 interaction with activating transcription factor 4 (ATF4), modifies gene expression patterns upon T. gondii infection. Our epidemiological data have also shown that DISC1 607 Phe/Phe genotype was associated with higher T. gondii antibody levels in sera. Although further studies are required, our study provides mechanistic insight into one of the few well-replicated serological observations in major mental illness.
Collapse
Affiliation(s)
- Shin-Ichi Kano
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Colin A Hodgkinson
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20892, USA
| | - Lorraine Jones-Brando
- Stanley Division of Developmental Neurovirology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Sharon Eastwood
- Department of Psychiatry, University of Oxford, Oxford, OX3 7JX, United Kingdom
| | - Koko Ishizuka
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Minae Niwa
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Eric Y Choi
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Daniel J Chang
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Yian Chen
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Swetha D Velivela
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Flora Leister
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Joel Wood
- Departments of Psychiatry and Human Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Kodavali Chowdari
- Departments of Psychiatry and Human Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Francesca Ducci
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20892, USA
| | - Daniel A Caycedo
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20892, USA
| | - Elizabeth Heinz
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20892, USA
| | - Emily R Newman
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20892, USA
| | - Nicola Cascella
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Preben B Mortensen
- National Centre for Register-Based Research, University of Aarhus, Aarhus, 8000, Denmark
| | - Peter P Zandi
- Department of Mental Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Faith Dickerson
- Stanley Research Program, Sheppard Pratt Health System, Baltimore, MD, 21204, USA
| | - Vishwajit Nimgaonkar
- Departments of Psychiatry and Human Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - David Goldman
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20892, USA
| | - Paul J Harrison
- Department of Psychiatry, University of Oxford, Oxford, OX3 7JX, United Kingdom
| | - Robert H Yolken
- Stanley Division of Developmental Neurovirology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Akira Sawa
- Department of Mental Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, 21205, USA.
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| |
Collapse
|
19
|
Yang K, Kondo MA, Jaaro-Peled H, Cash-Padgett T, Kano SI, Ishizuka K, Pevsner J, Tomoda T, Sawa A, Niwa M. The transcriptome landscape associated with Disrupted-in-Schizophrenia-1 locus impairment in early development and adulthood. Schizophr Res 2019; 210:149-156. [PMID: 31204062 PMCID: PMC8050833 DOI: 10.1016/j.schres.2019.05.032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 05/21/2019] [Accepted: 05/26/2019] [Indexed: 01/08/2023]
Abstract
DISC1 was originally expected to be a genetic risk factor for schizophrenia, but the genome wide association studies have not supported this idea. In contrast, neurobiological studies of DISC1 in cell and animal models have demonstrated that direct perturbation of DISC1 protein elicits neurobiological and behavioral abnormalities relevant to a wide range of psychiatric conditions, in particular psychosis. Thus, the utility of DISC1 as a biological lead for psychosis research is clear. In the present study, we aimed to capture changes in the molecular landscape in the prefrontal cortex upon perturbation of DISC1, using the Disc1 locus impairment (Disc1-LI) model in which the majority of Disc1 isoforms have been depleted, and to explore potential molecular mediators relevant to psychiatric conditions. We observed a robust change in gene expression profile elicited by Disc1-LI in which the stronger effects on molecular networks were observed in early stage compared with those in adulthood. Significant alterations were found in specific pathways relevant to psychiatric conditions, such as pathways of signaling by G protein-coupled receptor, neurotransmitter release cycle, and voltage gated potassium channels. The differentially expressed genes (DEGs) between Disc1-LI and wild-type mice are significantly enriched not only in neurons, but also in astrocytes and oligodendrocyte precursor cells. The brain-disorder-associated genes at the mRNA and protein levels rather than those at the genomic levels are enriched in the DEGs. Together, our present study supports the utility of Disc1-LI mice in biological research for psychiatric disorder-associated molecular networks.
Collapse
Affiliation(s)
- Kun Yang
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Mari A Kondo
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Hanna Jaaro-Peled
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Tyler Cash-Padgett
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Shin-Ichi Kano
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Koko Ishizuka
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jonathan Pevsner
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - Toshifumi Tomoda
- Medical Innovation Center, Kyoto University, Kyoto 606-8397, Japan
| | - Akira Sawa
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Mental Health, Johns Hopkins University Bloomberg School of Medicine, Baltimore, MD 21205, USA.
| | - Minae Niwa
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
20
|
Wang ZT, Lu MH, Zhang Y, Ji WL, Lei L, Wang W, Fang LP, Wang LW, Yu F, Wang J, Li ZY, Wang JR, Wang TH, Dou F, Wang QW, Wang XL, Li S, Ma QH, Xu RX. Disrupted-in-schizophrenia-1 protects synaptic plasticity in a transgenic mouse model of Alzheimer's disease as a mitophagy receptor. Aging Cell 2019; 18:e12860. [PMID: 30488644 PMCID: PMC6351828 DOI: 10.1111/acel.12860] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 08/09/2018] [Accepted: 09/06/2018] [Indexed: 12/14/2022] Open
Abstract
Mitochondrial dysfunction is an early feature of Alzheimer's disease (AD). Accumulated damaged mitochondria, which are associated with impaired mitophagy, contribute to neurodegeneration in AD. We show levels of Disrupted‐in‐schizophrenia‐1 (DISC1), which is genetically associated with psychiatric disorders and AD, decrease in the brains of AD patients and transgenic model mice and in Aβ‐treated cultured cells. Disrupted‐in‐schizophrenia‐1 contains a canonical LC3‐interacting region (LIR) motif (210FSFI213), through which DISC1 directly binds to LC3‐I/II. Overexpression of DISC1 enhances mitophagy through its binding to LC3, whereas knocking‐down of DISC1 blocks Aβ‐induced mitophagy. We further observe overexpression of DISC1, but not its mutant (muFSFI) which abolishes the interaction of DISC1 with LC3, rescues Aβ‐induced mitochondrial dysfunction, loss of spines, suppressed long‐term potentiation (LTP). Overexpression of DISC1 via adeno‐associated virus (serotype 8, AAV8) in the hippocampus of 8‐month‐old APP/PS1 transgenic mice for 4 months rescues cognitive deficits, synaptic loss, and Aβ plaque accumulation, in a way dependent on the interaction of DISC1 with LC3. These results indicate that DISC1 is a novel mitophagy receptor, which protects synaptic plasticity from Aβ accumulation‐induced toxicity through promoting mitophagy.
Collapse
Affiliation(s)
- Zhao-Tao Wang
- Department of Neurosurgery; Affiliated Bayi Brain Hospital, General Army Hospital; Southern Medical University; Beijing China
| | - Mei-Hong Lu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience; Soochow University; Suzhou China
| | - Yan Zhang
- Department of Neurosurgery; Affiliated Bayi Brain Hospital, General Army Hospital; Southern Medical University; Beijing China
| | - Wen-Li Ji
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience; Soochow University; Suzhou China
| | - Lei Lei
- Department of Physiology, Liaoning Provincial Key Laboratory of Cerebral Diseases; Dalian Medical University; Dalian China
| | - Wang Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience; Soochow University; Suzhou China
| | - Li-Pao Fang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience; Soochow University; Suzhou China
| | - Lu-Wen Wang
- Department of Pathology; Case Western Reserve University; Cleveland Ohio
| | - Fan Yu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience; Soochow University; Suzhou China
| | - Ji Wang
- Department of Neurosurgery; Affiliated Bayi Brain Hospital, General Army Hospital; Southern Medical University; Beijing China
| | - Zhen-Yu Li
- Department of Neurosurgery; Affiliated Bayi Brain Hospital, General Army Hospital; Southern Medical University; Beijing China
| | - Jian-Rong Wang
- Hematology Center of Cyrus Tang Medical Institute, Collaborative Innovation Center of Hematology, Key Laboratory of Stem Cells and Biomedical; Materials of Jiangsu Province and Chinese Ministry of Science and Technology, State Key Laboratory of Radiation Medicine and Radioprotection; Soochow University School of Medicine; Suzhou China
| | - Ting-Hua Wang
- Institute of Neuroscience; Kunming Medical University; Kunming China
| | - Fei Dou
- College of Life Sciences; Beijing Normal University; Beijing China
| | - Qin-Wen Wang
- Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine; Ningbo University; Ningbo China
| | - Xing-Long Wang
- Department of Pathology; Case Western Reserve University; Cleveland Ohio
| | - Shao Li
- Department of Physiology, Liaoning Provincial Key Laboratory of Cerebral Diseases; Dalian Medical University; Dalian China
| | - Quan-Hong Ma
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience; Soochow University; Suzhou China
| | - Ru-Xiang Xu
- Department of Neurosurgery; Affiliated Bayi Brain Hospital, General Army Hospital; Southern Medical University; Beijing China
| |
Collapse
|
21
|
Murphy E, Benítez-Burraco A. Toward the Language Oscillogenome. Front Psychol 2018; 9:1999. [PMID: 30405489 PMCID: PMC6206218 DOI: 10.3389/fpsyg.2018.01999] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 09/28/2018] [Indexed: 12/20/2022] Open
Abstract
Language has been argued to arise, both ontogenetically and phylogenetically, from specific patterns of brain wiring. We argue that it can further be shown that core features of language processing emerge from particular phasal and cross-frequency coupling properties of neural oscillations; what has been referred to as the language ‘oscillome.’ It is expected that basic aspects of the language oscillome result from genetic guidance, what we will here call the language ‘oscillogenome,’ for which we will put forward a list of candidate genes. We have considered genes for altered brain rhythmicity in conditions involving language deficits: autism spectrum disorders, schizophrenia, specific language impairment and dyslexia. These selected genes map on to aspects of brain function, particularly on to neurotransmitter function. We stress that caution should be adopted in the construction of any oscillogenome, given the range of potential roles particular localized frequency bands have in cognition. Our aim is to propose a set of genome-to-language linking hypotheses that, given testing, would grant explanatory power to brain rhythms with respect to language processing and evolution.
Collapse
Affiliation(s)
- Elliot Murphy
- Division of Psychology and Language Sciences, University College London, London, United Kingdom.,Department of Psychology, University of Westminster, London, United Kingdom
| | - Antonio Benítez-Burraco
- Department of Spanish Language, Linguistics and Literary Theory, University of Seville, Seville, Spain
| |
Collapse
|
22
|
Khan A, Powell SB. Sensorimotor gating deficits in "two-hit" models of schizophrenia risk factors. Schizophr Res 2018; 198:68-83. [PMID: 29070440 PMCID: PMC5911431 DOI: 10.1016/j.schres.2017.10.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 10/03/2017] [Accepted: 10/06/2017] [Indexed: 02/07/2023]
Abstract
Genetic and environmental models of neuropsychiatric disease have grown exponentially over the last 20years. One measure that is often used to evaluate the translational relevance of these models to human neuropsychiatric disease is prepulse inhibition of startle (PPI), an operational measure of sensorimotor gating. Deficient PPI characterizes several neuropsychiatric disorders but has been most extensively studied in schizophrenia. It has become a useful tool in translational neuropharmacological and molecular genetics studies because it can be measured across species using almost the same experimental parameters. Although initial studies of PPI in rodents were pharmacological because of the robust predictive validity of PPI for antipsychotic efficacy, more recently, PPI has become standard common behavioral measures used in genetic and neurodevelopmental models of schizophrenia. Here we review "two hit" models of schizophrenia and discuss the utility of PPI as a tool in phenotyping these models of relevant risk factors. In the review, we consider approaches to rodent models of genetic and neurodevelopmental risk factors and selectively review "two hit" models of gene×environment and environment×environment interactions in which PPI has been measured.
Collapse
Affiliation(s)
- Asma Khan
- Department of Psychiatry, University of California San Diego, 9500 Gilman Dr., La Jolla, CA 92093, United States; Research Service, VA San Diego Healthcare System, La Jolla, CA, United States
| | - Susan B Powell
- Department of Psychiatry, University of California San Diego, 9500 Gilman Dr., La Jolla, CA 92093, United States; Research Service, VA San Diego Healthcare System, La Jolla, CA, United States.
| |
Collapse
|
23
|
Ryskalin L, Limanaqi F, Frati A, Busceti CL, Fornai F. mTOR-Related Brain Dysfunctions in Neuropsychiatric Disorders. Int J Mol Sci 2018; 19:ijms19082226. [PMID: 30061532 PMCID: PMC6121884 DOI: 10.3390/ijms19082226] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 07/26/2018] [Accepted: 07/27/2018] [Indexed: 12/12/2022] Open
Abstract
The mammalian target of rapamycin (mTOR) is an ubiquitously expressed serine-threonine kinase, which senses and integrates several intracellular and environmental cues to orchestrate major processes such as cell growth and metabolism. Altered mTOR signalling is associated with brain malformation and neurological disorders. Emerging evidence indicates that even subtle defects in the mTOR pathway may produce severe effects, which are evident as neurological and psychiatric disorders. On the other hand, administration of mTOR inhibitors may be beneficial for a variety of neuropsychiatric alterations encompassing neurodegeneration, brain tumors, brain ischemia, epilepsy, autism, mood disorders, drugs of abuse, and schizophrenia. mTOR has been widely implicated in synaptic plasticity and autophagy activation. This review addresses the role of mTOR-dependent autophagy dysfunction in a variety of neuropsychiatric disorders, to focus mainly on psychiatric syndromes including schizophrenia and drug addiction. For instance, amphetamines-induced addiction fairly overlaps with some neuropsychiatric disorders including neurodegeneration and schizophrenia. For this reason, in the present review, a special emphasis is placed on the role of mTOR on methamphetamine-induced brain alterations.
Collapse
Affiliation(s)
- Larisa Ryskalin
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy.
| | - Fiona Limanaqi
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy.
| | | | | | - Francesco Fornai
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy.
- I.R.C.C.S. Neuromed, Via Atinense 18, 86077 Isernia, Italy.
| |
Collapse
|
24
|
Nafisi-Far N, Ghafouri-Fard S, Panah AST, Sayad A, Taheri M. A gender dimorphism in up-regulation of BACE1 gene expression in schizophrenia. Metab Brain Dis 2018; 33:933-937. [PMID: 29500546 DOI: 10.1007/s11011-018-0205-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 02/19/2018] [Indexed: 10/17/2022]
Abstract
Schizophrenia has long been considered as a devastating brain disorder in which both genetic and environmental factors are involved. The BACE1 gene is one of the most important susceptibility genes for this disorder. However, the changes in BACE1 expression in schizophrenic patients compared with healthy subjects have not been evaluated yet. In this case-control study, we examined BACE1 expression in a group of 50 patients with schizophrenia and 50 healthy controls. The level of BACE1 gene expression was measured using Real-Time PCR. Substantial increase in gene expression was detected in the patients compared with normal individuals (P = 0.001). Furthermore, a gender dimorphism was observed in BACE1 gene expression in the patients in a way that the male patients manifested a statistically significant higher levels of BACE1 expression (P = 0.002). BACE1 might be implicated in the pathogenesis of schizophrenia. Besides, BACE1 physiology may be gender -based at some levels. Our findings warrant an investigation of BACE1 gene in a larger number of cases and controls.
Collapse
Affiliation(s)
- Nafiseh Nafisi-Far
- Young Researchers and Elite Club, East Tehran Branch, Islamic Azad University, Tehran, Iran
- Department of Biology, East Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical sciences, Tehran, Iran
| | | | - Arezou Sayad
- Department of Medical Genetics, Shahid Beheshti University of Medical sciences, Tehran, Iran.
| | - Mohammad Taheri
- Department of Medical Genetics, Shahid Beheshti University of Medical sciences, Tehran, Iran.
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
25
|
Stewart EM, Wu Z, Huang XF, Kapsa RMI, Wallace GG. Use of conducting polymers to facilitate neurite branching in schizophrenia-related neuronal development. Biomater Sci 2018; 4:1244-51. [PMID: 27376413 DOI: 10.1039/c6bm00212a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Schizophrenia (SCZ) is a debilitating mental disorder which results in high healthcare and loss of productivity costs to society. This disease remains poorly understood, however there is increasing evidence suggesting a role for oxidative damage in the disease etiology. We aimed to examine the effect of the conducting polymer polypyrrole on the growth and morphology of both wildtype and neuregulin-1 knock out (NRG-1 +/-) explant cells. Polypyrrole is an organic conducting polymer known to be cytocompatible and capable of acting as a platform for effective stimulation of neurons. Here we demonstrate for the first time the ability of this material to mediate processes occurring in disease affected neurons: schizophrenic model cortical neurons. Prefrontal cortical cells were grown on conducting polymer scaffolds of specific composition and showed significantly increased neurite branching and outgrowth length on the polymers compared to controls. Concurrently, a more significant enhancement was seen in both parameters in the NRG-1 +/- model cells. This finding implies that conducting polymers such as polypyrrole may be utilised to overcome neuro-functional deficits associated with neurological disease in humans.
Collapse
Affiliation(s)
- Elise M Stewart
- ARC Centre of Excellence for Electromaterials Science, University of Wollongong, NSW, Australia.
| | - Zhixiang Wu
- Illawarra Health and Medical Research Institute, University of Wollongong, NSW, Australia
| | - Xu Feng Huang
- Illawarra Health and Medical Research Institute, University of Wollongong, NSW, Australia
| | - Robert M I Kapsa
- ARC Centre of Excellence for Electromaterials Science, University of Wollongong, NSW, Australia.
| | - Gordon G Wallace
- ARC Centre of Excellence for Electromaterials Science, University of Wollongong, NSW, Australia.
| |
Collapse
|
26
|
Zhang Z, Huang J, Shen Y, Li R. BACE1-Dependent Neuregulin-1 Signaling: An Implication for Schizophrenia. Front Mol Neurosci 2017; 10:302. [PMID: 28993723 PMCID: PMC5622153 DOI: 10.3389/fnmol.2017.00302] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 09/07/2017] [Indexed: 12/13/2022] Open
Abstract
Schizophrenia is a chronic psychiatric disorder with a lifetime prevalence of about 1% in the general population. Recent studies have shown that Neuregulin-1 (Nrg1) is a candidate gene for schizophrenia. At least 15 alternative splicing of NRG1 isoforms all contain an extracellular epidermal growth factor (EGF)-like domain, which is sufficient for Nrg1 biological activity including the formation of myelin sheaths and the regulation of synaptic plasticity. It is known that Nrg1 can be cleaved by β-secretase (BACE1) and the resulting N-terminal fragment (Nrg1-ntf) binds to receptor tyrosine kinase ErbB4, which activates Nrg1/ErbB4 signaling. While changes in Nrg1 expression levels in schizophrenia still remain controversial, understanding the BACE1-cleaved Nrg1-ntf and Nrg1/ErbB4 signaling in schizophrenia neuropathogenesis is essential and important. In this review paper, we included three major parts: (1) Nrg1 structure and cleavage pattern by BACE1; (2) BACE1-dependent Nrg1 cleavage associated with schizophrenia in human studies; and (3) Animal studies of Nrg1 and BACE1 mutations with behavioral observations. Our review will provide a better understanding of Nrg1 in schizophrenia and a potential strategy for using BACE1 cleavage of Nrg1 as a unique biomarker for diagnosis, as well as a new therapeutic target, of schizophrenia.
Collapse
Affiliation(s)
- Zhengrong Zhang
- National Clinical Research Center for Mental Disorders, Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical UniversityBeijing, China
| | - Jing Huang
- National Clinical Research Center for Mental Disorders, Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical UniversityBeijing, China
| | - Yong Shen
- Neurodegenerative Disorder Research Center, School of Life Sciences, University of Science and Technology of ChinaHefei, China.,Center for Therapeutic Strategies for Brain Disorders, Roskamp Institute, SarasotaFL, United States.,Center for Hormone Advanced Science and Education, Roskamp Institute, SarasotaFL, United States
| | - Rena Li
- National Clinical Research Center for Mental Disorders, Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical UniversityBeijing, China.,Center for Therapeutic Strategies for Brain Disorders, Roskamp Institute, SarasotaFL, United States.,Center for Hormone Advanced Science and Education, Roskamp Institute, SarasotaFL, United States.,Beijing Institute for Brain Disorders, Capital Medical UniversityBeijing, China
| |
Collapse
|
27
|
Norkett R, Modi S, Kittler JT. Mitochondrial roles of the psychiatric disease risk factor DISC1. Schizophr Res 2017; 187:47-54. [PMID: 28087269 DOI: 10.1016/j.schres.2016.12.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 12/17/2016] [Accepted: 12/22/2016] [Indexed: 12/31/2022]
Abstract
Ion transport during neuronal signalling utilizes the majority of the brain's energy supply. Mitochondria are key sites for energy provision through ATP synthesis and play other important roles including calcium buffering. Thus, tightly regulated distribution and function of these organelles throughout the intricate architecture of the neuron is essential for normal synaptic communication. Therefore, delineating mechanisms coordinating mitochondrial transport and function is essential for understanding nervous system physiology and pathology. While aberrant mitochondrial transport and dynamics have long been associated with neurodegenerative disease, they have also more recently been linked to major mental illness including schizophrenia, autism and depression. However, the underlying mechanisms have yet to be elucidated, due to an incomplete understanding of the combinations of genetic and environmental factors contributing to these conditions. Consequently, the DISC1 gene has undergone intense study since its discovery at the site of a balanced chromosomal translocation, segregating with mental illness in a Scottish pedigree. The precise molecular functions of DISC1 remain elusive. Reported functions of DISC1 include regulation of intracellular signalling pathways, neuronal migration and dendritic development. Intriguingly, a role for DISC1 in mitochondrial homeostasis and transport is fast emerging. Therefore, a major function of DISC1 in regulating mitochondrial distribution, ATP synthesis and calcium buffering may be disrupted in psychiatric disease. In this review, we discuss the links between DISC1 and mitochondria, considering both trafficking of these organelles and their function, and how, via these processes, DISC1 may contribute to the regulation of neuronal behavior in normal and psychiatric disease states.
Collapse
Affiliation(s)
- R Norkett
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, UK
| | - S Modi
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, UK
| | - J T Kittler
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, UK.
| |
Collapse
|
28
|
Iwakura Y, Wang R, Inamura N, Araki K, Higashiyama S, Takei N, Nawa H. Glutamate-dependent ectodomain shedding of neuregulin-1 type II precursors in rat forebrain neurons. PLoS One 2017; 12:e0174780. [PMID: 28350885 PMCID: PMC5370147 DOI: 10.1371/journal.pone.0174780] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 03/15/2017] [Indexed: 01/30/2023] Open
Abstract
The neurotrophic factor neuregulin 1 (NRG1) regulates neuronal development, glial differentiation, and excitatory synapse maturation. NRG1 is synthesized as a membrane-anchored precursor and is then liberated by proteolytic processing or exocytosis. Mature NRG1 then binds to its receptors expressed by neighboring neurons or glial cells. However, the molecular mechanisms that govern this process in the nervous system are not defined in detail. Here we prepared neuron-enriched and glia-enriched cultures from embryonic rat neocortex to investigate the role of neurotransmitters that regulate the liberation/release of NRG1 from the membrane of neurons or glial cells. Using a two-site enzyme immunoassay to detect soluble NRG1, we show that, of various neurotransmitters, glutamate was the most potent inducer of NRG1 release in neuron-enriched cultures. NRG1 release in glia-enriched cultures was relatively limited. Furthermore, among glutamate receptor agonists, N-Methyl-D-Aspartate (NMDA) and kainate (KA), but not AMPA or tACPD, mimicked the effects of glutamate. Similar findings were acquired from analysis of the hippocampus of rats with KA-induced seizures. To evaluate the contribution of members of a disintegrin and metalloproteinase (ADAM) families to NRG1 release, we transfected primary cultures of neurons with cDNA vectors encoding NRG1 types I, II, or III precursors, each tagged with the alkaline phosphatase reporter. Analysis of alkaline phosphatase activity revealed that the NRG1 type II precursor was subjected to tumor necrosis factor-α-converting enzyme (TACE) / a Disintegrin And Metalloproteinase 17 (ADAM17) -dependent ectodomain shedding in a protein kinase C-dependent manner. These results suggest that glutamatergic neurotransmission positively regulates the ectodomain shedding of NRG1 type II precursors and liberates the active NRG1 domain in an activity-dependent manner.
Collapse
Affiliation(s)
- Yuriko Iwakura
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
- * E-mail:
| | - Ran Wang
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Naoko Inamura
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Kazuaki Araki
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Shigeki Higashiyama
- Department of Biochemistry and Molecular Genetics, Ehime University, Graduate School of Medicine, Ehime, Japan
| | - Nobuyuki Takei
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Hiroyuki Nawa
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| |
Collapse
|
29
|
Zhang Q, Esrafilzadeh D, Crook JM, Kapsa R, Stewart EM, Tomaskovic-Crook E, Wallace GG, Huang XF. Electrical Stimulation Using Conductive Polymer Polypyrrole Counters Reduced Neurite Outgrowth of Primary Prefrontal Cortical Neurons from NRG1-KO and DISC1-LI Mice. Sci Rep 2017; 7:42525. [PMID: 28198409 PMCID: PMC5309772 DOI: 10.1038/srep42525] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 01/11/2017] [Indexed: 01/07/2023] Open
Abstract
Deficits in neurite outgrowth, possibly involving dysregulation of risk genes neuregulin-1 (NRG1) and disrupted in schizophrenia 1 (DISC1) have been implicated in psychiatric disorders including schizophrenia. Electrical stimulation using conductive polymers has been shown to stimulate neurite outgrowth of differentiating human neural stem cells. This study investigated the use of the electroactive conductive polymer polypyrrole (Ppy) to counter impaired neurite outgrowth of primary pre-frontal cortical (PFC) neurons from NRG1-knock out (NRG1-KO) and DISC1-locus impairment (DISC1-LI) mice. Whereas NRG1-KO and DISC1-LI exhibited reduced neurite length and number of neurite branches compared to wild-type controls, this was not apparent for cultures on electroactive Ppy. Additionally, the use of the Ppy substrate normalised the synaptophysin and PSD95 protein and mRNA expression whereas both are usually reduced by NRG1-KO or DISC1-LI. Our findings support the utility of Ppy mediated electrical stimulation to prevent the reduction of neurite outgrowth and related synaptic protein expression in the primary PFC neurons from NRG1-KO and DISC1-LI mice, providing proof-of-concept for treating neurodevelopmental diseases including schizophrenia.
Collapse
Affiliation(s)
- Qingsheng Zhang
- Centre for Translational Neuroscience, School of Medicine, University of Wollongong, Wollongong, NSW 2522, Australia.,ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, Innovation Campus, University of Wollongong, Squires Way, Fairy Meadow, NSW 2519, Australia.,Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
| | - Dorna Esrafilzadeh
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, Innovation Campus, University of Wollongong, Squires Way, Fairy Meadow, NSW 2519, Australia.,Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
| | - Jeremy M Crook
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, Innovation Campus, University of Wollongong, Squires Way, Fairy Meadow, NSW 2519, Australia.,Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia.,Departments of Surgery, St Vincent's Hospital, The University of Melbourne, Fitzroy, VIC 3065, Australia
| | - Robert Kapsa
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, Innovation Campus, University of Wollongong, Squires Way, Fairy Meadow, NSW 2519, Australia.,Departments of Medicine, St Vincent's Hospital, The University of Melbourne, Fitzroy, VIC 3065, Australia
| | - Elise M Stewart
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, Innovation Campus, University of Wollongong, Squires Way, Fairy Meadow, NSW 2519, Australia
| | - Eva Tomaskovic-Crook
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, Innovation Campus, University of Wollongong, Squires Way, Fairy Meadow, NSW 2519, Australia.,Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
| | - Gordon G Wallace
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, Innovation Campus, University of Wollongong, Squires Way, Fairy Meadow, NSW 2519, Australia
| | - Xu-Feng Huang
- Centre for Translational Neuroscience, School of Medicine, University of Wollongong, Wollongong, NSW 2522, Australia.,Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia.,Schizophrenia Research Institute, 384 Victoria Street, Darlinghurst, NSW 2010, Australia
| |
Collapse
|
30
|
Dahoun T, Trossbach SV, Brandon NJ, Korth C, Howes OD. The impact of Disrupted-in-Schizophrenia 1 (DISC1) on the dopaminergic system: a systematic review. Transl Psychiatry 2017; 7:e1015. [PMID: 28140405 PMCID: PMC5299392 DOI: 10.1038/tp.2016.282] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 11/16/2016] [Accepted: 11/27/2016] [Indexed: 12/21/2022] Open
Abstract
Disrupted-in-Schizophrenia 1 (DISC1) is a gene known as a risk factor for mental illnesses possibly associated with dopamine impairments. DISC1 is a scaffold protein interacting with proteins involved in the dopamine system. Here we summarise the impact of DISC1 disruption on the dopamine system in animal models, considering its effects on presynaptic dopaminergic function (tyrosine hydroxylase levels, dopamine transporter levels, dopamine levels at baseline and after amphetamine administration) and postsynaptic dopaminergic function (dopamine D1 and D2 receptor levels, dopamine receptor-binding potential and locomotor activity after amphetamine administration). Our findings show that many but not all DISC1 models display (1) increased locomotion after amphetamine administration, (2) increased dopamine levels after amphetamine administration in the nucleus accumbens, and (3) inconsistent basal dopamine levels, dopamine receptor levels and binding potentials. There is also limited evidence for decreased tyrosine hydroxylase levels in the frontal cortex and increased dopamine transporter levels in the striatum but not nucleus accumbens, but these conclusions warrant further replication. The main dopaminergic findings are seen across different DISC1 models, providing convergent evidence that DISC1 has a role in regulating dopaminergic function. These results implicate dopaminergic dysregulation as a mechanism underlying the increased rate of schizophrenia seen in DISC1 variant carriers, and provide insights into how DISC1, and potentially DISC1-interacting proteins such as AKT and GSK-3, could be used as novel therapeutic targets for schizophrenia.
Collapse
Affiliation(s)
- T Dahoun
- Psychiatric Imaging Group MRC London Institute of Medical Sciences, Hammersmith Hospital, London, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College, Hammersmith Hospital, London, UK
- Department of the Institute of Clinical Sciences, Psychiatric Imaging Group, MRC London Institute of Medical Sciences (LMS), Imperial College-Hammersmith Hospital Campus, London, UK
| | - S V Trossbach
- Department of Neuropathology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - N J Brandon
- AstraZeneca Neuroscience, Innovative Medicines and Early Development Biotech Unit, R&D Boston, Waltham, MA, USA
| | - C Korth
- Department of Neuropathology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - O D Howes
- Psychiatric Imaging Group MRC London Institute of Medical Sciences, Hammersmith Hospital, London, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College, Hammersmith Hospital, London, UK
- Department of the Institute of Clinical Sciences, Psychiatric Imaging Group, MRC London Institute of Medical Sciences (LMS), Imperial College-Hammersmith Hospital Campus, London, UK
- Department of Psychosis Studies, Institute of Psychiatry, Neurology and Neuroscience (IoPPN), King's College London, London, UK
| |
Collapse
|
31
|
Mulligan KA, Cheyette BNR. Neurodevelopmental Perspectives on Wnt Signaling in Psychiatry. MOLECULAR NEUROPSYCHIATRY 2017; 2:219-246. [PMID: 28277568 DOI: 10.1159/000453266] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mounting evidence indicates that Wnt signaling is relevant to pathophysiology of diverse mental illnesses including schizophrenia, bipolar disorder, and autism spectrum disorder. In the 35 years since Wnt ligands were first described, animal studies have richly explored how downstream Wnt signaling pathways affect an array of neurodevelopmental processes and how their disruption can lead to both neurological and behavioral phenotypes. Recently, human induced pluripotent stem cell (hiPSC) models have begun to contribute to this literature while pushing it in increasingly translational directions. Simultaneously, large-scale human genomic studies are providing evidence that sequence variation in Wnt signal pathway genes contributes to pathogenesis in several psychiatric disorders. This article reviews neurodevelopmental and postneurodevelopmental functions of Wnt signaling, highlighting mechanisms, whereby its disruption might contribute to psychiatric illness, and then reviews the most reliable recent genetic evidence supporting that mutations in Wnt pathway genes contribute to psychiatric illness. We are proponents of the notion that studies in animal and hiPSC models informed by the human genetic data combined with the deep knowledge base and tool kits generated over the last several decades of basic neurodevelopmental research will yield near-term tangible advances in neuropsychiatry.
Collapse
Affiliation(s)
- Kimberly A Mulligan
- Department of Biological Sciences, California State University, Sacramento, CA, USA
| | - Benjamin N R Cheyette
- Department of Psychiatry, Kavli Institute for Fundamental Neuroscience, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
32
|
O’Tuathaigh CMP, Fumagalli F, Desbonnet L, Perez-Branguli F, Moloney G, Loftus S, O’Leary C, Petit E, Cox R, Tighe O, Clarke G, Lai D, Harvey RP, Cryan JF, Mitchell KJ, Dinan TG, Riva MA, Waddington JL. Epistatic and Independent Effects on Schizophrenia-Related Phenotypes Following Co-disruption of the Risk Factors Neuregulin-1 × DISC1. Schizophr Bull 2017; 43:214-225. [PMID: 27613806 PMCID: PMC5216856 DOI: 10.1093/schbul/sbw120] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Few studies have addressed likely gene × gene (ie, epistatic) interactions in mediating risk for schizophrenia. Using a preclinical genetic approach, we investigated whether simultaneous disruption of the risk factors Neuregulin-1 (NRG1) and Disrupted-in-schizophrenia 1 (DISC1) would produce a disease-relevant phenotypic profile different from that observed following disruption to either gene alone. NRG1 heterozygotes exhibited hyperactivity and disruption to prepulse inhibition, both reversed by antipsychotic treatment, and accompanied by reduced striatal dopamine D2 receptor protein expression, impaired social cognition, and altered glutamatergic synaptic protein expression in selected brain areas. Single gene DISC1 mutants demonstrated a disruption in social cognition and nest-building, altered brain 5-hydroxytryptamine levels and hippocampal ErbB4 expression, and decreased cortical expression of the schizophrenia-associated microRNA miR-29b. Co-disruption of DISC1 and NRG1, indicative of epistasis, evoked an impairment in sociability and enhanced self-grooming, accompanied by changes in hypothalamic oxytocin/vasopressin gene expression. The findings indicate specific behavioral correlates and underlying cellular pathways downstream of main effects of DNA variation in the schizophrenia-associated genes NRG1 and DISC1.
Collapse
Affiliation(s)
- Colm M. P. O’Tuathaigh
- School of Medicine, Brookfield Health Sciences Complex, University College Cork, Cork, Ireland;,Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin 2,Ireland;,*To whom correspondence should be addressed; School of Medicine, Brookfield Health Sciences Complex, University College Cork, Cork T12 YN60, Ireland; tel: +353-(0)21-420-5303, fax: +353-(0)21-490-1594, e-mail:
| | - Fabio Fumagalli
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Universita` degli Studi di Milano, Milan,
Italy
| | - Lieve Desbonnet
- Neurogastroenterology Laboratory, Alimentary Pharmabiotic Centre, Biosciences Institute, University College Cork, Cork, Ireland
| | - Francesc Perez-Branguli
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland;,IZKF Junior Research Group and BMBF Research Group Neuroscience, IZKF, Friedrich-Alexander-Universitaet Erlangen-Nuernberg, Erlangen, Germany
| | - Gerard Moloney
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Samim Loftus
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Claire O’Leary
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin 2,Ireland
| | - Emilie Petit
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin 2,Ireland
| | - Rachel Cox
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin 2,Ireland
| | - Orna Tighe
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin 2,Ireland
| | - Gerard Clarke
- Neurogastroenterology Laboratory, Alimentary Pharmabiotic Centre, Biosciences Institute, University College Cork, Cork, Ireland;,Department of Psychiatry, University College Cork, Cork, Ireland
| | - Donna Lai
- Victor Chang Cardiac Research Institute, Sydney, Australia
| | | | - John F. Cryan
- Neurogastroenterology Laboratory, Alimentary Pharmabiotic Centre, Biosciences Institute, University College Cork, Cork, Ireland;,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Kevin J. Mitchell
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland
| | - Timothy G. Dinan
- Neurogastroenterology Laboratory, Alimentary Pharmabiotic Centre, Biosciences Institute, University College Cork, Cork, Ireland;,Department of Psychiatry, University College Cork, Cork, Ireland
| | - Marco A. Riva
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Universita` degli Studi di Milano, Milan,
Italy
| | - John L. Waddington
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin 2,Ireland;,Jiangsu Key Laboratory of Translational Research & Therapy for Neuro-Psychiatric-Disorders and Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
33
|
Misassembly of full-length Disrupted-in-Schizophrenia 1 protein is linked to altered dopamine homeostasis and behavioral deficits. Mol Psychiatry 2016; 21:1561-1572. [PMID: 26754951 PMCID: PMC5078859 DOI: 10.1038/mp.2015.194] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 10/16/2015] [Accepted: 10/22/2015] [Indexed: 12/21/2022]
Abstract
Disrupted-in-schizophrenia 1 (DISC1) is a mental illness gene first identified in a Scottish pedigree. So far, DISC1-dependent phenotypes in animal models have been confined to expressing mutant DISC1. Here we investigated how pathology of full-length DISC1 protein could be a major mechanism in sporadic mental illness. We demonstrate that a novel transgenic rat model, modestly overexpressing the full-length DISC1 transgene, showed phenotypes consistent with a significant role of DISC1 misassembly in mental illness. The tgDISC1 rat displayed mainly perinuclear DISC1 aggregates in neurons. Furthermore, the tgDISC1 rat showed a robust signature of behavioral phenotypes that includes amphetamine supersensitivity, hyperexploratory behavior and rotarod deficits, all pointing to changes in dopamine (DA) neurotransmission. To understand the etiology of the behavioral deficits, we undertook a series of molecular studies in the dorsal striatum of tgDISC1 rats. We observed an 80% increase in high-affinity DA D2 receptors, an increased translocation of the dopamine transporter to the plasma membrane and a corresponding increase in DA inflow as observed by cyclic voltammetry. A reciprocal relationship between DISC1 protein assembly and DA homeostasis was corroborated by in vitro studies. Elevated cytosolic dopamine caused an increase in DISC1 multimerization, insolubility and complexing with the dopamine transporter, suggesting a physiological mechanism linking DISC1 assembly and dopamine homeostasis. DISC1 protein pathology and its interaction with dopamine homeostasis is a novel cellular mechanism that is relevant for behavioral control and may have a role in mental illness.
Collapse
|
34
|
Neuregulin-1 Regulates Cortical Inhibitory Neuron Dendrite and Synapse Growth through DISC1. Neural Plast 2016; 2016:7694385. [PMID: 27847649 PMCID: PMC5099462 DOI: 10.1155/2016/7694385] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Revised: 07/05/2016] [Accepted: 09/05/2016] [Indexed: 11/20/2022] Open
Abstract
Cortical inhibitory neurons play crucial roles in regulating excitatory synaptic networks and cognitive function and aberrant development of these cells have been linked to neurodevelopmental disorders. The secreted neurotrophic factor Neuregulin-1 (NRG1) and its receptor ErbB4 are established regulators of inhibitory neuron connectivity, but the developmental signalling mechanisms regulating this process remain poorly understood. Here, we provide evidence that NRG1-ErbB4 signalling functions through the multifunctional scaffold protein, Disrupted in Schizophrenia 1 (DISC1), to regulate the development of cortical inhibitory interneuron dendrite and synaptic growth. We found that NRG1 increases inhibitory neuron dendrite complexity and glutamatergic synapse formation onto inhibitory neurons and that this effect is blocked by expression of a dominant negative DISC1 mutant, or DISC1 knockdown. We also discovered that NRG1 treatment increases DISC1 expression and its localization to glutamatergic synapses being made onto cortical inhibitory neurons. Mechanistically, we determined that DISC1 binds ErbB4 within cortical inhibitory neurons. Collectively, these data suggest that a NRG1-ErbB4-DISC1 signalling pathway regulates the development of cortical inhibitory neuron dendrite and synaptic growth. Given that NRG1, ErbB4, and DISC1 are schizophrenia-linked genes, these findings shed light on how independent risk factors may signal in a common developmental pathway that contributes to neural connectivity defects and disease pathogenesis.
Collapse
|
35
|
Early postnatal GABAA receptor modulation reverses deficits in neuronal maturation in a conditional neurodevelopmental mouse model of DISC1. Mol Psychiatry 2016; 21:1449-59. [PMID: 26728564 PMCID: PMC4935661 DOI: 10.1038/mp.2015.203] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 10/21/2015] [Accepted: 11/09/2015] [Indexed: 01/28/2023]
Abstract
Exploring drug targets based on disease-associated molecular mechanisms during development is crucial for the generation of novel prevention and treatment strategies for neurodevelopmental psychiatric conditions. We report that prefrontal cortex (PFC)-specific postnatal knockdown of DISC1 via in utero electroporation combined with an inducible knockdown expression system drives deficits in synaptic GABAA function and dendritic development in pyramidal neurons, as well as abnormalities in sensorimotor gating, albeit without profound memory deficits. We show for the first time that DISC1 is specifically involved in regulating cell surface expression of α2 subunit-containing GABAA receptors in immature developing neurons, but not after full maturation. Notably, pharmacological intervention with α2/3 subtype-selective GABAA receptor positive allosteric modulators during the early postnatal period ameliorates dendritic deficits and behavioral abnormalities induced by knockdown of DISC1. These findings highlight a critical role of DISC1-mediated disruption of postnatal GABA signaling in aberrant PFC maturation and function.
Collapse
|
36
|
Increased density of DISC1-immunoreactive oligodendroglial cells in fronto-parietal white matter of patients with paranoid schizophrenia. Eur Arch Psychiatry Clin Neurosci 2016; 266:495-504. [PMID: 26315603 DOI: 10.1007/s00406-015-0640-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 08/20/2015] [Indexed: 12/11/2022]
Abstract
Profound white matter abnormalities have repeatedly been described in schizophrenia, which involve the altered expression of numerous oligodendrocyte-associated genes. Transcripts of the disrupted-in-schizophrenia 1 (DISC1) gene, a key susceptibility factor in schizophrenia, have recently been shown to be expressed by oligodendroglial cells and to negatively regulate oligodendrocyte differentiation and maturation. To learn more about the putative role(s) of oligodendroglia-associated DISC1 in schizophrenia, we analyzed the density of DISC1-immunoreactive oligodendrocytes in the fronto-parietal white matter in postmortem brains of patients with schizophrenia. Compared with controls (N = 12) and cases with undifferentiated/residual schizophrenia (N = 6), there was a significantly increased density of DISC1-expressing glial cells in paranoid schizophrenia (N = 12), which unlikely resulted from neuroleptic treatment. Pathophysiologically, over-expression of DISC1 protein(s) in white matter oligodendrocytes might add to the reduced levels of two myelin markers, 2',3'-cyclic-nucleotide 3'-phosphodiesterase and myelin basic protein in schizophrenia. Moreover, it might significantly contribute to cell cycle abnormalities as well as to deficits in oligodendroglial cell differentiation and maturation found in schizophrenia.
Collapse
|
37
|
Devine MJ, Norkett R, Kittler JT. DISC1 is a coordinator of intracellular trafficking to shape neuronal development and connectivity. J Physiol 2016; 594:5459-69. [PMID: 27121900 DOI: 10.1113/jp272187] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 03/31/2016] [Indexed: 01/14/2023] Open
Abstract
The long, asymmetric and specialised architecture of neuronal processes necessitates a properly regulated transport network of molecular motors and cytoskeletal tracks. This allows appropriate distribution of cargo for correct formation and activity of the synapse, and thus normal neuronal communication. This communication is impaired in psychiatric disease, and ongoing studies have proposed that Disrupted in schizophrenia 1 (DISC1) is an important genetic risk factor for these disorders. The mechanisms by which DISC1 dysfunction might increase propensity to psychiatric disease are not completely understood; however, an emerging theme is that DISC1 can function as a key regulator of neuronal intracellular trafficking. Transport of a wide range of potential cargoes - including mRNAs, neurotransmitter receptors, vesicles and mitochondria - can be modulated by DISC1, and therefore is susceptible to DISC1 dysfunction. This theme highlights the importance of understanding precisely how DISC1 can regulate intracellular trafficking, and suggests that a novel approach to the treatment of psychiatric disorders could be provided by targeting this protein and the trafficking machinery with which it interacts.
Collapse
Affiliation(s)
- M J Devine
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, UK
| | - R Norkett
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, UK
| | - J T Kittler
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, UK.
| |
Collapse
|
38
|
Shao L, Golbaz K, Honer WG, Beasley CL. Deficits in axon-associated proteins in prefrontal white matter in bipolar disorder but not schizophrenia. Bipolar Disord 2016; 18:342-51. [PMID: 27218831 DOI: 10.1111/bdi.12395] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 01/25/2016] [Accepted: 02/26/2016] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Brain imaging studies have implicated white matter dysfunction in the pathophysiology of both bipolar disorder (BD) and schizophrenia (SCZ). However, the contribution of axons to white matter pathology in these disorders is not yet understood. Maintenance of neuronal function is dependent on the active transport of biological material, including synaptic proteins, along the axon. In this study, the expression of six proteins associated with axonal transport of synaptic cargoes was quantified in postmortem samples of prefrontal white matter in subjects with BD, those with SCZ, and matched controls, as a measure of axonal dysfunction in these disorders. METHODS Levels of the microtubule-associated proteins β-tubulin and microtubule-associated protein 6 (MAP6), the motor and accessory proteins kinesin-1 and disrupted-in-schizophrenia 1 (DISC1), and the synaptic cargoes synaptotagmin and synaptosomal-associated protein-25 (SNAP-25) were quantified in white matter adjacent to the dorsolateral prefrontal cortex in subjects with BD (n = 34), subjects with SCZ (n = 35), and non-psychiatric controls (n = 35) using immunoblotting and an enzyme-linked immunosorbent assay (ELISA). RESULTS Protein expression of β-tubulin, kinesin-1, DISC1, synaptotagmin, and SNAP-25 was significantly lower in subjects with BD compared to controls. Levels of axon-associated proteins were also lower in subjects with SCZ, but failed to reach statistical significance. CONCLUSIONS These data provide evidence for deficits in axon-associated proteins in prefrontal white matter in BD. Findings are suggestive of decreased axonal density or dysregulation of axonal function in this disorder.
Collapse
Affiliation(s)
- Li Shao
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| | - Khashayar Golbaz
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| | - William G Honer
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| | - Clare L Beasley
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
39
|
Tomoda T, Sumitomo A, Jaaro-Peled H, Sawa A. Utility and validity of DISC1 mouse models in biological psychiatry. Neuroscience 2016; 321:99-107. [PMID: 26768401 PMCID: PMC4803604 DOI: 10.1016/j.neuroscience.2015.12.061] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Revised: 12/31/2015] [Accepted: 12/31/2015] [Indexed: 11/26/2022]
Abstract
We have seen an era of explosive progress in translating neurobiology into etiological understanding of mental disorders for the past 10-15 years. The discovery of Disrupted-in-schizophrenia 1 (DISC1) gene was one of the major driving forces that have contributed to the progress. The finding that DISC1 plays crucial roles in neurodevelopment and synapse regulation clearly underscored the utility and validity of DISC1-related biology in advancing our understanding of pathophysiological processes underlying psychiatric conditions. Despite recent genetic studies that failed to identify DISC1 as a risk gene for sporadic cases of schizophrenia, DISC1 mutant mice, coupled with various environmental stressors, have proven successful in satisfying face validity as models of a wide range of human psychiatric conditions. Investigating mental disorders using these models is expected to further contribute to the circuit-level understanding of the pathological mechanisms, as well as to the development of novel therapeutic strategies in the future.
Collapse
Affiliation(s)
- T Tomoda
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan.
| | - A Sumitomo
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - H Jaaro-Peled
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - A Sawa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
40
|
Xia M, Broek JAC, Jouroukhin Y, Schoenfelder J, Abazyan S, Jaaro-Peled H, Sawa A, Bahn S, Pletnikov M. Cell Type-Specific Effects of Mutant DISC1: A Proteomics Study. MOLECULAR NEUROPSYCHIATRY 2016; 2:28-36. [PMID: 27606318 DOI: 10.1159/000444587] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 02/08/2016] [Indexed: 12/19/2022]
Abstract
Despite the recent progress in psychiatric genetics, very few studies have focused on genetic risk factors in glial cells that, compared to neurons, can manifest different molecular pathologies underlying psychiatric disorders. In order to address this issue, we studied the effects of mutant disrupted in schizophrenia 1 (DISC1), a genetic risk factor for schizophrenia, in cultured primary neurons and astrocytes using an unbiased mass spectrometry-based proteomic approach. We found that selective expression of mutant DISC1 in neurons affects a wide variety of proteins predominantly involved in neuronal development (e.g., SOX1) and vesicular transport (Rab proteins), whereas selective expression of mutant DISC1 in astrocytes produces changes in the levels of mitochondrial (GDPM), nuclear (TMM43) and cell adhesion (ECM2) proteins. The present study demonstrates that DISC1 variants can perturb distinct molecular pathways in a cell type-specific fashion to contribute to psychiatric disorders through heterogenic effects in diverse brain cells.
Collapse
Affiliation(s)
- Meng Xia
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Md., USA; Preclinical College, Guangxi University of Chinese Medicine, Nanning, PR China
| | - Jantine A C Broek
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Yan Jouroukhin
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Md., USA
| | - Jeannine Schoenfelder
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Sofya Abazyan
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Md., USA
| | - Hanna Jaaro-Peled
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Md., USA
| | - Akira Sawa
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Md., USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Md., USA
| | - Sabine Bahn
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Mikhail Pletnikov
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Md., USA; Departments of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Md., USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Md., USA; Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Md., USA
| |
Collapse
|
41
|
Vomund S, de Souza Silva MA, Huston JP, Korth C. Behavioral Resilience and Sensitivity to Locally Restricted Cortical Migration Deficits Induced by In Utero Knockdown of Disabled-1 in the Adult Rat. Cereb Cortex 2016; 27:2052-2063. [DOI: 10.1093/cercor/bhw060] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
42
|
Enriquez-Barreto L, Morales M. The PI3K signaling pathway as a pharmacological target in Autism related disorders and Schizophrenia. MOLECULAR AND CELLULAR THERAPIES 2016; 4:2. [PMID: 26877878 PMCID: PMC4751644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 01/25/2016] [Indexed: 11/21/2023]
Abstract
This review is focused in PI3K's involvement in two widespread mental disorders: Autism and Schizophrenia. A large body of evidence points to synaptic dysfunction as a cause of these diseases, either during the initial phases of brain synaptic circuit's development or later modulating synaptic function and plasticity. Autism related disorders and Schizophrenia are complex genetic conditions in which the identification of gene markers has proved difficult, although the existence of single-gene mutations with a high prevalence in both diseases offers insight into the role of the PI3K signaling pathway. In the brain, components of the PI3K pathway regulate synaptic formation and plasticity; thus, disruption of this pathway leads to synapse dysfunction and pathological behaviors. Here, we recapitulate recent evidences that demonstrate the imbalance of several PI3K elements as leading causes of Autism and Schizophrenia, together with the plausible new pharmacological paths targeting this signaling pathway.
Collapse
Affiliation(s)
- Lilian Enriquez-Barreto
- Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Miguel Morales
- Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
43
|
Enriquez-Barreto L, Morales M. The PI3K signaling pathway as a pharmacological target in Autism related disorders and Schizophrenia. MOLECULAR AND CELLULAR THERAPIES 2016; 4:2. [PMID: 26877878 PMCID: PMC4751644 DOI: 10.1186/s40591-016-0047-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 01/25/2016] [Indexed: 01/01/2023]
Abstract
This review is focused in PI3K’s involvement in two widespread mental disorders: Autism and Schizophrenia. A large body of evidence points to synaptic dysfunction as a cause of these diseases, either during the initial phases of brain synaptic circuit’s development or later modulating synaptic function and plasticity. Autism related disorders and Schizophrenia are complex genetic conditions in which the identification of gene markers has proved difficult, although the existence of single-gene mutations with a high prevalence in both diseases offers insight into the role of the PI3K signaling pathway. In the brain, components of the PI3K pathway regulate synaptic formation and plasticity; thus, disruption of this pathway leads to synapse dysfunction and pathological behaviors. Here, we recapitulate recent evidences that demonstrate the imbalance of several PI3K elements as leading causes of Autism and Schizophrenia, together with the plausible new pharmacological paths targeting this signaling pathway.
Collapse
Affiliation(s)
- Lilian Enriquez-Barreto
- Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Miguel Morales
- Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
44
|
Deng QS, Dong XY, Wu H, Wang W, Wang ZT, Zhu JW, Liu CF, Jia WQ, Zhang Y, Schachner M, Ma QH, Xu RX. Disrupted-in-Schizophrenia-1 Attenuates Amyloid-β Generation and Cognitive Deficits in APP/PS1 Transgenic Mice by Reduction of β-Site APP-Cleaving Enzyme 1 Levels. Neuropsychopharmacology 2016; 41:440-53. [PMID: 26062786 PMCID: PMC5130120 DOI: 10.1038/npp.2015.164] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 04/27/2015] [Accepted: 05/05/2015] [Indexed: 02/05/2023]
Abstract
Disrupted-in-Schizophrenia-1 (DISC1) is a genetic risk factor for a wide range of major mental disorders, including schizophrenia, major depression, and bipolar disorders. Recent reports suggest a potential role of DISC1 in the pathogenesis of Alzheimer's disease (AD), by referring to an interaction between DISC1 and amyloid precursor protein (APP), and to an association of a single-nucleotide polymorphism in a DISC1 intron and late onset of AD. However, the function of DISC1 in AD remains unknown. In this study, decreased levels of DISC1 were observed in the cortex and hippocampus of 8-month-old APP/PS1 transgenic mice, an animal model of AD. Overexpression of DISC1 reduced, whereas knockdown of DISC1 increased protein levels, but not mRNA levels of β-site APP-Cleaving Enzyme 1 (BACE1), a key enzyme in amyloid-β (Aβ) generation. Reduction of BACE1 protein levels by overexpression of DISC1 was accompanied by an accelerating decline rate of BACE1, and was blocked by the lysosomal inhibitor chloroquine, rather than proteasome inhibitor MG-132. Moreover, overexpression of DISC1 in the hippocampus with an adeno-associated virus reduced the levels of BACE1, soluble Aβ40/42, amyloid plaque density, and rescued cognitive deficits of APP/PS1 transgenic mice. These results indicate that DISC1 attenuates Aβ generation and cognitive deficits of APP/PS1 transgenic mice through promoting lysosomal degradation of BACE1. Our findings provide new insights into the role of DISC1 in AD pathogenesis and link a potential function of DISC1 to the psychiatric symptoms of AD.
Collapse
Affiliation(s)
- Qing-Shan Deng
- Affiliated Bayi Brain Hospital and Affiliated Beijing Military Hospital of Southern Medical University, Beijing, China
| | - Xing-Yu Dong
- Affiliated Bayi Brain Hospital and Affiliated Beijing Military Hospital of Southern Medical University, Beijing, China
| | - Hao Wu
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Wang Wang
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Zhao-Tao Wang
- Affiliated Bayi Brain Hospital and Affiliated Beijing Military Hospital of Southern Medical University, Beijing, China
| | - Jian-Wei Zhu
- Affiliated Bayi Brain Hospital and Affiliated Beijing Military Hospital of Southern Medical University, Beijing, China
| | - Chun-Feng Liu
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Wei-Qiang Jia
- Affiliated Bayi Brain Hospital and Affiliated Beijing Military Hospital of Southern Medical University, Beijing, China
| | - Yan Zhang
- Affiliated Bayi Brain Hospital and Affiliated Beijing Military Hospital of Southern Medical University, Beijing, China
| | - Melitta Schachner
- Center for Neuroscience, Shantou University Medical College, Shantou, China
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Quan-Hong Ma
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and Institute, of Neuroscience, Soochow University, Blk402, Renai Road 199, Suzhou, Jiangsu 215021, China, Tel: +86 18015504376, Fax: +86 512 65880829 E-mail:
| | - Ru-Xiang Xu
- Affiliated Bayi Brain Hospital and Affiliated Beijing Military Hospital of Southern Medical University, Beijing, China
- Affiliated Bayi Brain Hospital and Affiliated Beijing Military Hospital of Southern Medical University, Beijing 100070, China, Tel: +8613391788118, Fax: +86 10 64057752, E-mail:
| |
Collapse
|
45
|
Logical design of anti-prion agents using NAGARA. Biochem Biophys Res Commun 2016; 469:930-5. [DOI: 10.1016/j.bbrc.2015.12.106] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 12/22/2015] [Indexed: 11/20/2022]
|
46
|
Interneuronal DISC1 regulates NRG1-ErbB4 signalling and excitatory-inhibitory synapse formation in the mature cortex. Nat Commun 2015; 6:10118. [PMID: 26656849 PMCID: PMC4682104 DOI: 10.1038/ncomms10118] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Accepted: 11/05/2015] [Indexed: 12/11/2022] Open
Abstract
Neuregulin-1 (NRG1) and its receptor ErbB4 influence several processes of neurodevelopment, but the mechanisms regulating this signalling in the mature brain are not well known. DISC1 is a multifunctional scaffold protein that mediates many cellular processes. Here we present a functional relationship between DISC1 and NRG1-ErbB4 signalling in mature cortical interneurons. By cell type-specific gene modulation in vitro and in vivo including in a mutant DISC1 mouse model, we demonstrate that DISC1 inhibits NRG1-induced ErbB4 activation and signalling. This effect is likely mediated by competitive inhibition of binding of ErbB4 to PSD95. Finally, we show that interneuronal DISC1 affects NRG1-ErbB4-mediated phenotypes in the fast spiking interneuron-pyramidal neuron circuit. Post-mortem brain analyses and some genetic studies have reported interneuronal deficits and involvement of the DISC1, NRG1 and ErbB4 genes in schizophrenia, respectively. Our results suggest a mechanism by which cross-talk between DISC1 and NRG1-ErbB4 signalling may contribute to these deficits. Neuregulin-1 and DISC1 signalling pathways have both been linked to neurodevelopment and schizophrenia. Here, Seshadri et al. demonstrate that DISC1 negatively regulates NRG1-induced ErbB4 signalling in adult cortical interneurons both in vitro and in vivo, possibly via competitive binding to PSD95.
Collapse
|
47
|
Boyd PJ, Cunliffe VT, Roy S, Wood JD. Sonic hedgehog functions upstream of disrupted-in-schizophrenia 1 (disc1): implications for mental illness. Biol Open 2015; 4:1336-43. [PMID: 26405049 PMCID: PMC4610215 DOI: 10.1242/bio.012005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
DISRUPTED-IN-SCHIZOPHRENIA (DISC1) has been one of the most intensively studied genetic risk factors for mental illness since it was discovered through positional mapping of a translocation breakpoint in a large Scottish family where a balanced chromosomal translocation was found to segregate with schizophrenia and affective disorders. While the evidence for it being central to disease pathogenesis in the original Scottish family is compelling, recent genome-wide association studies have not found evidence for common variants at the DISC1 locus being associated with schizophrenia in the wider population. It may therefore be the case that DISC1 provides an indication of biological pathways that are central to mental health issues and functional studies have shown that it functions in multiple signalling pathways. However, there is little information regarding factors that function upstream of DISC1 to regulate its expression and function. We herein demonstrate that Sonic hedgehog (Shh) signalling promotes expression of disc1 in the zebrafish brain. Expression of disc1 is lost in smoothened mutants that have a complete loss of Shh signal transduction, and elevated in patched mutants which have constitutive activation of Shh signalling. We previously demonstrated that disc1 knockdown has a dramatic effect on the specification of oligodendrocyte precursor cells (OPC) in the hindbrain and Shh signalling is known to be essential for the specification of these cells. We show that disc1 is prominently expressed in olig2-positive midline progenitor cells that are absent in smo mutants, while cyclopamine treatment blocks disc1 expression in these cells and mimics the effect of disc1 knock down on OPC specification. Various features of a number of psychiatric conditions could potentially arise through aberrant Hedgehog signalling. We therefore suggest that altered Shh signalling may be an important neurodevelopmental factor in the pathobiology of mental illness.
Collapse
Affiliation(s)
- Penelope J Boyd
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield S10 2HQ, UK Institute of Molecular and Cell Biology, 61 Biopolis Drive, 138673, Singapore Bateson Centre, Department of Biomedical Science, University of Sheffield, Firth Court, Western Bank, Sheffield S10 2TN, UK
| | - Vincent T Cunliffe
- Bateson Centre, Department of Biomedical Science, University of Sheffield, Firth Court, Western Bank, Sheffield S10 2TN, UK
| | - Sudipto Roy
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, 138673, Singapore Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, 117543, Singapore Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, 119288, Singapore
| | - Jonathan D Wood
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield S10 2HQ, UK Bateson Centre, Department of Biomedical Science, University of Sheffield, Firth Court, Western Bank, Sheffield S10 2TN, UK
| |
Collapse
|
48
|
Rodríguez-Seoane C, Ramos A, Korth C, Requena JR. DISC1 regulates expression of the neurotrophin VGF through the PI3K/AKT/CREB pathway. J Neurochem 2015. [DOI: 10.1111/jnc.13258] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Carmen Rodríguez-Seoane
- CIMUS Biomedical Research Institute & Department of Medicine; University of Santiago de Compostela-IDIS; Santiago de Compostela Spain
| | - Adriana Ramos
- CIMUS Biomedical Research Institute & Department of Medicine; University of Santiago de Compostela-IDIS; Santiago de Compostela Spain
- Department of Psychiatry and Behavioral Sciences; The Johns Hopkins University School of Medicine; Baltimore Maryland USA
| | - Carsten Korth
- Department of Neuropathology; Heinrich Heine University; Medical School; Düsseldorf Germany
| | - Jesús R. Requena
- CIMUS Biomedical Research Institute & Department of Medicine; University of Santiago de Compostela-IDIS; Santiago de Compostela Spain
| |
Collapse
|
49
|
DISC1 regulates trafficking and processing of APP and Aβ generation. Mol Psychiatry 2015; 20:874-9. [PMID: 25224257 PMCID: PMC4362789 DOI: 10.1038/mp.2014.100] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Revised: 06/23/2014] [Accepted: 07/14/2014] [Indexed: 12/13/2022]
Abstract
We report the novel regulation of proteolytic processing of amyloid precursor protein (APP) by DISC1, a major risk factor for psychiatric illnesses, such as depression and schizophrenia. RNAi knockdown of DISC1 in mature primary cortical neurons led to a significant increase in the levels of intracellular α-C-terminal fragment of APP (APP-CTFα) and the corresponding N-terminal-secreted ectodomain product sAPPα. DISC1 knockdown also elicited a significant decrease in the levels of amyloid beta (Aβ)42 and Aβ40. These aberrant proteolytic events were successfully rescued by co-expression of wild-type DISC1, but not by mutant DISC1 lacking the amino acids required for the interaction with APP, suggesting that APP-DISC1 protein interactions are crucial for the regulation of the C-terminal proteolysis. In a genetically engineered model in which a major full-length DISC1 isoform is depleted, consistent changes in APP processing were seen: an increase in APP-CTFα and decrease in Aβ42 and Aβ40 levels. Finally, we found that knockdown of DISC1 increased the expression of APP at the cell surface and decreased its internalization. The presented DISC1 mechanism of APP proteolytic processing and Aβ peptide generation, which is central to Alzheimer's disease pathology, suggests a novel interface between neurological and psychiatric conditions.
Collapse
|
50
|
Yamamuro K, Kimoto S, Rosen KM, Kishimoto T, Makinodan M. Potential primary roles of glial cells in the mechanisms of psychiatric disorders. Front Cell Neurosci 2015; 9:154. [PMID: 26029044 PMCID: PMC4432872 DOI: 10.3389/fncel.2015.00154] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Accepted: 04/06/2015] [Indexed: 01/05/2023] Open
Abstract
While neurons have long been considered the major player in multiple brain functions such as perception, emotion, and memory, glial cells have been relegated to a far lesser position, acting as merely a “glue” to support neurons. Multiple lines of recent evidence, however, have revealed that glial cells such as oligodendrocytes, astrocytes, and microglia, substantially impact on neuronal function and activities and are significantly involved in the underlying pathobiology of psychiatric disorders. Indeed, a growing body of evidence indicates that glial cells interact extensively with neurons both chemically (e.g., through neurotransmitters, neurotrophic factors, and cytokines) and physically (e.g., through gap junctions), supporting a role for these cells as likely significant modifiers not only of neural function in brain development but also disease pathobiology. Since questions have lingered as to whether glial dysfunction plays a primary role in the biology of neuropsychiatric disorders or a role related solely to their support of neuronal physiology in these diseases, informative and predictive animal models have been developed over the last decade. In this article, we review recent findings uncovered using glia-specific genetically modified mice with which we can evaluate both the causation of glia dysfunction and its potential role in neuropsychiatric disorders such as autism and schizophrenia.
Collapse
Affiliation(s)
- Kazuhiko Yamamuro
- Department of Psychiatry, Faculty of Medicine, Nara Medical University, Kashihara Japan
| | - Sohei Kimoto
- Department of Psychiatry, Faculty of Medicine, Nara Medical University, Kashihara Japan
| | | | - Toshifumi Kishimoto
- Department of Psychiatry, Faculty of Medicine, Nara Medical University, Kashihara Japan
| | - Manabu Makinodan
- Department of Psychiatry, Faculty of Medicine, Nara Medical University, Kashihara Japan
| |
Collapse
|