1
|
Rodríguez-Prieto Á, Mateos-White I, Aníbal-Martínez M, Navarro-González C, Gil-Sanz C, Domínguez-Canterla Y, González-Manteiga A, Del Buey Furió V, López-Bendito G, Fazzari P. Nrg1 intracellular signaling regulates the development of interhemispheric callosal axons in mice. Life Sci Alliance 2024; 7:e202302250. [PMID: 38918041 PMCID: PMC11200272 DOI: 10.26508/lsa.202302250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 06/14/2024] [Accepted: 06/14/2024] [Indexed: 06/27/2024] Open
Abstract
Schizophrenia is associated with altered cortical circuitry. Although the schizophrenia risk gene NRG1 is known to affect the wiring of inhibitory interneurons, its role in excitatory neurons and axonal development is unclear. Here, we investigated the role of Nrg1 in the development of the corpus callosum, the major interhemispheric connection formed by cortical excitatory neurons. We found that deletion of Nrg1 impaired callosal axon development in vivo. Experiments in vitro and in vivo demonstrated that Nrg1 is cell-autonomously required for axonal outgrowth and that intracellular signaling of Nrg1 is sufficient to promote axonal development in cortical neurons and specifically in callosal axons. Furthermore, our data suggest that Nrg1 signaling regulates the expression of Growth Associated Protein 43, a key regulator of axonal growth. In conclusion, our study demonstrates that NRG1 is involved in the formation of interhemispheric callosal connections and provides a novel perspective on the relevance of NRG1 in excitatory neurons and in the etiology of schizophrenia.
Collapse
Affiliation(s)
- Ángela Rodríguez-Prieto
- Lab of Cortical Circuits in Health and Disease, CIPF Centro de Investigación Príncipe, Valencia, Spain
| | - Isabel Mateos-White
- Lab of Neural Development, BIOTECMED Institute, Universidad de Valencia, Valencia, Spain
| | - Mar Aníbal-Martínez
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d'Alacant, Spain
| | - Carmen Navarro-González
- Lab of Cortical Circuits in Health and Disease, CIPF Centro de Investigación Príncipe, Valencia, Spain
- Department of Biotechnology, Universitat Politècnica de València, Valencia, Spain
| | - Cristina Gil-Sanz
- Lab of Neural Development, BIOTECMED Institute, Universidad de Valencia, Valencia, Spain
| | - Yaiza Domínguez-Canterla
- Lab of Cortical Circuits in Health and Disease, CIPF Centro de Investigación Príncipe, Valencia, Spain
| | - Ana González-Manteiga
- Lab of Cortical Circuits in Health and Disease, CIPF Centro de Investigación Príncipe, Valencia, Spain
| | - Verónica Del Buey Furió
- Lab of Cortical Circuits in Health and Disease, CIPF Centro de Investigación Príncipe, Valencia, Spain
| | - Guillermina López-Bendito
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d'Alacant, Spain
| | - Pietro Fazzari
- Lab of Cortical Circuits in Health and Disease, CIPF Centro de Investigación Príncipe, Valencia, Spain
| |
Collapse
|
2
|
Fu WY, Ip NY. The role of genetic risk factors of Alzheimer's disease in synaptic dysfunction. Semin Cell Dev Biol 2023; 139:3-12. [PMID: 35918217 DOI: 10.1016/j.semcdb.2022.07.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 07/24/2022] [Accepted: 07/25/2022] [Indexed: 12/31/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by the progressive deterioration of cognitive functions. Due to the extended global life expectancy, the prevalence of AD is increasing among aging populations worldwide. While AD is a multifactorial disease, synaptic dysfunction is one of the major neuropathological changes that occur early in AD, before clinical symptoms appear, and is associated with the progression of cognitive deterioration. However, the underlying pathological mechanisms leading to this synaptic dysfunction remains unclear. Recent large-scale genomic analyses have identified more than 40 genetic risk factors that are associated with AD. In this review, we discuss the functional roles of these genes in synaptogenesis and synaptic functions under physiological conditions, and how their functions are dysregulated in AD. This will provide insights into the contributions of these encoded proteins to synaptic dysfunction during AD pathogenesis.
Collapse
Affiliation(s)
- Wing-Yu Fu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
| | - Nancy Y Ip
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China; Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, Guangdong 518057, China.
| |
Collapse
|
3
|
Mitochondrial DNA Deficiency and Supplementation in Sus scrofa Oocytes Influence Transcriptome Profiles in Oocytes and Blastocysts. Int J Mol Sci 2023; 24:ijms24043783. [PMID: 36835193 PMCID: PMC9963854 DOI: 10.3390/ijms24043783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023] Open
Abstract
Mitochondrial DNA (mtDNA) deficiency correlates with poor oocyte quality and fertilisation failure. However, the supplementation of mtDNA deficient oocytes with extra copies of mtDNA improves fertilisation rates and embryo development. The molecular mechanisms associated with oocyte developmental incompetence, and the effects of mtDNA supplementation on embryo development are largely unknown. We investigated the association between the developmental competence of Sus scrofa oocytes, assessed with Brilliant Cresyl Blue, and transcriptome profiles. We also analysed the effects of mtDNA supplementation on the developmental transition from the oocyte to the blastocyst by longitudinal transcriptome analysis. mtDNA deficient oocytes revealed downregulation of genes associated with RNA metabolism and oxidative phosphorylation, including 56 small nucleolar RNA genes and 13 mtDNA protein coding genes. We also identified the downregulation of a large subset of genes for meiotic and mitotic cell cycle process, suggesting that developmental competence affects the completion of meiosis II and first embryonic cell division. The supplementation of oocytes with mtDNA in combination with fertilisation improves the maintenance of the expression of several key developmental genes and the patterns of parental allele-specific imprinting gene expression in blastocysts. These results suggest associations between mtDNA deficiency and meiotic cell cycle and the developmental effects of mtDNA supplementation on Sus scrofa blastocysts.
Collapse
|
4
|
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
|
5
|
Fekete CD, Nishiyama A. Presentation and integration of multiple signals that modulate oligodendrocyte lineage progression and myelination. Front Cell Neurosci 2022; 16:1041853. [PMID: 36451655 PMCID: PMC9701731 DOI: 10.3389/fncel.2022.1041853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 10/17/2022] [Indexed: 11/15/2022] Open
Abstract
Myelination is critical for fast saltatory conduction of action potentials. Recent studies have revealed that myelin is not a static structure as previously considered but continues to be made and remodeled throughout adulthood in tune with the network requirement. Synthesis of new myelin requires turning on the switch in oligodendrocytes (OL) to initiate the myelination program that includes synthesis and transport of macromolecules needed for myelin production as well as the metabolic and other cellular functions needed to support this process. A significant amount of information is available regarding the individual intrinsic and extrinsic signals that promote OL commitment, expansion, terminal differentiation, and myelination. However, it is less clear how these signals are made available to OL lineage cells when needed, and how multiple signals are integrated to generate the correct amount of myelin that is needed in a given neural network state. Here we review the pleiotropic effects of some of the extracellular signals that affect myelination and discuss the cellular processes used by the source cells that contribute to the variation in the temporal and spatial availability of the signals, and how the recipient OL lineage cells might integrate the multiple signals presented to them in a manner dialed to the strength of the input.
Collapse
Affiliation(s)
| | - Akiko Nishiyama
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States
| |
Collapse
|
6
|
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
|
7
|
Shiosaka S. Kallikrein 8: A key sheddase to strengthen and stabilize neural plasticity. Neurosci Biobehav Rev 2022; 140:104774. [PMID: 35820483 DOI: 10.1016/j.neubiorev.2022.104774] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/01/2022] [Accepted: 07/06/2022] [Indexed: 11/19/2022]
Abstract
Neural networks are modified and reorganized throughout life, even in the matured brain. Synapses in the networks form, change, or disappear dynamically in the plasticity state. The pre- and postsynaptic signaling, transmission, and structural dynamics have been studied considerably well. However, not many studies have shed light on the events in the synaptic cleft and intercellular space. Neural activity-dependent protein shedding is a phenomenon in which (1) presynaptic excitation evokes secretion or activation of sheddases, (2) sheddases are involved not only in cleavage of membrane- or matrix-bound proteins but also in mechanical modulation of cell-to-cell connectivity, and (3) freed activity domains of protein factors play a role in receptor-mediated or non-mediated biological actions. Kallikrein 8/neuropsin (KLK8) is a kallikrein family serine protease rich in the mammalian limbic brain. Accumulated evidence has suggested that KLK8 is an important modulator of neural plasticity and consequently, cognition. Insufficiency, as well as excess of KLK8 may have detrimental effects on limbic functions.
Collapse
Affiliation(s)
- Sadao Shiosaka
- Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka Prefectural Hospital Organization, Miyanosaka 3-16-21, Hirakata-shi, Osaka 573-0022, Japan.
| |
Collapse
|
8
|
Li L, Shang L, Kang W, Lingqian D, Ge S. Neuregulin‐1 promotes the proliferation, migration and angiogenesis of human periodontal ligament stem cells
in vitro. Cell Biol Int 2022; 46:792-805. [PMID: 35077607 DOI: 10.1002/cbin.11770] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 01/04/2022] [Accepted: 01/18/2022] [Indexed: 11/07/2022]
Affiliation(s)
- Ling Li
- Department of PeriodontologySchool and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration
- Department of StomatologyLinyi People's HospitalLinyiShandong ProvinceChina
| | - Lingling Shang
- Department of PeriodontologySchool and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration
| | - Wenyan Kang
- Department of PeriodontologySchool and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration
| | - Du Lingqian
- Department of StomatologyThe Second Hospital, Cheeloo College of Medicine, Shandong UniversityJinanShandong ProvinceChina
| | - Shaohua Ge
- Department of PeriodontologySchool and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration
| |
Collapse
|
9
|
Cao Q, Wei Y, Deng J, Li J, Huang Y, Li Y, Zhang JC, Zhang Z, Lin S. NRG1 accelerates the forgetting of fear memories and facilitates the induction of long-term depression in adult mice. Psychopharmacology (Berl) 2021; 238:2535-2542. [PMID: 34189597 DOI: 10.1007/s00213-021-05877-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 05/17/2021] [Indexed: 11/26/2022]
Abstract
RATIONALE Forgetting of fear memory is a current medical therapy for posttraumatic stress disorder (PTSD), and hippocampal long-term depression (LTD) may be the underlying mechanism. Neuregulin 1 (NRG1), a trophic factor, reportedly modulates memory consolidation and synaptic plasticity. METHODS Fear memory was assessed using contextual fear conditioning. Electrophysiology was used to measure LTD and GABAergic transmission in the hippocampus. OBJECTIVES To determine the contribution of hippocampal NRG1 to fear memory forgetting and low-frequency stimulation (LFS)-induced LTD. RESULTS Administration of NRG1 in the hippocampus accelerated forgetting of contextual fear memories. Furthermore, NRG1 had no effect on low-frequency stimulation-induced LTD in young mice but significantly facilitated the induction of LTD and GABAergic transmission in adult animals. More importantly, NRG1-facilitated LTD induction in adult mice could be blocked by inhibition of GABAA receptors and NMDAR activation. CONCLUSION These findings suggest a role for NRG1 in fear memory forgetting and hippocampal LTD, providing a potential target for the development of drug-assisted PTSD therapy.
Collapse
Affiliation(s)
- Qianqian Cao
- Department of Physiology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Yuan Wei
- Center for Scientific Research and Institute of Exercise and Health, Guangzhou Sports University, Guangzhou, 510500, China
| | - Jialin Deng
- Department of Physiology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Junfeng Li
- Department of Physiology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Yanhua Huang
- Department of Physiology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Yuke Li
- Department of Physiology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Ji-Chun Zhang
- Department of Physiology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Zili Zhang
- Department of Reproductive Medicine Center, The First People's Hospital of Foshan (Affiliated FoShan Hospital of Sun Yat-Sen University), Foshan, China.
| | - Song Lin
- Department of Physiology, School of Medicine, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
10
|
Navarro-Gonzalez C, Carceller H, Benito Vicente M, Serra I, Navarrete M, Domínguez-Canterla Y, Rodríguez-Prieto Á, González-Manteiga A, Fazzari P. Nrg1 haploinsufficiency alters inhibitory cortical circuits. Neurobiol Dis 2021; 157:105442. [PMID: 34246770 DOI: 10.1016/j.nbd.2021.105442] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 05/19/2021] [Accepted: 06/30/2021] [Indexed: 11/19/2022] Open
Abstract
Neuregulin 1 (NRG1) and its receptor ERBB4 are schizophrenia (SZ) risk genes that control the development of both excitatory and inhibitory cortical circuits. Most studies focused on the characterization ErbB4 deficient mice. However, ErbB4 deletion concurrently perturbs the signaling of Nrg1 and Neuregulin 3 (Nrg3), another ligand expressed in the cortex. In addition, NRG1 polymorphisms linked to SZ locate mainly in non-coding regions and they may partially reduce Nrg1 expression. Here, to study the relevance of Nrg1 partial loss-of-function in cortical circuits we characterized a recently developed haploinsufficient mouse model of Nrg1 (Nrg1tm1Lex). These mice display SZ-like behavioral deficits. The cellular and molecular underpinnings of the behavioral deficits in Nrg1tm1Lex mice remain to be established. With multiple approaches including Magnetic Resonance Spectroscopy (MRS), electrophysiology, quantitative imaging and molecular analysis we found that Nrg1 haploinsufficiency impairs the inhibitory cortical circuits. We observed changes in the expression of molecules involved in GABAergic neurotransmission, decreased density of Vglut1 excitatory buttons onto Parvalbumin interneurons and decreased frequency of spontaneous inhibitory postsynaptic currents. Moreover, we found a decreased number of Parvalbumin positive interneurons in the cortex and altered expression of Calretinin. Interestingly, we failed to detect other alterations in excitatory neurons that were previously reported in ErbB4 null mice suggesting that the Nrg1 haploinsufficiency does not entirely phenocopies ErbB4 deletions. Altogether, this study suggests that Nrg1 haploinsufficiency primarily affects the cortical inhibitory circuits in the cortex and provides new insights into the structural and molecular synaptic impairment caused by NRG1 hypofunction in a preclinical model of SZ.
Collapse
Affiliation(s)
- Carmen Navarro-Gonzalez
- Lab of Cortical Circuits in Health and Disease, CIPF Centro de Investigación Príncipe Felipe, Valencia, Spain.
| | - Héctor Carceller
- Lab of Cortical Circuits in Health and Disease, CIPF Centro de Investigación Príncipe Felipe, Valencia, Spain.
| | - Marina Benito Vicente
- Laboratorio Resonancia Magnética de Investigación, Hospital Nacional de Parapléjicos, Toledo, Spain.
| | - Irene Serra
- Consejo Superior de Investigaciones Científicas (CSIC), Instituto Cajal, Madrid, Spain.
| | - Marta Navarrete
- Consejo Superior de Investigaciones Científicas (CSIC), Instituto Cajal, Madrid, Spain.
| | - Yaiza Domínguez-Canterla
- Lab of Cortical Circuits in Health and Disease, CIPF Centro de Investigación Príncipe Felipe, Valencia, Spain.
| | - Ángela Rodríguez-Prieto
- Lab of Cortical Circuits in Health and Disease, CIPF Centro de Investigación Príncipe Felipe, Valencia, Spain.
| | - Ana González-Manteiga
- Lab of Cortical Circuits in Health and Disease, CIPF Centro de Investigación Príncipe Felipe, Valencia, Spain.
| | - Pietro Fazzari
- Lab of Cortical Circuits in Health and Disease, CIPF Centro de Investigación Príncipe Felipe, Valencia, Spain; Consejo Superior de Investigaciones Científicas (CSIC), Centro de Biología Molecular Severo Ochoa, Madrid, Spain.
| |
Collapse
|
11
|
Kim MH, Kim IB, Lee J, Cha DH, Park SM, Kim JH, Kim R, Park JS, An Y, Kim K, Kim S, Webster MJ, Kim S, Lee JH. Low-Level Brain Somatic Mutations Are Implicated in Schizophrenia. Biol Psychiatry 2021; 90:35-46. [PMID: 33867114 DOI: 10.1016/j.biopsych.2021.01.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/08/2021] [Accepted: 01/25/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Somatic mutations arising from the brain have recently emerged as significant contributors to neurodevelopmental disorders, including childhood intractable epilepsy and cortical malformations. However, whether brain somatic mutations are implicated in schizophrenia (SCZ) is not well established. METHODS We performed deep whole exome sequencing (average read depth > 550×) of matched dorsolateral prefrontal cortex and peripheral tissues from 27 patients with SCZ and 31 age-matched control individuals, followed by comprehensive and strict analysis of somatic mutations, including mutagenesis signature, substitution patterns, and involved pathways. In particular, we explored the impact of deleterious mutations in GRIN2B through primary neural culture. RESULTS We identified an average of 4.9 and 5.6 somatic mutations per exome per brain in patients with SCZ and control individuals, respectively. These mutations presented with average variant allele frequencies of 8.0% in patients with SCZ and 7.6% in control individuals. Although mutational profiles, such as the number and type of mutations, showed no significant difference between patients with SCZ and control individuals, somatic mutations in SCZ brains were significantly enriched for SCZ-related pathways, including dopamine receptor, glutamate receptor, and long-term potentiation pathways. Furthermore, we showed that brain somatic mutations in GRIN2B (encoding glutamate ionotropic NMDA receptor subunit 2B), which were found in two patients with SCZ, disrupted the location of GRIN2B across the surface of dendrites among primary cultured neurons. CONCLUSIONS Taken together, this study shows that brain somatic mutations are associated with the pathogenesis of SCZ.
Collapse
Affiliation(s)
- Myeong-Heui Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute for Science and Technology, Daejeon, Republic of Korea
| | - Il Bin Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute for Science and Technology, Daejeon, Republic of Korea; Department of Psychiatry, Hanyang University Guri Hospital, Guri, Republic of Korea
| | - Junehawk Lee
- Center for Computational Science Platform, National Institute of Supercomputing and Networking, Korea Institute of Science and Technology Information, Daejeon, Republic of Korea
| | - Do Hyeon Cha
- Graduate School of Medical Science and Engineering, Korea Advanced Institute for Science and Technology, Daejeon, Republic of Korea
| | - Sang Min Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute for Science and Technology, Daejeon, Republic of Korea
| | - Ja Hye Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute for Science and Technology, Daejeon, Republic of Korea
| | - Ryunhee Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute for Science and Technology, Daejeon, Republic of Korea
| | - Jun Sung Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute for Science and Technology, Daejeon, Republic of Korea; European Bioinformatics Institute, Hinxton, Cambridgeshire, United Kingdom
| | - Yohan An
- Graduate School of Medical Science and Engineering, Korea Advanced Institute for Science and Technology, Daejeon, Republic of Korea
| | - Kyungdeok Kim
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology, Daejeon, Republic of Korea
| | - Seyeon Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute for Science and Technology, Daejeon, Republic of Korea
| | - Maree J Webster
- Stanley Medical Research Institute, Laboratory of Brain Research, Rockville, Maryland
| | - Sanghyeon Kim
- Stanley Medical Research Institute, Laboratory of Brain Research, Rockville, Maryland.
| | - Jeong Ho Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute for Science and Technology, Daejeon, Republic of Korea; SoVarGen Inc., Daejeon, Republic of Korea.
| |
Collapse
|
12
|
Mahaman YAR, Huang F, Embaye KS, Wang X, Zhu F. The Implication of STEP in Synaptic Plasticity and Cognitive Impairments in Alzheimer's Disease and Other Neurological Disorders. Front Cell Dev Biol 2021; 9:680118. [PMID: 34195199 PMCID: PMC8236946 DOI: 10.3389/fcell.2021.680118] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 05/06/2021] [Indexed: 12/31/2022] Open
Abstract
STriatal-Enriched protein tyrosine Phosphatase (STEP) is a tyrosine phosphatase that has been implicated in Alzheimer’s disease (AD), the most common form of dementia, and many other neurological diseases. The protein level and activity of STEP have been found to be elevated in most of these disorders, and specifically in AD as a result of dysregulation of different pathways including PP2B/DARPP32/PP1, PKA as well as impairments of both proteasomal and lysosomal systems. The upregulation in STEP leads to increased binding to, and dephosphorylation of, its substrates which are mainly found to be synaptic plasticity and thus learning and memory related proteins. These proteins include kinases like Fyn, Pyk2, ERK1/2 and both NMDA and AMPA receptor subunits GluN2B and GluA2. The dephosphorylation of these molecules results in inactivation of these kinases and internalization of NMDA and AMPA receptor complexes leading to synapse loss and cognitive impairments. In this study, we aim to review STEP regulation and its implications in AD as well as other neurological disorders and then summarize data on targeting STEP as therapeutic strategy in these diseases.
Collapse
Affiliation(s)
- Yacoubou Abdoul Razak Mahaman
- Cognitive Impairment Ward of Neurology Department, The Third Affiliated Hospital, Shenzhen University, Shenzhen, China.,Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Huang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kidane Siele Embaye
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaochuan Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Feiqi Zhu
- Cognitive Impairment Ward of Neurology Department, The Third Affiliated Hospital, Shenzhen University, Shenzhen, China
| |
Collapse
|
13
|
Srivastava S, Ahmad R, Khare SK. Alzheimer's disease and its treatment by different approaches: A review. Eur J Med Chem 2021; 216:113320. [PMID: 33652356 DOI: 10.1016/j.ejmech.2021.113320] [Citation(s) in RCA: 181] [Impact Index Per Article: 60.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 02/04/2021] [Accepted: 02/13/2021] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that impairs mental ability development and interrupts neurocognitive function. This neuropathological condition is depicted by neurodegeneration, neural loss, and development of neurofibrillary tangles and Aβ plaques. There is also a greater risk of developing AD at a later age for people with cardiovascular diseases, hypertension and diabetes. In the biomedical sciences, effective treatment for Alzheimer's disease is a severe obstacle. There is no such treatment to cure Alzheimer's disease. The drug present in the market show only symptomatic relief. The cause of Alzheimer's disease is not fully understood and the blood-brain barrier restricts drug efficacy are two main factors that hamper research. Stem cell-based therapy has been seen as an effective, secure, and creative therapeutic solution to overcoming AD because of AD's multifactorial nature and inadequate care. Current developments in nanotechnology often offer possibilities for the delivery of active drug candidates to address certain limitations. The key nanoformulations being tested against AD include polymeric nanoparticles (NP), inorganic NPs and lipid-based NPs. Nano drug delivery systems are promising vehicles for targeting several therapeutic moieties by easing drug molecules' penetration across the CNS and improving their bioavailability. In this review, we focus on the causes of the AD and their treatment by different approaches.
Collapse
Affiliation(s)
- Sukriti Srivastava
- Enzyme and Microbial Biochemistry Laboratory, Department of Chemistry, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Razi Ahmad
- Enzyme and Microbial Biochemistry Laboratory, Department of Chemistry, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Sunil Kumar Khare
- Enzyme and Microbial Biochemistry Laboratory, Department of Chemistry, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India.
| |
Collapse
|
14
|
van Gastel J, Leysen H, Boddaert J, Vangenechten L, Luttrell LM, Martin B, Maudsley S. Aging-related modifications to G protein-coupled receptor signaling diversity. Pharmacol Ther 2020; 223:107793. [PMID: 33316288 DOI: 10.1016/j.pharmthera.2020.107793] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 11/26/2020] [Indexed: 02/06/2023]
Abstract
Aging is a highly complex molecular process, affecting nearly all tissue systems in humans and is the highest risk factor in developing neurodegenerative disorders such as Alzheimer's and Parkinson's disease, cardiovascular disease and Type 2 diabetes mellitus. The intense complexity of the aging process creates an incentive to develop more specific drugs that attenuate or even reverse some of the features of premature aging. As our current pharmacopeia is dominated by therapeutics that target members of the G protein-coupled receptor (GPCR) superfamily it may be prudent to search for effective anti-aging therapeutics in this fertile domain. Since the first demonstration of GPCR-based β-arrestin signaling, it has become clear that an enhanced appreciation of GPCR signaling diversity may facilitate the creation of therapeutics with selective signaling activities. Such 'biased' ligand signaling profiles can be effectively investigated using both standard molecular biological techniques as well as high-dimensionality data analyses. Through a more nuanced appreciation of the quantitative nature across the multiple dimensions of signaling bias that drugs possess, researchers may be able to further refine the efficacy of GPCR modulators to impact the complex aberrations that constitute the aging process. Identifying novel effector profiles could expand the effective pharmacopeia and assist in the design of precision medicines. This review discusses potential non-G protein effectors, and specifically their potential therapeutic suitability in aging and age-related disorders.
Collapse
Affiliation(s)
- Jaana van Gastel
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium; Faculty of Pharmacy, Biomedical and Veterinary Science, University of Antwerp, Antwerp, Belgium
| | - Hanne Leysen
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium; Faculty of Pharmacy, Biomedical and Veterinary Science, University of Antwerp, Antwerp, Belgium
| | - Jan Boddaert
- Molecular Pathology Group, Faculty of Medicine and Health Sciences, Laboratory of Cell Biology and Histology, Antwerp, Belgium
| | - Laura Vangenechten
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Louis M Luttrell
- Division of Endocrinology, Diabetes & Medical Genetics, Medical University of South Carolina, USA
| | - Bronwen Martin
- Faculty of Pharmacy, Biomedical and Veterinary Science, University of Antwerp, Antwerp, Belgium
| | - Stuart Maudsley
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium; Faculty of Pharmacy, Biomedical and Veterinary Science, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
15
|
Rajasekaran A, Shivakumar V, Kalmady SV, Parlikar R, Chhabra H, Prabhu A, Subbanna M, Venugopal D, Amaresha AC, Agarwal SM, Bose A, Narayanaswamy JC, Debnath M, Venkatasubramanian G. Impact of NRG1 HapICE gene variants on digit ratio and dermatoglyphic measures in schizophrenia. Asian J Psychiatr 2020; 54:102363. [PMID: 33271685 DOI: 10.1016/j.ajp.2020.102363] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 08/24/2020] [Indexed: 12/14/2022]
Abstract
Multiple lines of evidence have suggested a potential role of Neuregulin-1 (NRG1) in the neurodevelopmental pathogenesis of schizophrenia. Interaction between genetic risk variants present within NRG1 locus and non-specific gestational putative insults can significantly impair crucial processes of brain development. Such genetic effects can be analyzed through the assessment of digit ratio and dermatoglyphic patterns. We examined the role of two well-replicated polymorphisms of NRG1 (SNP8NRG221533 and SNP8NRG243177) on schizophrenia risk and its probable impact on the digit ratio and dermatoglyphic measures in patients (N = 221) and healthy controls (N = 200). In schizophrenia patients, but not in healthy controls, a significant association between NRG1 SNP8NRG221533 C/C genotype with lower left 2D:4D ratio, as well as with higher FA_TbcRC and DA_TbcRC. The substantial effect of SNP8NRG221533 on both digit ratio and dermatoglyphic measures suggest a potential role for NRG1 gene variants on neurodevelopmental pathogenesis of schizophrenia.
Collapse
Affiliation(s)
- Ashwini Rajasekaran
- Translational Psychiatry Laboratory, Neurobiology Research Center, National Institute of Mental Health and Neurosciences, Bangalore, India; Department of Human Genetics, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Venkataram Shivakumar
- Translational Psychiatry Laboratory, Neurobiology Research Center, National Institute of Mental Health and Neurosciences, Bangalore, India; InSTAR Program, Schizophrenia Clinic, Department of Psychiatry, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Sunil V Kalmady
- Canadian VIGOUR Centre, University of Alberta, Edmonton, AB, Canada
| | - Rujuta Parlikar
- Translational Psychiatry Laboratory, Neurobiology Research Center, National Institute of Mental Health and Neurosciences, Bangalore, India; InSTAR Program, Schizophrenia Clinic, Department of Psychiatry, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Harleen Chhabra
- Translational Psychiatry Laboratory, Neurobiology Research Center, National Institute of Mental Health and Neurosciences, Bangalore, India; InSTAR Program, Schizophrenia Clinic, Department of Psychiatry, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Ananya Prabhu
- Translational Psychiatry Laboratory, Neurobiology Research Center, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Manjula Subbanna
- Translational Psychiatry Laboratory, Neurobiology Research Center, National Institute of Mental Health and Neurosciences, Bangalore, India; Department of Human Genetics, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Deepthi Venugopal
- Translational Psychiatry Laboratory, Neurobiology Research Center, National Institute of Mental Health and Neurosciences, Bangalore, India; Department of Human Genetics, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Anekal C Amaresha
- Translational Psychiatry Laboratory, Neurobiology Research Center, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Sri Mahavir Agarwal
- Translational Psychiatry Laboratory, Neurobiology Research Center, National Institute of Mental Health and Neurosciences, Bangalore, India; InSTAR Program, Schizophrenia Clinic, Department of Psychiatry, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Anushree Bose
- Translational Psychiatry Laboratory, Neurobiology Research Center, National Institute of Mental Health and Neurosciences, Bangalore, India; InSTAR Program, Schizophrenia Clinic, Department of Psychiatry, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Janardhanan C Narayanaswamy
- Translational Psychiatry Laboratory, Neurobiology Research Center, National Institute of Mental Health and Neurosciences, Bangalore, India; InSTAR Program, Schizophrenia Clinic, Department of Psychiatry, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Monojit Debnath
- Translational Psychiatry Laboratory, Neurobiology Research Center, National Institute of Mental Health and Neurosciences, Bangalore, India; Department of Human Genetics, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Ganesan Venkatasubramanian
- Translational Psychiatry Laboratory, Neurobiology Research Center, National Institute of Mental Health and Neurosciences, Bangalore, India; InSTAR Program, Schizophrenia Clinic, Department of Psychiatry, National Institute of Mental Health and Neurosciences, Bangalore, India.
| |
Collapse
|
16
|
Ledonne A, Mercuri NB. On the Modulatory Roles of Neuregulins/ErbB Signaling on Synaptic Plasticity. Int J Mol Sci 2019; 21:ijms21010275. [PMID: 31906113 PMCID: PMC6981567 DOI: 10.3390/ijms21010275] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 12/27/2019] [Accepted: 12/29/2019] [Indexed: 12/14/2022] Open
Abstract
Neuregulins (NRGs) are a family of epidermal growth factor-related proteins, acting on tyrosine kinase receptors of the ErbB family. NRGs play an essential role in the development of the nervous system, since they orchestrate vital functions such as cell differentiation, axonal growth, myelination, and synapse formation. They are also crucially involved in the functioning of adult brain, by directly modulating neuronal excitability, neurotransmission, and synaptic plasticity. Here, we provide a review of the literature documenting the roles of NRGs/ErbB signaling in the modulation of synaptic plasticity, focusing on evidence reported in the hippocampus and midbrain dopamine (DA) nuclei. The emerging picture shows multifaceted roles of NRGs/ErbB receptors, which critically modulate different forms of synaptic plasticity (LTP, LTD, and depotentiation) affecting glutamatergic, GABAergic, and DAergic synapses, by various mechanisms. Further, we discuss the relevance of NRGs/ErbB-dependent synaptic plasticity in the control of brain processes, like learning and memory and the known involvement of NRGs/ErbB signaling in the modulation of synaptic plasticity in brain’s pathological conditions. Current evidence points to a central role of NRGs/ErbB receptors in controlling glutamatergic LTP/LTD and GABAergic LTD at hippocampal CA3–CA1 synapses, as well as glutamatergic LTD in midbrain DA neurons, thus supporting that NRGs/ErbB signaling is essential for proper brain functions, cognitive processes, and complex behaviors. This suggests that dysregulated NRGs/ErbB-dependent synaptic plasticity might contribute to mechanisms underlying different neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Ada Ledonne
- Department of Experimental Neuroscience, Santa Lucia Foundation, Via del Fosso di Fiorano, no 64, 00143 Rome, Italy;
- Correspondence: ; Tel.: +3906-501703160; Fax: +3906-501703307
| | - Nicola B. Mercuri
- Department of Experimental Neuroscience, Santa Lucia Foundation, Via del Fosso di Fiorano, no 64, 00143 Rome, Italy;
- Department of Systems Medicine, University of Rome “Tor Vergata”, Via Montpellier no 1, 00133 Rome, Italy
| |
Collapse
|
17
|
Zieba J, Morris MJ, Karl T. Behavioural effects of high fat diet exposure starting in late adolescence in neuregulin 1 transmembrane domain mutant mice. Behav Brain Res 2019; 373:112074. [DOI: 10.1016/j.bbr.2019.112074] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 07/04/2019] [Accepted: 07/05/2019] [Indexed: 12/17/2022]
|
18
|
Wang Y, Wang P, Chen F, Lulu M, Huang S, Liu Z. Potential synaptic plasticity-based Shenzhiling oral liquid for a SAD Mouse Model. Brain Behav 2019; 9:e01385. [PMID: 31429527 PMCID: PMC6749598 DOI: 10.1002/brb3.1385] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 07/02/2019] [Accepted: 07/03/2019] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Synaptic plasticity is the basis of memory formation. The pathological manifestations of abnormal glucose metabolism in the nervous system of sporadic Alzheimer's disease (SAD) may affect synaptic plasticity, thus causing memory damage. As a traditional Chinese medicine compound preparation, the mechanism by which Shenzhiling (SZL) oral liquid can alleviate the cognitive impairment of SAD by improving synaptic plasticity remains unclear. OBJECTIVE This article mainly discusses whether SZL can exert a protective synaptic effect as mediated by glutamate receptors and glycogen synthesis kinase 3β (GSK3β); further, it discusses whether SZL can improve cognitive function in SAD. METHODS C57BL/6 mice were used as a SAD model after injection with streptozotocin (STZ) into the bilateral lateral ventricles; mice of the same background were injected with artificial cerebrospinal fluid into bilateral ventricles and were used as a control group. After 3 months of exposure to the intervention, the step-down test was carried out. Western blot was used to detect the levels of NMDAR2B, p-NMDAR2B, mGlu5, GSK3β, and p-GSK3β in the hippocampus of mice. Immunohistochemical analysis was used to observe the number of GSK3β-positive cells in the CA1 region of mouse hippocampus. RESULTS The memory retention ability of mice was significantly improved after 3 months of SZL treatment, and the expression levels of NMDAR2B, mGlu5, and GSK3β were significantly changed. CONCLUSION Shenzhiling provides a potential for the treatment for SAD with traditional Chinese medicine.
Collapse
Affiliation(s)
- Yahan Wang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, China.,Key Laboratory of Pharmacology of Dongzhimen Hospital (BUCM), State Administration of Traditional Chinese Medicine, Beijing, China
| | - Pengwen Wang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, China.,Key Laboratory of Pharmacology of Dongzhimen Hospital (BUCM), State Administration of Traditional Chinese Medicine, Beijing, China
| | - Fang Chen
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, China.,Key Laboratory of Pharmacology of Dongzhimen Hospital (BUCM), State Administration of Traditional Chinese Medicine, Beijing, China.,Rehabilitation centre, The Xinjiang Uygur Autonomous Region Traditional Chinese Medicine Hospital affiliated to Xinjiang Medical University, Urumqi, China
| | - Mana Lulu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, China.,Key Laboratory of Pharmacology of Dongzhimen Hospital (BUCM), State Administration of Traditional Chinese Medicine, Beijing, China
| | - Shuaiyang Huang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, China.,Key Laboratory of Pharmacology of Dongzhimen Hospital (BUCM), State Administration of Traditional Chinese Medicine, Beijing, China
| | - Zhenhong Liu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, China.,Key Laboratory of Pharmacology of Dongzhimen Hospital (BUCM), State Administration of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
19
|
Kizner V, Naujock M, Fischer S, Jäger S, Reich S, Schlotthauer I, Zuckschwerdt K, Geiger T, Hildebrandt T, Lawless N, Macartney T, Dorner-Ciossek C, Gillardon F. CRISPR/Cas9-mediated Knockout of the Neuropsychiatric Risk Gene KCTD13 Causes Developmental Deficits in Human Cortical Neurons Derived from Induced Pluripotent Stem Cells. Mol Neurobiol 2019; 57:616-634. [PMID: 31402430 DOI: 10.1007/s12035-019-01727-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 07/31/2019] [Indexed: 02/08/2023]
Abstract
The human KCTD13 gene is located within the 16p11.2 locus and copy number variants of this locus are associated with a high risk for neuropsychiatric diseases including autism spectrum disorder and schizophrenia. Studies in zebrafish point to a role of KCTD13 in proliferation of neural precursor cells which may contribute to macrocephaly in 16p11.2 deletion carriers. KCTD13 is highly expressed in the fetal human brain and in mouse cortical neurons, but its contribution to the development and function of mammalian neurons is not completely understood. In the present study, we deleted the KCTD13 gene in human-induced pluripotent stem cells (iPSCs) using CRISPR/Cas9 nickase. Following neural differentiation of KCTD13 deficient and isogenic control iPSC lines, we detected a moderate but significant inhibition of DNA synthesis and proliferation in KCTD13 deficient human neural precursor cells. KCTD13 deficient cortical neurons derived from iPSCs showed decreased neurite formation and reduced spontaneous network activity. RNA-sequencing and pathway analysis pointed to a role for ERBB signaling in these phenotypic changes. Consistently, activating and inhibiting ERBB kinases rescued and aggravated, respectively, impaired neurite formation. In contrast to findings in non-neuronal human HeLa cells, we did not detect an accumulation of the putative KCTD13/Cullin-3 substrate RhoA, and treatment with inhibitors of RhoA signaling did not rescue decreased neurite formation in human KCTD13 knockout neurons. Taken together, our data provide insight into the role of KCTD13 in neurodevelopmental disorders, and point to ERBB signaling as a potential target for neuropsychiatric disorders associated with KCTD13 deficiency.
Collapse
Affiliation(s)
- Valeria Kizner
- CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co KG, 88397, Biberach an der Riss, Germany
| | - Maximilian Naujock
- CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co KG, 88397, Biberach an der Riss, Germany
| | - Sandra Fischer
- CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co KG, 88397, Biberach an der Riss, Germany
| | - Stefan Jäger
- CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co KG, 88397, Biberach an der Riss, Germany
| | - Selina Reich
- CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co KG, 88397, Biberach an der Riss, Germany
| | - Ines Schlotthauer
- CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co KG, 88397, Biberach an der Riss, Germany
| | - Kai Zuckschwerdt
- Target Discovery Research, Boehringer Ingelheim Pharma GmbH & Co KG, 88397, Biberach an der Riss, Germany
| | - Tobias Geiger
- Cardio-metabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co KG, 88397, Biberach an der Riss, Germany
| | - Tobias Hildebrandt
- Target Discovery Research, Boehringer Ingelheim Pharma GmbH & Co KG, 88397, Biberach an der Riss, Germany
| | - Nathan Lawless
- Target Discovery Research, Boehringer Ingelheim Pharma GmbH & Co KG, 88397, Biberach an der Riss, Germany
| | - Thomas Macartney
- MRC Protein Phosphorylation and Ubiquitylation Unit, Sir James Black Centre, University of Dundee, Dundee, DD1 5EH, UK
| | - Cornelia Dorner-Ciossek
- CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co KG, 88397, Biberach an der Riss, Germany
| | - Frank Gillardon
- CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co KG, 88397, Biberach an der Riss, Germany.
| |
Collapse
|
20
|
Kataria H, Alizadeh A, Karimi-Abdolrezaee S. Neuregulin-1/ErbB network: An emerging modulator of nervous system injury and repair. Prog Neurobiol 2019; 180:101643. [PMID: 31229498 DOI: 10.1016/j.pneurobio.2019.101643] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 06/07/2019] [Accepted: 06/11/2019] [Indexed: 12/20/2022]
Abstract
Neuregulin-1 (Nrg-1) is a member of the Neuregulin family of growth factors with essential roles in the developing and adult nervous system. Six different types of Nrg-1 (Nrg-1 type I-VI) and over 30 isoforms have been discovered; however, their specific roles are not fully determined. Nrg-1 signals through a complex network of protein-tyrosine kinase receptors, ErbB2, ErbB3, ErbB4 and multiple intracellular pathways. Genetic and pharmacological studies of Nrg-1 and ErbB receptors have identified a critical role for Nrg-1/ErbB network in neurodevelopment including neuronal migration, neural differentiation, myelination as well as formation of synapses and neuromuscular junctions. Nrg-1 signaling is best known for its characterized role in development and repair of the peripheral nervous system (PNS) due to its essential role in Schwann cell development, survival and myelination. However, our knowledge of the impact of Nrg-1/ErbB on the central nervous system (CNS) has emerged in recent years. Ongoing efforts have uncovered a multi-faceted role for Nrg-1 in regulating CNS injury and repair processes. In this review, we provide a timely overview of the most recent updates on Nrg-1 signaling and its role in nervous system injury and diseases. We will specifically highlight the emerging role of Nrg-1 in modulating the glial and immune responses and its capacity to foster neuroprotection and remyelination in CNS injury. Nrg-1/ErbB network is a key regulatory pathway in the developing nervous system; therefore, unraveling its role in neuropathology and repair can aid in development of new therapeutic approaches for nervous system injuries and associated disorders.
Collapse
Affiliation(s)
- Hardeep Kataria
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Arsalan Alizadeh
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Soheila Karimi-Abdolrezaee
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
21
|
Grieco SF, Holmes TC, Xu X. Neuregulin directed molecular mechanisms of visual cortical plasticity. J Comp Neurol 2019; 527:668-678. [PMID: 29464684 PMCID: PMC6103898 DOI: 10.1002/cne.24414] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 01/18/2018] [Accepted: 01/18/2018] [Indexed: 02/06/2023]
Abstract
Experience-dependent critical period (CP) plasticity has been extensively studied in the visual cortex. Monocular deprivation during the CP affects ocular dominance, limits visual performance, and contributes to the pathological etiology of amblyopia. Neuregulin-1 (NRG1) signaling through its tyrosine kinase receptor ErbB4 is essential for the normal development of the nervous system and has been linked to neuropsychiatric disorders such as schizophrenia. We discovered recently that NRG1/ErbB4 signaling in PV neurons is critical for the initiation of CP visual cortical plasticity by controlling excitatory synaptic inputs onto PV neurons and thus PV-cell mediated cortical inhibition that occurs following visual deprivation. Building on this discovery, we review the existing literature of neuregulin signaling in developing and adult cortex and address the implication of NRG/ErbB4 signaling in visual cortical plasticity at the cellular and circuit levels. NRG-directed research may lead to therapeutic approaches to reactivate plasticity in the adult cortex.
Collapse
Affiliation(s)
- Steven F Grieco
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, California
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, California
- Department of Biomedical Engineering, University of California, Irvine, California
- Department of Microbiology and Molecular Genetics, University of California, Irvine, California
| | - Todd C Holmes
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, California
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, California
- Department of Biomedical Engineering, University of California, Irvine, California
- Department of Microbiology and Molecular Genetics, University of California, Irvine, California
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, California
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, California
- Department of Biomedical Engineering, University of California, Irvine, California
- Department of Microbiology and Molecular Genetics, University of California, Irvine, California
| |
Collapse
|
22
|
Bygrave AM, Kilonzo K, Kullmann DM, Bannerman DM, Kätzel D. Can N-Methyl-D-Aspartate Receptor Hypofunction in Schizophrenia Be Localized to an Individual Cell Type? Front Psychiatry 2019; 10:835. [PMID: 31824347 PMCID: PMC6881463 DOI: 10.3389/fpsyt.2019.00835] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 10/21/2019] [Indexed: 01/07/2023] Open
Abstract
Hypofunction of N-methyl-D-aspartate glutamate receptors (NMDARs), whether caused by endogenous factors like auto-antibodies or mutations, or by pharmacological or genetic manipulations, produces a wide variety of deficits which overlap with-but do not precisely match-the symptom spectrum of schizophrenia. In order to understand how NMDAR hypofunction leads to different components of the syndrome, it is necessary to take into account which neuronal subtypes are particularly affected by it in terms of detrimental functional alterations. We provide a comprehensive overview detailing findings in rodent models with cell type-specific knockout of NMDARs. Regarding inhibitory cortical cells, an emerging model suggests that NMDAR hypofunction in parvalbumin (PV) positive interneurons is a potential risk factor for this disease. PV interneurons display a selective vulnerability resulting from a combination of genetic, cellular, and environmental factors that produce pathological multi-level positive feedback loops. Central to this are two antioxidant mechanisms-NMDAR activity and perineuronal nets-which are themselves impaired by oxidative stress, amplifying disinhibition. However, NMDAR hypofunction in excitatory pyramidal cells also produces a range of schizophrenia-related deficits, in particular maladaptive learning and memory recall. Furthermore, NMDAR blockade in the thalamus disturbs thalamocortical communication, and NMDAR ablation in dopaminergic neurons may provoke over-generalization in associative learning, which could relate to the positive symptom domain. Therefore, NMDAR hypofunction can produce schizophrenia-related effects through an action on various different circuits and cell types.
Collapse
Affiliation(s)
- Alexei M Bygrave
- Department of Neuroscience, Johns Hopkins University, Baltimore, MD, United States
| | - Kasyoka Kilonzo
- Institute of Applied Physiology, Ulm University, Ulm, Germany
| | - Dimitri M Kullmann
- UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - David M Bannerman
- Department of Experimental Psychology, University of Oxford, Oxford, United Kingdom
| | - Dennis Kätzel
- Institute of Applied Physiology, Ulm University, Ulm, Germany
| |
Collapse
|
23
|
Olaya JC, Heusner CL, Matsumoto M, Shannon Weickert C, Karl T. Schizophrenia-relevant behaviours of female mice overexpressing neuregulin 1 type III. Behav Brain Res 2018; 353:227-235. [DOI: 10.1016/j.bbr.2018.03.026] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 03/15/2018] [Accepted: 03/15/2018] [Indexed: 12/20/2022]
|
24
|
Ledonne A, Mercuri NB. mGluR1-Dependent Long Term Depression in Rodent Midbrain Dopamine Neurons Is Regulated by Neuregulin 1/ErbB Signaling. Front Mol Neurosci 2018; 11:346. [PMID: 30327588 PMCID: PMC6174199 DOI: 10.3389/fnmol.2018.00346] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 09/04/2018] [Indexed: 11/22/2022] Open
Abstract
Increasing evidence demonstrates that the neurotrophic factor Neuregulin 1 (NRG1) and its receptors, ErbB tyrosine kinases, modulate midbrain dopamine (DA) transmission. We have previously reported that NRG1/ErbB signaling is essential for proper metabotropic glutamate receptors 1 (mGluR1) functioning in midbrain DA neurons, thus the functional interaction between ErbB receptors and mGluR1 regulates neuronal excitation and in vivo striatal DA release. While it is widely recognized that mGluR1 play a pivotal role in long-term modifications of synaptic transmission in several brain areas, specific mGluR1-dependent forms of synaptic plasticity in substantia nigra pars compacta (SNpc) DA neurons have not been described yet. Here, first we aimed to detect and characterize mGluR1-dependent glutamatergic long-term depression (LTD) in SNpc DA neurons. Second, we tested the hypothesis that endogenous ErbB signaling, by affecting mGluR1, fine-tunes glutamatergic synaptic plasticity in DA cells. We found that either pharmacological or synaptic activation of mGluR1 causes an LTD of AMPAR-mediated transmission in SNpc DA neurons from mice and rat slices, which is reliant on endogenous NRG1/ErbB signaling. Indeed, LTD is counteracted by a broad spectrum ErbB inhibitor. Moreover, the intracellular injection of pan-ErbB- or ErbB2 inhibitors inside DA neurons reduces mGluR1-dependent LTD, suggesting an involvement of ErbB2/ErbB4-containing receptors. Interestingly, exogenous NRG1 fosters LTD expression during minimal mGluRI activation. These results enlarge our cognizance on mGluR1 relevance in the induction of a novel form of long-term synaptic plasticity in SNpc DA neurons and describe a new NRG1/ErbB-dependent mechanism shaping glutamatergic transmission in DA cells. This might have important implications either in DA-dependent behaviors and learning/memory processes or in DA-linked diseases.
Collapse
Affiliation(s)
- Ada Ledonne
- Department of Experimental Neuroscience, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Nicola Biagio Mercuri
- Department of Experimental Neuroscience, IRCCS Santa Lucia Foundation, Rome, Italy.,Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
25
|
Cao SX, Zhang Y, Hu XY, Hong B, Sun P, He HY, Geng HY, Bao AM, Duan SM, Yang JM, Gao TM, Lian H, Li XM. ErbB4 deletion in noradrenergic neurons in the locus coeruleus induces mania-like behavior via elevated catecholamines. eLife 2018; 7:39907. [PMID: 30179154 PMCID: PMC6185106 DOI: 10.7554/elife.39907] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 09/02/2018] [Indexed: 11/13/2022] Open
Abstract
Dysfunction of the noradrenergic (NE) neurons is implicated in the pathogenesis of bipolar disorder (BPD). ErbB4 is highly expressed in NE neurons, and its genetic variation has been linked to BPD; however, how ErbB4 regulates NE neuronal function and contributes to BPD pathogenesis is unclear. Here we find that conditional deletion of ErbB4 in locus coeruleus (LC) NE neurons increases neuronal spontaneous firing through NMDA receptor hyperfunction, and elevates catecholamines in the cerebrospinal fluid (CSF). Furthermore, Erbb4-deficient mice present mania-like behaviors, including hyperactivity, reduced anxiety and depression, and increased sucrose preference. These behaviors are completely rescued by the anti-manic drug lithium or antagonists of catecholaminergic receptors. Our study demonstrates the critical role of ErbB4 signaling in regulating LC-NE neuronal function, reinforcing the view that dysfunction of the NE system may contribute to the pathogenesis of mania-associated disorder. Bipolar disorder is a mental illness that affects roughly 1 in 100 people worldwide. It features periods of depression interspersed with episodes of mania – a state of delusion, heightened excitation and increased activity. Evidence suggests that changes in a brain region called the locus coeruleus contribute to bipolar disorder. Cells within this area produce a chemical called norepinephrine, whose levels increase during mania and decrease during depression. But it is unclear exactly how norepinephrine-producing cells, also known as noradrenergic cells, contribute to bipolar disorder. The answer may lie in a protein called ErbB4, which is found within the outer membrane of many noradrenergic neurons. ErbB4 is active in both the developing and adult brain, and certain people with bipolar disorder have mutations in the gene that codes for the protein. Might changes in ErbB4 disrupt the activity of noradrenergic neurons? And could these changes increase the risk of bipolar disorder? To find out, Cao, Zhang et al. deleted the gene for ErbB4 from noradrenergic neurons in the locus coeruleus of mice. The mutant mice showed mania-like behaviors: compared to normal animals, they were hyperactive, less anxious, and consumed more of a sugary solution. Treating the mice with lithium, a medication used in bipolar disorder, reversed these changes and made the rodents behave more like non-mutant animals. Further experiments revealed that noradrenergic neurons in the mutant mice showed increased spontaneous activity. These animals also had more of the chemicals noradrenaline and dopamine in the fluid circulating around their brains and spinal cords. The results thus suggest that losing ErbB4 enhances the spontaneous firing of noradrenergic neurons in the locus coeruleus. This increases release of noradrenaline and dopamine, which in turn leads to mania-like behaviors. Future research should examine whether drugs that target ErbB4 could treat mania and improve the lives of people with bipolar disorder and related conditions.
Collapse
Affiliation(s)
- Shu-Xia Cao
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ying Zhang
- Center for Neuroscience, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, China
| | - Xing-Yue Hu
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Bin Hong
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Peng Sun
- Center for Neuroscience, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, China
| | - Hai-Yang He
- Center for Neuroscience, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, China
| | - Hong-Yan Geng
- Center for Neuroscience, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, China
| | - Ai-Min Bao
- Center for Neuroscience, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, China
| | - Shu-Min Duan
- Center for Neuroscience, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian-Ming Yang
- Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Tian-Ming Gao
- Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Hong Lian
- Center for Neuroscience, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao-Ming Li
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Center for Neuroscience, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
26
|
Müller T, Braud S, Jüttner R, Voigt BC, Paulick K, Sheean ME, Klisch C, Gueneykaya D, Rathjen FG, Geiger JR, Poulet JF, Birchmeier C. Neuregulin 3 promotes excitatory synapse formation on hippocampal interneurons. EMBO J 2018; 37:embj.201798858. [PMID: 30049711 PMCID: PMC6120667 DOI: 10.15252/embj.201798858] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 06/19/2018] [Accepted: 06/21/2018] [Indexed: 01/09/2023] Open
Abstract
Hippocampal GABAergic interneurons are crucial for cortical network function and have been implicated in psychiatric disorders. We show here that Neuregulin 3 (Nrg3), a relatively little investigated low-affinity ligand, is a functionally dominant interaction partner of ErbB4 in parvalbumin-positive (PV) interneurons. Nrg3 and ErbB4 are located pre- and postsynaptically, respectively, in excitatory synapses on PV interneurons in vivo Additionally, we show that ablation of Nrg3 results in a similar phenotype as the one described for ErbB4 ablation, including reduced excitatory synapse numbers on PV interneurons, altered short-term plasticity, and disinhibition of the hippocampal network. In culture, presynaptic Nrg3 increases excitatory synapse numbers on ErbB4+ interneurons and affects short-term plasticity. Nrg3 mutant neurons are poor donors of presynaptic terminals in the presence of competing neurons that produce recombinant Nrg3, and this bias requires postsynaptic ErbB4 but not ErbB4 kinase activity. Furthermore, when presented by non-neuronal cells, Nrg3 induces postsynaptic membrane specialization. Our data indicate that Nrg3 provides adhesive cues that facilitate excitatory neurons to synapse onto ErbB4+ interneurons.
Collapse
Affiliation(s)
- Thomas Müller
- Developmental Biology/Signal Transduction Group, Max-Delbrueck-Centrum in the Helmholtz Association, Berlin, Germany
| | - Stephanie Braud
- Institute of Neurophysiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - René Jüttner
- Developmental Neurobiology Group, Max-Delbrueck-Centrum in the Helmholtz Association, Berlin, Germany
| | - Birgit C Voigt
- Neural Circuits and Behaviour Group, Max-Delbrueck-Centrum in the Helmholtz Association, Berlin, Germany
| | - Katharina Paulick
- Developmental Biology/Signal Transduction Group, Max-Delbrueck-Centrum in the Helmholtz Association, Berlin, Germany
| | - Maria E Sheean
- Developmental Biology/Signal Transduction Group, Max-Delbrueck-Centrum in the Helmholtz Association, Berlin, Germany
| | - Constantin Klisch
- Institute of Neurophysiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Dilansu Gueneykaya
- Cellular Neuroscience Group, Max-Delbrueck-Centrum in the Helmholtz Association, Berlin, Germany
| | - Fritz G Rathjen
- Developmental Neurobiology Group, Max-Delbrueck-Centrum in the Helmholtz Association, Berlin, Germany
| | - Jörg Rp Geiger
- Institute of Neurophysiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - James Fa Poulet
- Neural Circuits and Behaviour Group, Max-Delbrueck-Centrum in the Helmholtz Association, Berlin, Germany.,Neuroscience Research Center and Cluster of Excellence NeuroCure, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Carmen Birchmeier
- Developmental Biology/Signal Transduction Group, Max-Delbrueck-Centrum in the Helmholtz Association, Berlin, Germany
| |
Collapse
|
27
|
Lloyd D, Talmage D, Shannon Weickert C, Karl T. Reduced type III neuregulin 1 expression does not modulate the behavioural sensitivity of mice to acute Δ 9 -tetrahydrocannabinol (D 9 -THC). Pharmacol Biochem Behav 2018; 170:64-70. [DOI: 10.1016/j.pbb.2018.05.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 04/12/2018] [Accepted: 05/07/2018] [Indexed: 12/15/2022]
|
28
|
Huang XF, Song X. Effects of antipsychotic drugs on neurites relevant to schizophrenia treatment. Med Res Rev 2018; 39:386-403. [PMID: 29785841 DOI: 10.1002/med.21512] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Revised: 04/06/2018] [Accepted: 04/30/2018] [Indexed: 12/31/2022]
Abstract
Although antipsychotic drugs are mainly used for treating schizophrenia, they are widely used for treating various psychiatric diseases in adults, the elderly, adolescents and even children. Today, about 1.2% of the worldwide population suffers from psychosis and related disorders, which translates to about 7.5 million subjects potentially targeted by antipsychotic drugs. Neurites project from the cell body of neurons and connect neurons to each other to form neural networks. Deficits in neurite outgrowth and integrity are implicated in psychiatric diseases including schizophrenia. Neurite deficits contribute to altered brain development, neural networking and connectivity as well as symptoms including psychosis and altered cognitive function. This review revealed that (1) antipsychotic drugs could have profound effects on neurites, synaptic spines and synapse, by which they may influence and regulate neural networking and plasticity; (2) antipsychotic drugs target not only neurotransmitter receptors but also intracellular signaling molecules regulating the signaling pathways responsible for neurite outgrowth and maintenance; (3) high doses and chronic administration of antipsychotic drugs may cause some loss of neurites, synaptic spines, or synapsis in the cortical structures. In addition, confounding effects causing neurite deficits may include elevated inflammatory cytokines and antipsychotic drug-induced metabolic side effects in patients on chronic antipsychotic therapy. Unraveling how antipsychotic drugs affect neurites and neural connectivity is essential for improving therapeutic outcomes and preventing aversive effects for patients on antipsychotic drug treatment.
Collapse
Affiliation(s)
- Xu-Feng Huang
- Henan Medical Key Laboratory of Translational Research on Psychiatric Diseases, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, China.,The Illawarra Health and Medical Research Institute and School of Medicine, University of Wollongong, Wollongong, Australia
| | - Xueqin Song
- Henan Medical Key Laboratory of Translational Research on Psychiatric Diseases, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
29
|
Kotzadimitriou D, Nissen W, Paizs M, Newton K, Harrison PJ, Paulsen O, Lamsa K. Neuregulin 1 Type I Overexpression Is Associated with Reduced NMDA Receptor-Mediated Synaptic Signaling in Hippocampal Interneurons Expressing PV or CCK. eNeuro 2018; 5:ENEURO.0418-17.2018. [PMID: 29740596 PMCID: PMC5938717 DOI: 10.1523/eneuro.0418-17.2018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 02/25/2018] [Accepted: 02/28/2018] [Indexed: 11/21/2022] Open
Abstract
Hypofunction of N-methyl-d-aspartate receptors (NMDARs) in inhibitory GABAergic interneurons is implicated in the pathophysiology of schizophrenia (SZ), a heritable disorder with many susceptibility genes. However, it is still unclear how SZ risk genes interfere with NMDAR-mediated synaptic transmission in diverse inhibitory interneuron populations. One putative risk gene is neuregulin 1 (NRG1), which signals via the receptor tyrosine kinase ErbB4, itself a schizophrenia risk gene. The type I isoform of NRG1 shows increased expression in the brain of SZ patients, and ErbB4 is enriched in GABAergic interneurons expressing parvalbumin (PV) or cholecystokinin (CCK). Here, we investigated ErbB4 expression and synaptic transmission in interneuronal populations of the hippocampus of transgenic mice overexpressing NRG1 type I (NRG1tg-type-I mice). Immunohistochemical analyses confirmed that ErbB4 was coexpressed with either PV or CCK in hippocampal interneurons, but we observed a reduced number of ErbB4-immunopositive interneurons in the NRG1tg-type-I mice. NMDAR-mediated currents in interneurons expressing PV (including PV+ basket cells) or CCK were reduced in NRG1tg-type-I mice compared to their littermate controls. We found no difference in AMPA receptor-mediated currents. Optogenetic activation (5 pulses at 20 Hz) of local glutamatergic fibers revealed a decreased NMDAR-mediated contribution to disynaptic GABAergic inhibition of pyramidal cells in the NRG1tg-type-I mice. GABAergic synaptic transmission from either PV+ or CCK+ interneurons, and glutamatergic transmission onto pyramidal cells, did not significantly differ between genotypes. The results indicate that synaptic NMDAR-mediated signaling in hippocampal interneurons is sensitive to chronically elevated NGR1 type I levels. This may contribute to the pathophysiological consequences of increased NRG1 expression in SZ.
Collapse
Affiliation(s)
| | - Wiebke Nissen
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK
| | - Melinda Paizs
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged, 6720, Hungary
| | - Kathryn Newton
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK
| | - Paul J. Harrison
- Department of Psychiatry, University of Oxford, and Oxford Health NHS Foundation Trust, Oxford, UK
| | - Ole Paulsen
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Karri Lamsa
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged, 6720, Hungary
| |
Collapse
|
30
|
Marballi KK, Gallitano AL. Immediate Early Genes Anchor a Biological Pathway of Proteins Required for Memory Formation, Long-Term Depression and Risk for Schizophrenia. Front Behav Neurosci 2018; 12:23. [PMID: 29520222 PMCID: PMC5827560 DOI: 10.3389/fnbeh.2018.00023] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 01/29/2018] [Indexed: 01/02/2023] Open
Abstract
While the causes of myriad medical and infectious illnesses have been identified, the etiologies of neuropsychiatric illnesses remain elusive. This is due to two major obstacles. First, the risk for neuropsychiatric disorders, such as schizophrenia, is determined by both genetic and environmental factors. Second, numerous genes influence susceptibility for these illnesses. Genome-wide association studies have identified at least 108 genomic loci for schizophrenia, and more are expected to be published shortly. In addition, numerous biological processes contribute to the neuropathology underlying schizophrenia. These include immune dysfunction, synaptic and myelination deficits, vascular abnormalities, growth factor disruption, and N-methyl-D-aspartate receptor (NMDAR) hypofunction. However, the field of psychiatric genetics lacks a unifying model to explain how environment may interact with numerous genes to influence these various biological processes and cause schizophrenia. Here we describe a biological cascade of proteins that are activated in response to environmental stimuli such as stress, a schizophrenia risk factor. The central proteins in this pathway are critical mediators of memory formation and a particular form of hippocampal synaptic plasticity, long-term depression (LTD). Each of these proteins is also implicated in schizophrenia risk. In fact, the pathway includes four genes that map to the 108 loci associated with schizophrenia: GRIN2A, nuclear factor of activated T-cells (NFATc3), early growth response 1 (EGR1) and NGFI-A Binding Protein 2 (NAB2); each of which contains the "Index single nucleotide polymorphism (SNP)" (most SNP) at its respective locus. Environmental stimuli activate this biological pathway in neurons, resulting in induction of EGR immediate early genes: EGR1, EGR3 and NAB2. We hypothesize that dysfunction in any of the genes in this pathway disrupts the normal activation of Egrs in response to stress. This may result in insufficient electrophysiologic, immunologic, and neuroprotective, processes that these genes normally mediate. Continued adverse environmental experiences, over time, may thereby result in neuropathology that gives rise to the symptoms of schizophrenia. By combining multiple genes associated with schizophrenia susceptibility, in a functional cascade triggered by neuronal activity, the proposed biological pathway provides an explanation for both the polygenic and environmental influences that determine the complex etiology of this mental illness.
Collapse
Affiliation(s)
- Ketan K. Marballi
- Department of Basic Medical Sciences and Psychiatry, University of Arizona College of Medicine—Phoenix, Phoenix, AZ, United States
| | - Amelia L. Gallitano
- Department of Basic Medical Sciences and Psychiatry, University of Arizona College of Medicine—Phoenix, Phoenix, AZ, United States
| |
Collapse
|
31
|
Xu J, Hartley BJ, Kurup P, Phillips A, Topol A, Xu M, Ononenyi C, Foscue E, Ho SM, Baguley TD, Carty N, Barros CS, Müller U, Gupta S, Gochman P, Rapoport J, Ellman JA, Pittenger C, Aronow B, Nairn AC, Nestor MW, Lombroso PJ, Brennand KJ. Inhibition of STEP 61 ameliorates deficits in mouse and hiPSC-based schizophrenia models. Mol Psychiatry 2018; 23:271-281. [PMID: 27752082 PMCID: PMC5395367 DOI: 10.1038/mp.2016.163] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 07/13/2016] [Accepted: 08/11/2016] [Indexed: 02/03/2023]
Abstract
The brain-specific tyrosine phosphatase, STEP (STriatal-Enriched protein tyrosine Phosphatase) is an important regulator of synaptic function. STEP normally opposes synaptic strengthening by increasing N-methyl D-aspartate glutamate receptor (NMDAR) internalization through dephosphorylation of GluN2B and inactivation of the kinases extracellular signal-regulated kinase 1/2 and Fyn. Here we show that STEP61 is elevated in the cortex in the Nrg1+/- knockout mouse model of schizophrenia (SZ). Genetic reduction or pharmacological inhibition of STEP prevents the loss of NMDARs from synaptic membranes and reverses behavioral deficits in Nrg1+/- mice. STEP61 protein is also increased in cortical lysates from the central nervous system-specific ErbB2/4 mouse model of SZ, as well as in human induced pluripotent stem cell (hiPSC)-derived forebrain neurons and Ngn2-induced excitatory neurons, from two independent SZ patient cohorts. In these selected SZ models, increased STEP61 protein levels likely reflect reduced ubiquitination and degradation. These convergent findings from mouse and hiPSC SZ models provide evidence for STEP61 dysfunction in SZ.
Collapse
Affiliation(s)
- J Xu
- Child Study Center, Yale University, New Haven, CT, USA
| | - B J Hartley
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - P Kurup
- Child Study Center, Yale University, New Haven, CT, USA
| | - A Phillips
- Hussman Institute for Autism, Baltimore, MD, USA
| | - A Topol
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - M Xu
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - C Ononenyi
- Child Study Center, Yale University, New Haven, CT, USA
| | - E Foscue
- Child Study Center, Yale University, New Haven, CT, USA
| | - S-M Ho
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Department of Developmental and Stem Cell Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - T D Baguley
- Department of Chemistry, Yale University, New Haven, CT, USA
| | - N Carty
- Child Study Center, Yale University, New Haven, CT, USA
| | - C S Barros
- Dorris Neuroscience Center, Department of Cell Biology, The Scripps Research Institute, La Jolla, CA, USA,Plymouth University School of Medicine, Plymouth UK
| | - U Müller
- Dorris Neuroscience Center, Department of Cell Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - S Gupta
- UC Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - P Gochman
- Childhood Psychiatry Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - J Rapoport
- Childhood Psychiatry Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - J A Ellman
- Department of Chemistry, Yale University, New Haven, CT, USA
| | - C Pittenger
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - B Aronow
- UC Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - A C Nairn
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - M W Nestor
- Hussman Institute for Autism, Baltimore, MD, USA
| | - P J Lombroso
- Child Study Center, Yale University, New Haven, CT, USA,Department of Psychiatry, Yale University, New Haven, CT, USA,Department of Neurobiology, Yale University, New Haven, CT, USA,Department of Psychiatry, Yale University, New Haven, CT, USA E-mail:
| | - K J Brennand
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY, 10029, USA. E-mail:
| |
Collapse
|
32
|
Jha MK, Kim JH, Song GJ, Lee WH, Lee IK, Lee HW, An SSA, Kim S, Suk K. Functional dissection of astrocyte-secreted proteins: Implications in brain health and diseases. Prog Neurobiol 2017; 162:37-69. [PMID: 29247683 DOI: 10.1016/j.pneurobio.2017.12.003] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 10/23/2017] [Accepted: 12/08/2017] [Indexed: 02/07/2023]
Abstract
Astrocytes, which are homeostatic cells of the central nervous system (CNS), display remarkable heterogeneity in their morphology and function. Besides their physical and metabolic support to neurons, astrocytes modulate the blood-brain barrier, regulate CNS synaptogenesis, guide axon pathfinding, maintain brain homeostasis, affect neuronal development and plasticity, and contribute to diverse neuropathologies via secreted proteins. The identification of astrocytic proteome and secretome profiles has provided new insights into the maintenance of neuronal health and survival, the pathogenesis of brain injury, and neurodegeneration. Recent advances in proteomics research have provided an excellent catalog of astrocyte-secreted proteins. This review categorizes astrocyte-secreted proteins and discusses evidence that astrocytes play a crucial role in neuronal activity and brain function. An in-depth understanding of astrocyte-secreted proteins and their pathways is pivotal for the development of novel strategies for restoring brain homeostasis, limiting brain injury/inflammation, counteracting neurodegeneration, and obtaining functional recovery.
Collapse
Affiliation(s)
- Mithilesh Kumar Jha
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jong-Heon Kim
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Gyun Jee Song
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Won-Ha Lee
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - In-Kyu Lee
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Ho-Won Lee
- Department of Neurology, Brain Science and Engineering Institute, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Seong Soo A An
- Department of BioNano Technology, Gachon University, Gyeonggi-do, Republic of Korea
| | - SangYun Kim
- Department of Neurology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Gyeonggi-do, Republic of Korea
| | - Kyoungho Suk
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea.
| |
Collapse
|
33
|
O’Tuathaigh CMP, Mathur N, O’Callaghan MJ, MacIntyre L, Harvey R, Lai D, Waddington JL, Pickard BS, Watson DG, Moran PM. Specialized Information Processing Deficits and Distinct Metabolomic Profiles Following TM-Domain Disruption of Nrg1. Schizophr Bull 2017; 43:1100-1113. [PMID: 28338897 PMCID: PMC5581893 DOI: 10.1093/schbul/sbw189] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Although there is considerable genetic and pathologic evidence for an association between neuregulin 1 (NRG1) dysregulation and schizophrenia, the underlying molecular and cellular mechanisms remain unclear. Mutant mice containing disruption of the transmembrane (TM) domain of the NRG1 gene constitute a heuristic model for dysregulation of NRG1-ErbB4 signaling in schizophrenia. The present study focused on hitherto uncharacterized information processing phenotypes in this mutant line. Using a mass spectrometry-based metabolomics approach, we also quantified levels of unique metabolites in brain. Across 2 different sites and protocols, Nrg1 mutants demonstrated deficits in prepulse inhibition, a measure of sensorimotor gating, that is, disrupted in schizophrenia; these deficits were partially reversed by acute treatment with second, but not first-, generation antipsychotic drugs. However, Nrg1 mutants did not show a specific deficit in latent inhibition, a measure of selective attention that is also disrupted in schizophrenia. In contrast, in a "what-where-when" object recognition memory task, Nrg1 mutants displayed sex-specific (males only) disruption of "what-when" performance, indicative of impaired temporal aspects of episodic memory. Differential metabolomic profiling revealed that these behavioral phenotypes were accompanied, most prominently, by alterations in lipid metabolism pathways. This study is the first to associate these novel physiological mechanisms, previously independently identified as being abnormal in schizophrenia, with disruption of NRG1 function. These data suggest novel mechanisms by which compromised neuregulin function from birth might lead to schizophrenia-relevant behavioral changes in adulthood.
Collapse
Affiliation(s)
| | - Naina Mathur
- School of Psychology, University of Nottingham, Nottingham, UK
| | | | - Lynsey MacIntyre
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Richard Harvey
- Victor Chang Cardiac Research Institute, Sydney, Australia
| | - Donna Lai
- Victor Chang Cardiac Research Institute, Sydney, Australia
| | - John L Waddington
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland
- Jiangsu Key Laboratory of Translational Research & Therapy for Neuro-Psychiatric-Disorders and Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Benjamin S Pickard
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - David G Watson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Paula M Moran
- School of Psychology, University of Nottingham, Nottingham, UK
| |
Collapse
|
34
|
Shang K, Talmage DA, Karl T. Parent-of-origin effects on schizophrenia-relevant behaviours of type III neuregulin 1 mutant mice. Behav Brain Res 2017; 332:250-258. [DOI: 10.1016/j.bbr.2017.05.057] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 04/29/2017] [Accepted: 05/24/2017] [Indexed: 12/18/2022]
|
35
|
Chakraborty A, Murphy S, Coleman N. The Role of NMDA Receptors in Neural Stem Cell Proliferation and Differentiation. Stem Cells Dev 2017; 26:798-807. [DOI: 10.1089/scd.2016.0325] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Affiliation(s)
- Adri Chakraborty
- Department of Biological Sciences, University of the Sciences in Philadelphia, Philadelphia, Pennsylvania
| | - Suzanne Murphy
- Department of Biological Sciences, University of the Sciences in Philadelphia, Philadelphia, Pennsylvania
| | - Natalia Coleman
- Biology, New Jersey City University, Jersey City, New Jersey
| |
Collapse
|
36
|
Variation at NRG1 genotype related to modulation of small-world properties of the functional cortical network. Eur Arch Psychiatry Clin Neurosci 2017; 267:25-32. [PMID: 26650688 DOI: 10.1007/s00406-015-0659-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 11/17/2015] [Indexed: 01/11/2023]
Abstract
Functional brain networks possess significant small-world (SW) properties. Genetic variation relevant to both inhibitory and excitatory transmission may contribute to modulate these properties. In healthy controls, genotypic variation in Neuregulin 1 (NRG1) related to the risk of psychosis (risk alleles) would contribute to functional SW modulation of the cortical network. Electroencephalographic activity during an odd-ball task was recorded in 144 healthy controls. Then, small-worldness (SWn) was calculated in five frequency bands (i.e., theta, alpha, beta1, beta2 and gamma) for baseline (from -300 to the stimulus onset) and response (150-450 ms post-target stimulus) windows. The SWn modulation was defined as the difference in SWn between both windows. Association between SWn modulation and carrying the risk allele for three single nucleotide polymorphisms (SNP) of NRG1 (i.e., rs6468119, rs6994992 and rs7005606) was assessed. A significant association between three SNPs of NRG1 and the SWn modulation was found, specifically: NRG1 rs6468119 in alpha and beta1 bands; NRG1 rs6994992 in theta band; and NRG1 rs7005606 in theta and beta1 bands. Genetic variation at NRG1 may influence functional brain connectivity through the modulation of SWn properties of the cortical network.
Collapse
|
37
|
Koványi B, Csölle C, Calovi S, Hanuska A, Kató E, Köles L, Bhattacharya A, Haller J, Sperlágh B. The role of P2X7 receptors in a rodent PCP-induced schizophrenia model. Sci Rep 2016; 6:36680. [PMID: 27824163 PMCID: PMC5099752 DOI: 10.1038/srep36680] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 10/18/2016] [Indexed: 02/07/2023] Open
Abstract
P2X7 receptors (P2X7Rs) are ligand-gated ion channels sensitive to extracellular ATP. Here we examined for the first time the role of P2X7R in an animal model of schizophrenia. Using the PCP induced schizophrenia model we show that both genetic deletion and pharmacological inhibition of P2X7Rs alleviate schizophrenia-like behavioral alterations. In P2rx7+/+ mice, PCP induced hyperlocomotion, stereotype behavior, ataxia and social withdrawal. In P2X7 receptor deficient mice (P2rx7−/−), the social interactions were increased, whereas the PCP induced hyperlocomotion and stereotype behavior were alleviated. The selective P2X7 receptor antagonist JNJ-47965567 partly replicated the effect of gene deficiency on PCP-induced behavioral changes and counteracted PCP-induced social withdrawal. We also show that PCP treatment upregulates and increases the functional responsiveness of P2X7Rs in the prefrontal cortex of young adult animals. The amplitude of NMDA evoked currents recorded from layer V pyramidal neurons of cortical slices were slightly decreased by both genetic deletion of P2rx7 and by JNJ-47965567. PCP induced alterations in mRNA expression encoding schizophrenia-related genes, such as NR2A, NR2B, neuregulin 1, NR1 and GABA α1 subunit were absent in the PFC of young adult P2rx7−/− animals. Our findings point to P2X7R as a potential therapeutic target in schizophrenia.
Collapse
Affiliation(s)
- Bence Koványi
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences (IEM HAS), H-1450 Budapest, Hungary.,János Szentágothai School of Neurosciences, Semmelweis University School of PhD Studies, Budapest, Hungary
| | - Cecilia Csölle
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences (IEM HAS), H-1450 Budapest, Hungary
| | - Stefano Calovi
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences (IEM HAS), H-1450 Budapest, Hungary
| | - Adrienn Hanuska
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, H-1089 Budapest, Hungary
| | - Erzsébet Kató
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, H-1089 Budapest, Hungary
| | - László Köles
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, H-1089 Budapest, Hungary
| | | | - József Haller
- Department of Behavioral Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences (IEM HAS), H-1450, Budapest, Hungary
| | - Beáta Sperlágh
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences (IEM HAS), H-1450 Budapest, Hungary
| |
Collapse
|
38
|
Yamazaki Y, Sumikawa K. Nicotine-induced neuroplasticity counteracts the effect of schizophrenia-linked neuregulin 1 signaling on NMDAR function in the rat hippocampus. Neuropharmacology 2016; 113:386-395. [PMID: 27784625 DOI: 10.1016/j.neuropharm.2016.10.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 10/10/2016] [Accepted: 10/21/2016] [Indexed: 12/13/2022]
Abstract
A high rate of heavy tobacco smoking among people with schizophrenia has been suggested to reflect self-medication and amelioration of cognitive dysfunction, a core feature of schizophrenia. NMDAR hypofunction is hypothesized to be a mechanism of cognitive dysfunction, and excessive schizophrenia-linked neuregulin 1 (NRG1) signaling through its receptor ErbB4 can suppress NMDAR function by preventing Src-mediated enhancement of NMDAR responses. Here we investigated whether chronic nicotine exposure in rats by subcutaneous injection of nicotine (0.5-1 mg/kg, twice daily for 10-15 days) counteracts the suppressive effect of NRG1β on NMDAR-mediated responses recorded from CA1 pyramidal cells in acute hippocampal slices. We found that NRG1β, which prevents the enhancement of NMDAR responses by the Src-family-kinase-activating peptide pYEEI in naive rats, failed to block the effect of pYEEI in nicotine-exposed rats. In naive rats, NRG1β acts only on GluN2B-NMDARs by blocking their Src-mediated upregulation. Chronic nicotine exposure causes enhanced GluN2B-NMDAR responses via Src upregulation and recruits Fyn for the enhancement of GluN2A-NMDAR responses. NRG1β has no effect on both enhanced basal GluN2B-NMDAR responses and Fyn-mediated enhancement of GluN2A-NMDAR responses. Src-mediated enhancement of GluN2B-NMDAR responses and Fyn-mediated enhancement of GluN2A-NMDAR responses initiate long-term potentiation (LTP) of AMPAR synaptic responses in naive and nicotine-exposed CA1 pyramidal cells, respectively. These results suggest that NRG1β suppresses LTP by blocking Src-mediated enhancement of GluN2B-NMDAR responses, but has no effect on LTP in nicotine-exposed rats. These effects of chronic nicotine exposure may counteract the negative effect of increased NRG1-ErbB4 signaling on the cellular mechanisms of learning and memory in individuals with schizophrenia, and therefore may motivate heavy smoking.
Collapse
Affiliation(s)
- Yoshihiko Yamazaki
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697-4550, USA; Department of Neurophysiology, Yamagata University School of Medicine, Yamagata 990-9585, Japan
| | - Katumi Sumikawa
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697-4550, USA.
| |
Collapse
|
39
|
Lecca D, Marangon D, Coppolino GT, Méndez AM, Finardi A, Costa GD, Martinelli V, Furlan R, Abbracchio MP. MiR-125a-3p timely inhibits oligodendroglial maturation and is pathologically up-regulated in human multiple sclerosis. Sci Rep 2016; 6:34503. [PMID: 27698367 PMCID: PMC5048305 DOI: 10.1038/srep34503] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 09/09/2016] [Indexed: 12/31/2022] Open
Abstract
In the mature central nervous system (CNS), oligodendrocytes provide support and insulation to axons thanks to the production of a myelin sheath. During their maturation to myelinating cells, oligodendroglial precursors (OPCs) follow a very precise differentiation program, which is finely orchestrated by transcription factors, epigenetic factors and microRNAs (miRNAs), a class of small non-coding RNAs involved in post-transcriptional regulation. Any alterations in this program can potentially contribute to dysregulated myelination, impaired remyelination and neurodegenerative conditions, as it happens in multiple sclerosis (MS). Here, we identify miR-125a-3p, a developmentally regulated miRNA, as a new actor of oligodendroglial maturation, that, in the mammalian CNS regulates the expression of myelin genes by simultaneously acting on several of its already validated targets. In cultured OPCs, over-expression of miR-125a-3p by mimic treatment impairs while its inhibition with an antago-miR stimulates oligodendroglial maturation. Moreover, we show that miR-125a-3p levels are abnormally high in the cerebrospinal fluid of MS patients bearing active demyelinating lesions, suggesting that its pathological upregulation may contribute to MS development, at least in part by blockade of OPC differentiation leading to impaired repair of demyelinated lesions.
Collapse
Affiliation(s)
- Davide Lecca
- Laboratory of Molecular and Cellular Pharmacology of the Purinergic Transmission, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, 20133, Italy
| | - Davide Marangon
- Laboratory of Molecular and Cellular Pharmacology of the Purinergic Transmission, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, 20133, Italy
| | - Giusy T Coppolino
- Laboratory of Molecular and Cellular Pharmacology of the Purinergic Transmission, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, 20133, Italy
| | - Aida Menéndez Méndez
- Departamento de Bioquímica y Biología Molecular IV, Universidad Complutense de Madrid, 28040, Spain
| | - Annamaria Finardi
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, San Raffaele Scientific Institute, Milan, 20132, Italy
| | - Gloria Dalla Costa
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, San Raffaele Scientific Institute, Milan, 20132, Italy
| | - Vittorio Martinelli
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, San Raffaele Scientific Institute, Milan, 20132, Italy
| | - Roberto Furlan
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, San Raffaele Scientific Institute, Milan, 20132, Italy
| | - Maria P Abbracchio
- Laboratory of Molecular and Cellular Pharmacology of the Purinergic Transmission, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, 20133, Italy
| |
Collapse
|
40
|
Mostaid MS, Lloyd D, Liberg B, Sundram S, Pereira A, Pantelis C, Karl T, Weickert CS, Everall IP, Bousman CA. Neuregulin-1 and schizophrenia in the genome-wide association study era. Neurosci Biobehav Rev 2016; 68:387-409. [DOI: 10.1016/j.neubiorev.2016.06.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 05/30/2016] [Accepted: 06/03/2016] [Indexed: 12/22/2022]
|
41
|
Proteolytic processing of Neuregulin-1. Brain Res Bull 2016; 126:178-182. [PMID: 27393467 DOI: 10.1016/j.brainresbull.2016.07.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Revised: 06/28/2016] [Accepted: 07/03/2016] [Indexed: 02/07/2023]
Abstract
Neuregulin-1 (NRG1), known also as heregulin, acetylcholine receptor inducing activity (ARIA), glial growth factor (GGF), or sensory and motor neuron derived factor (SMDF), is a key factor for many developmental processes and in adult brain. All known splice variants contain an epidermal growth factor (EGF)-like domain, which is mediating signaling via receptors of the ErbB family. In particular, NRG1 acts as an essential signaling molecule expressed on the axonal surface, where it signals to Schwann cells throughout development and regulates the thickness of the myelin sheath. NRG1 is required also by other cell types in the nervous system, for instance as an axonal signal released by proprioceptive afferents to induce development of the muscle spindle, and it controls aspects of cortical interneuron development as well as the formation of thalamo-cortical projections. The precursor protein of NRG1 can be activated and released from the membrane through limited proteolysis by the β-Secretase (β-site amyloid precursor protein cleaving enzyme 1, BACE1) which was first identified through its function as the rate limiting enzyme of amyloid-β-peptide (Aβ) production. Aβ is the major component of amyloid plaques in Alzheimer's disease (AD). Due to the hairpin nature of NRG1 type III two membrane-bound stubs with a type 1 and a type 2 orientation are generated by an initial proteolytic cleavage and successive release of the EGF-like domain either by dual cleavage by BACE1 or by ADAM17 (a disintegrin and metalloprotease) which is also called TACE (Tumor Necrosis Factor-α-converting enzyme). The cleavages activate NRG1 to allow juxtacrine or paracrine signaling. The type 1 oriented stub is further cleaved by γ-secretase in the transmembrane domain with a putative role in intracellular domain (ICD) signaling, while the type II oriented stub is cleaved by signal peptidase like proteases (SPPLs). Neuregulin-1 was identified as a major physiological substrate of BACE1 during early postnatal development when similarities in BACE1 KO mice and NRG1 heterozygous mice were discovered. Both display severe hypomyelination of peripheral nerves. Later it was shown with genetic and pharmacological evidence that the developmental effect of type I NRG1 on the formation and the maintenance of muscle spindles is BACE1 dependent. Thus, NRG1 functions in PNS and CNS are likely to set limits to an Alzheimer disease therapy with relatively strong BACE1 inhibition.
Collapse
|
42
|
Sigurdsson T. Neural circuit dysfunction in schizophrenia: Insights from animal models. Neuroscience 2016; 321:42-65. [DOI: 10.1016/j.neuroscience.2015.06.059] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 06/15/2015] [Accepted: 06/26/2015] [Indexed: 12/17/2022]
|
43
|
Holm-Hansen S, Low JK, Zieba J, Gjedde A, Bergersen LH, Karl T. Behavioural effects of high fat diet in a mutant mouse model for the schizophrenia risk gene neuregulin 1. GENES BRAIN AND BEHAVIOR 2016; 15:295-304. [PMID: 26707035 DOI: 10.1111/gbb.12267] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 07/29/2015] [Accepted: 07/29/2015] [Indexed: 11/29/2022]
Abstract
Schizophrenia patients are often obese or overweight and poor dietary choices appear to be a factor in this phenomenon. Poor diet has been found to have complex consequences for the mental state of patients. Thus, this study investigated whether an unhealthy diet [i.e. high fat diet (HFD)] impacts on the behaviour of a genetic mouse model for the schizophrenia risk gene neuregulin 1 (i.e. transmembrane domain Nrg1 mutant mice: Nrg1 HET). Female Nrg1 HET and wild-type-like littermates (WT) were fed with either HFD or a control chow diet. The mice were tested for baseline (e.g. anxiety) and schizophrenia-relevant behaviours after 7 weeks of diet exposure. HFD increased body weight and impaired glucose tolerance in all mice. Only Nrg1 females on HFD displayed a hyper-locomotive phenotype as locomotion-suppressive effects of HFD were only evident in WT mice. HFD also induced an anxiety-like response and increased freezing in the context and the cued version of the fear conditioning task. Importantly, CHOW-fed Nrg1 females displayed impaired social recognition memory, which was absent in HFD-fed mutants. Sensorimotor gating deficits of Nrg1 females were not affected by diet. In summary, HFD had complex effects on the behavioural phenotype of test mice and attenuated particular cognitive deficits of Nrg1 mutant females. This topic requires further investigations thereby also considering other dietary factors of relevance for schizophrenia as well as interactive effects of diet with medication and sex.
Collapse
Affiliation(s)
- S Holm-Hansen
- Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark.,Brain and Muscle Energy Group, Electron Microscopy Laboratory, Institute of Oral Biology, University of Oslo, Oslo, Norway
| | - J K Low
- Neuroscience Research Australia, Sydney, Australia.,Schizophrenia Research Institute, Randwick, Australia
| | - J Zieba
- Neuroscience Research Australia, Sydney, Australia.,Schizophrenia Research Institute, Randwick, Australia
| | - A Gjedde
- Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark.,Center for Healthy Aging, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - L H Bergersen
- Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark.,Brain and Muscle Energy Group, Electron Microscopy Laboratory, Institute of Oral Biology, University of Oslo, Oslo, Norway.,Center for Healthy Aging, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - T Karl
- Neuroscience Research Australia, Sydney, Australia.,Schizophrenia Research Institute, Randwick, Australia.,School of Medical Sciences, University of New South Wales, Sydney, Australia
| |
Collapse
|
44
|
Zhang Q, Yu Y, Huang XF. Olanzapine Prevents the PCP-induced Reduction in the Neurite Outgrowth of Prefrontal Cortical Neurons via NRG1. Sci Rep 2016; 6:19581. [PMID: 26781398 PMCID: PMC4726088 DOI: 10.1038/srep19581] [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: 10/01/2015] [Accepted: 12/15/2015] [Indexed: 11/09/2022] Open
Abstract
Accumulating evidence suggests that reducing neurite outgrowth and synaptic plasticity plays a critical role in the pathology of cognitive deficits in schizophrenia. The N-methyl-D-aspartate receptor antagonist phencyclidine (PCP) can induce symptoms of schizophrenia as well as reduce dendritic spine density and neurite growth. The antipsychotic drug olanzapine may improve these deficits. This study aimed to investigate: (1) if olanzapine prevents PCP-induced suppression of neurite outgrowth and synaptic protein expression; (2) if olanzapine affects the Akt-GSK3 signaling pathway; and (3) the role of neuregulin 1 (NRG1) in this process. Immunofluorescence revealed that PCP treatment for 24 hours reduces both neurite length (28.5%) and the number of neurite branches (35.6%) in primary prefrontal cortical neuron cultures. PCP reduced protein and mRNA expressions of synaptophysin (24.9% and 23.2%, respectively) and PSD95 (31.5% and 21.4%, respectively), and the protein expression of p-Akt (26.7%) and p-GSK3β (35.2%). Olanzapine co-treatment prevented these PCP-induced effects in normal neurons but not in neurons from NRG1-knockout mice. These results indicate that NRG1 mediates the preventive effects of olanzapine on the PCP-induced impairment of neurite outgrowth and synaptic protein expression. This study provides potential targets for interventions on improving the efficacy of olanzapine on preventing cognitive deficits in schizophrenia.
Collapse
Affiliation(s)
- Qingsheng Zhang
- Centre for Translational Neuroscience, School of Medicine, University of Wollongong, Wollongong, 2522, NSW, Australia.,Illawarra Health and Medical Research Institute, Wollongong, 2522, NSW, Australia
| | - Yinghua Yu
- Centre for Translational Neuroscience, School of Medicine, University of Wollongong, Wollongong, 2522, NSW, Australia.,Illawarra Health and Medical Research Institute, Wollongong, 2522, NSW, Australia.,Schizophrenia Research Institute, 384 Victoria Street, Darlinghurst, 2010, NSW, Australia
| | - Xu-Feng Huang
- Centre for Translational Neuroscience, School of Medicine, University of Wollongong, Wollongong, 2522, NSW, Australia.,Illawarra Health and Medical Research Institute, Wollongong, 2522, NSW, Australia.,Schizophrenia Research Institute, 384 Victoria Street, Darlinghurst, 2010, NSW, Australia
| |
Collapse
|
45
|
Hu X, Fan Q, Hou H, Yan R. Neurological dysfunctions associated with altered BACE1-dependent Neuregulin-1 signaling. J Neurochem 2016; 136:234-49. [PMID: 26465092 PMCID: PMC4833723 DOI: 10.1111/jnc.13395] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 09/23/2015] [Accepted: 09/25/2015] [Indexed: 01/09/2023]
Abstract
Inhibition of BACE1 is being pursued as a therapeutic target to treat patients suffering from Alzheimer's disease because BACE1 is the sole β-secretase that generates β-amyloid peptide. Knowledge regarding other cellular functions of BACE1 is therefore critical for the safe use of BACE1 inhibitors in human patients. Neuregulin-1 (Nrg1) is a BACE1 substrate and BACE1 cleavage of Nrg1 is critical for signaling functions in myelination, remyelination, synaptic plasticity, normal psychiatric behaviors, and maintenance of muscle spindles. This review summarizes the most recent discoveries associated with BACE1-dependent Nrg1 signaling in these areas. This body of knowledge will help to provide guidance for preventing unwanted Nrg1-based side effects following BACE1 inhibition in humans. To initiate its signaling cascade, membrane anchored Neuregulin (Nrg), mainly type I and III β1 Nrg1 isoforms and Nrg3, requires ectodomain shedding. BACE1 is one of such indispensable sheddases to release the functional Nrg signaling fragment. The dependence of Nrg on the cleavage by BACE1 is best manifested by disrupting the critical role of Nrg in the control of axonal myelination, schizophrenic behaviors as well as the formation and maintenance of muscle spindles.
Collapse
Affiliation(s)
- Xiangyou Hu
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Qingyuan Fan
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Hailong Hou
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Riqiang Yan
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| |
Collapse
|
46
|
Jajodia A, Kaur H, Kumari K, Kanojia N, Gupta M, Baghel R, Sood M, Jain S, Chadda RK, Kukreti R. Evaluation of genetic association of neurodevelopment and neuroimmunological genes with antipsychotic treatment response in schizophrenia in Indian populations. Mol Genet Genomic Med 2015; 4:18-27. [PMID: 26788534 PMCID: PMC4707035 DOI: 10.1002/mgg3.169] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 07/10/2015] [Indexed: 12/14/2022] Open
Abstract
Neurodevelopmental and neuroimmunological genes critically regulate antipsychotic treatment outcome. We report genetic associations of antipsychotic response in 742 schizophrenia patients from Indian populations of Indo‐European and Dravidian ancestry, segregated by disease severity. Meta‐analysis comparing the two populations identified CCL2 [rs4795893: OR (95% CI) = 1.79 (1.27–2.52), P = 7.62 × 10−4; rs4586: OR (95% CI) = 1.74 (1.24–2.43), P = 1.13 × 10−3] and GRIA4 [rs2513265: OR (95% CI) = 0.53 (0.36–0.78), P = 1.44 × 10−3] in low severity group; and, ADCY2 [rs1544938: OR (95% CI) = 0.36 (0.19–0.65), P = 7.68 × 10−4] and NRG1 [rs13250975, OR (95% CI) = 0.42 (0.23–0.79), P = 6.81 × 10−3; rs17716295, OR (95% CI) = 1.78 (1.15–2.75), P = 8.71 × 10−3] in high severity group, with incomplete response toward antipsychotics. To our knowledge, this is the first study to identify genetic polymorphisms associated with the efficacy of antipsychotic treatment of schizophrenia patients from two major India populations.
Collapse
Affiliation(s)
- Ajay Jajodia
- Genomics and Molecular Medicine CSIR-Institute of Genomics and Integrative Biology Mall Road Delhi 110007 India
| | - Harpreet Kaur
- Genomics and Molecular Medicine CSIR-Institute of Genomics and Integrative Biology Mall Road Delhi 110007 India
| | - Kalpana Kumari
- Department of Psychiatry All India Institute of Medical Sciences Ansari Nagar New Delhi 110029 India
| | - Neha Kanojia
- Genomics and Molecular Medicine CSIR-Institute of Genomics and Integrative Biology Mall Road Delhi 110007 India
| | - Meenal Gupta
- Genomics and Molecular Medicine CSIR-Institute of Genomics and Integrative Biology Mall Road Delhi 110007 India
| | - Ruchi Baghel
- Genomics and Molecular Medicine CSIR-Institute of Genomics and Integrative Biology Mall Road Delhi 110007 India
| | - Mamta Sood
- Department of Psychiatry All India Institute of Medical Sciences Ansari Nagar New Delhi 110029 India
| | - Sanjeev Jain
- Molecular Genetic Laboratory Department of Psychiatry National Institute of Mental Health and Neuro Sciences Hosur Road Bengaluru 560029 India
| | - Rakesh K Chadda
- Department of Psychiatry All India Institute of Medical Sciences Ansari Nagar New Delhi 110029 India
| | - Ritushree Kukreti
- Genomics and Molecular Medicine CSIR-Institute of Genomics and Integrative Biology Mall Road Delhi 110007 India
| |
Collapse
|
47
|
Tang Y, Ye M, Du Y, Qiu X, Lv X, Yang W, Luo J. EGFR signaling upregulates surface expression of the GluN2B-containing NMDA receptor and contributes to long-term potentiation in the hippocampus. Neuroscience 2015. [PMID: 26204818 DOI: 10.1016/j.neuroscience.2015.07.021] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
N-methyl-d-aspartate receptors (NMDARs) have been known to be regulated by various receptor tyrosine kinases. Activation of epidermal growth factor receptor (EGFR) specifically increases NMDAR-mediated currents and enhances long-term potentiation (LTP) in the hippocampus. However, the mechanism through which EGFR regulates NMDARs remains to be elucidated. In this study we found that EGFR was highly expressed in the hippocampus and mainly localized in the non-synaptic region including the soma and neurites of cultured hippocampal neurons. EGFR activation led to an increase in ifenprodil-sensitive NMDAR currents. Consistent with this, we also observed that surface expression of GluN2B-containing NMDAR was upregulated. Our biochemical data from hippocampal slices and hippocampal cultured neurons demonstrated that EGF treatment in vitro significantly increased phosphorylation of the GluN2B subunit at Y1472 with a coincidental activation of Src family kinases (SFKs). EGFR blockade with a specific antagonist BIBX-1382 attenuated an increase of GluN2B in the postsynaptic density during high-frequency stimulation (HFS)-induced LTP. Moreover, BIBX blockade significantly impaired HFS-induced LTP. In conclusion, our findings suggest that EGFR signaling upregulates NMDARs through modification of the GluN2B subunit, and is required for HFS-induced LTP in the hippocampus.
Collapse
Affiliation(s)
- Y Tang
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - M Ye
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Y Du
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - X Qiu
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - X Lv
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - W Yang
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - J Luo
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
48
|
Xu J, Kurup P, Foscue E, Lombroso PJ. Striatal-enriched protein tyrosine phosphatase regulates the PTPα/Fyn signaling pathway. J Neurochem 2015; 134:629-41. [PMID: 25951993 DOI: 10.1111/jnc.13160] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 04/05/2015] [Accepted: 04/27/2015] [Indexed: 12/13/2022]
Abstract
The tyrosine kinase Fyn has two regulatory tyrosine residues that when phosphorylated either activate (Tyr(420)) or inhibit (Tyr(531)) Fyn activity. Within the central nervous system, two protein tyrosine phosphatases (PTPs) target these regulatory tyrosines in Fyn. PTPα dephosphorylates Tyr(531) and activates Fyn, while STEP (STriatal-Enriched protein tyrosine Phosphatase) dephosphorylates Tyr(420) and inactivates Fyn. Thus, PTPα and STEP have opposing functions in the regulation of Fyn; however, whether there is cross talk between these two PTPs remains unclear. Here, we used molecular techniques in primary neuronal cultures and in vivo to demonstrate that STEP negatively regulates PTPα by directly dephosphorylating PTPα at its regulatory Tyr(789). Dephosphorylation of Tyr(789) prevents the translocation of PTPα to synaptic membranes, blocking its ability to interact with and activate Fyn. Genetic or pharmacologic reduction in STEP61 activity increased the phosphorylation of PTPα at Tyr(789), as well as increased translocation of PTPα to synaptic membranes. Activation of PTPα and Fyn and trafficking of GluN2B to synaptic membranes are necessary for ethanol (EtOH) intake behaviors in rodents. We tested the functional significance of STEP61 in this signaling pathway by EtOH administration to primary cultures as well as in vivo, and demonstrated that the inactivation of STEP61 by EtOH leads to the activation of PTPα, its translocation to synaptic membranes, and the activation of Fyn. These findings indicate a novel mechanism by which STEP61 regulates PTPα and suggest that STEP and PTPα coordinate the regulation of Fyn. STEP61 , PTPα, Fyn, and NMDA receptor (NMDAR) have been implicated in ethanol intake behaviors in the dorsomedial striatum (DMS) in rodents. Here, we report that PTPα is a novel substrate for STEP61. Upon ethanol exposure, STEP61 is phosphorylated and inactivated by protein kinase A (PKA) signaling in the DMS. As a result of STEP61 inhibition, there is an increase in the phosphorylation of PTPα, which translocates to lipid rafts and activates Fyn and subsequent NMDAR signaling. The results demonstrate a synergistic regulation of Fyn-NMDAR signaling by STEP61 and PTPα, which may contribute to the regulation of ethanol-related behaviors. NMDA, N-methyl-D-aspartate; PTPα, receptor-type protein tyrosine phosphatase alpha; STEP, STriatal-Enriched protein tyrosine Phosphatase.
Collapse
Affiliation(s)
- Jian Xu
- Child Study Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Pradeep Kurup
- Child Study Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Ethan Foscue
- Child Study Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Paul J Lombroso
- Child Study Center, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
49
|
Long LE, Anderson P, Frank E, Shaw A, Liu S, Huang XF, Pinault D, Karl T, O’Brien TJ, Shannon Weickert C, Jones NC. Neuregulin 1 expression and electrophysiological abnormalities in the Neuregulin 1 transmembrane domain heterozygous mutant mouse. PLoS One 2015; 10:e0124114. [PMID: 25992564 PMCID: PMC4437646 DOI: 10.1371/journal.pone.0124114] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Accepted: 03/10/2015] [Indexed: 01/29/2023] Open
Abstract
Background The Neuregulin 1 transmembrane domain heterozygous mutant (Nrg1 TM HET) mouse is used to investigate the role of Nrg1 in brain function and schizophrenia-like behavioural phenotypes. However, the molecular alterations in brain Nrg1 expression that underpin the behavioural observations have been assumed, but not directly determined. Here we comprehensively characterise mRNA Nrg1 transcripts throughout development of the Nrg1 TM HET mouse. In addition, we investigate the regulation of high-frequency (gamma) electrophysiological oscillations in this mutant mouse to associate molecular changes in Nrg1 with a schizophrenia-relevant neurophysiological profile. Methods Using exonic probes spanning the cysteine-rich, epidermal growth factor (EGF)-like, transmembrane and intracellular domain encoding regions of Nrg1, mRNA levels were measured using qPCR in hippocampus and frontal cortex from male and female Nrg1 TM HET and wild type-like (WT) mice throughout development. We also performed electrophysiological recordings in adult mice and analysed gamma oscillatory at baseline, in responses to auditory stimuli and to ketamine. Results In both hippocampus and cortex, Nrg1 TM HET mice show significantly reduced expression of the exon encoding the transmembrane domain of Nrg1 compared with WT, but unaltered mRNA expression encoding the extracellular bioactive EGF-like and the cysteine-rich (type III) domains, and development-specific and region-specific reductions in the mRNA encoding the intracellular domain. Hippocampal Nrg1 protein expression was not altered, but NMDA receptor NR2B subunit phosphorylation was lower in Nrg1 TM HET mice. We identified elevated ongoing and reduced sensory-evoked gamma power in Nrg1 TM HET mice. Interpretation We found no evidence to support the claim that the Nrg1 TM HET mouse represents a simple haploinsufficient model. Further research is required to explore the possibility that mutation results in a gain of Nrg1 function.
Collapse
Affiliation(s)
- Leonora E. Long
- Schizophrenia Research Institute, Sydney, New South Wales, Australia
- Neuroscience Research Australia, Randwick, New South Wales, Australia
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Paul Anderson
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia
| | - Elisabeth Frank
- Schizophrenia Research Institute, Sydney, New South Wales, Australia
- University of Wollongong, Wollongong, New South Wales, Australia
| | - Alex Shaw
- Schizophrenia Research Institute, Sydney, New South Wales, Australia
- Neuroscience Research Australia, Randwick, New South Wales, Australia
| | - Shijie Liu
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia
| | - Xu-Feng Huang
- Schizophrenia Research Institute, Sydney, New South Wales, Australia
- University of Wollongong, Wollongong, New South Wales, Australia
| | - Didier Pinault
- INSERM U1114, psychopathologie cognitive et physiopathologie de la schizophrénie, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Tim Karl
- Schizophrenia Research Institute, Sydney, New South Wales, Australia
- Neuroscience Research Australia, Randwick, New South Wales, Australia
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Terence J. O’Brien
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia
| | - Cynthia Shannon Weickert
- Schizophrenia Research Institute, Sydney, New South Wales, Australia
- Neuroscience Research Australia, Randwick, New South Wales, Australia
- School of Psychiatry, University of New South Wales, Sydney, New South Wales, Australia
- * E-mail: (CSW); (NCJ)
| | - Nigel C. Jones
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia
- * E-mail: (CSW); (NCJ)
| |
Collapse
|
50
|
Deng C, Pan B, Hu CH, Han M, Huang XF. Differential effects of short- and long-term antipsychotic treatment on the expression of neuregulin-1 and ErbB4 receptors in the rat brain. Psychiatry Res 2015; 225:347-54. [PMID: 25576368 DOI: 10.1016/j.psychres.2014.12.014] [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: 04/08/2014] [Revised: 10/08/2014] [Accepted: 12/03/2014] [Indexed: 01/09/2023]
Abstract
Neuregulin-1 (NRG1) and ErbB4 genes have been identified as candidate genes for schizophrenia. Post-mortem studies indicated that NRG1-ErbB4 signalling is impaired in schizophrenia subjects. This study investigated whether short- or long-term antipsychotic treatment has different effects on the expression of NRG1 and ErbB4 receptors. Female Sprague-Dawley rats were treated orally with either aripiprazole (0.75 mg/kg), haloperidol (0.1 mg/kg), olanzapine (0.5 mg/kg), or vehicle, 3 times/day for 1 or 12 weeks. Western blotting was performed to examine the expression of NRG1 isoforms (135 kDa, 70 kDa and 40 kDa) and ErbB4 receptors. Both 1-week haloperidol and olanzapine treatment increased NRG1-70kDa expression in the hippocampus; haloperidol also up-regulated ErbB4 levels in the prefrontal cortex (PFC). In the 12-week group, aripiprazole decreased the expression of all three NRG1 isoforms and ErbB4 receptors in the PFC, NRG1-70 kDa and -40 kDa in the cingulate cortex (Cg), and NRG1-135 kDa, -70 kDa and ErbB4 receptors in the hippocampus; haloperidol reduced NRG1-135 kDa in the PFC, NRG1-40 kDa in all three brain regions, and ErbB4 receptor levels in the PFC and hippocampus; NRG1-40 kDa in the PFC and Cg was also down-regulated by olanzapine. These results suggest that the time-dependent and region-specific effects of antipsychotics on NRG1-ErbB4 signalling may contribute to the efficacy of antipsychotics to treat schizophrenia.
Collapse
Affiliation(s)
- Chao Deng
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia; Centre for Translational Neuroscience, School of Medicine, University of Wollongong, Wollongong, NSW, Australia; Schizophrenia Research Institute, 384 Victoria Street, Darlinghurst, NSW, Australia.
| | - Bo Pan
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia; Centre for Translational Neuroscience, School of Medicine, University of Wollongong, Wollongong, NSW, Australia
| | - Chang-Hua Hu
- Centre for Translational Neuroscience, School of Medicine, University of Wollongong, Wollongong, NSW, Australia; Institute of Modern Biopharmaceuticals, College of Pharmaceutical Sciences, Southwest University, Beibei, Chongqing, People׳s Republic of China
| | - Mei Han
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia; Centre for Translational Neuroscience, School of Medicine, University of Wollongong, Wollongong, NSW, Australia; Schizophrenia Research Institute, 384 Victoria Street, Darlinghurst, NSW, Australia
| | - Xu-Feng Huang
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia; Centre for Translational Neuroscience, School of Medicine, University of Wollongong, Wollongong, NSW, Australia; Schizophrenia Research Institute, 384 Victoria Street, Darlinghurst, NSW, Australia
| |
Collapse
|