1
|
Walker SL, Glasper ER. Unraveling sex differences in maternal and paternal care impacts on social behaviors and neurobiological responses to early-life adversity. Front Neuroendocrinol 2025; 76:101162. [PMID: 39561882 PMCID: PMC11811932 DOI: 10.1016/j.yfrne.2024.101162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/11/2024] [Accepted: 11/11/2024] [Indexed: 11/21/2024]
Abstract
Early-life stress (ELS) affects the development of prosocial behaviors and social-cognitive function, often leading to structural brain changes and increased psychosocial disorders. Recent studies suggest that mother- and father-child relationships independently influence social development in a sex-specific manner, but the effects of impaired father-child relationships are often overlooked. This review examines preclinical rodent studies to explore how parental neglect impacts neuroplasticity and social behaviors in offspring. We highlight that disruptions in maternal interactions may affect male pups more in uniparental rodents, while impaired paternal interactions in biparental rodents tend to impact female pups more. Due to limited research, the separate effects of maternal and paternal neglect on brain development and social behaviors in biparental species remain unclear. Addressing these gaps could clarify the sex-specific mechanisms underlying social and neurobiological deficits following parental neglect.
Collapse
Affiliation(s)
- Shakeera L Walker
- Department of Neuroscience, The Ohio State University, Columbus, OH, 43210, United States; Neuroscience Graduate Program, The Ohio State University, Columbus, OH, 43210, United States
| | - Erica R Glasper
- Department of Neuroscience, The Ohio State University, Columbus, OH, 43210, United States; Neuroscience Graduate Program, The Ohio State University, Columbus, OH, 43210, United States; Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, 43210, United States.
| |
Collapse
|
2
|
Wu B, Post L, Lin Z, Schonewille M. PP2B-Dependent Cerebellar Plasticity Sets the Amplitude of the Vestibulo-ocular Reflex during Juvenile Development. J Neurosci 2024; 44:e1211232024. [PMID: 38527808 PMCID: PMC11044099 DOI: 10.1523/jneurosci.1211-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 02/01/2024] [Accepted: 02/07/2024] [Indexed: 03/27/2024] Open
Abstract
Throughout life, the cerebellum plays a central role in the coordination and optimization of movements, using cellular plasticity to adapt a range of behaviors. Whether these plasticity processes establish a fixed setpoint during development, or continuously adjust behaviors throughout life, is currently unclear. Here, by spatiotemporally manipulating the activity of protein phosphatase 2B (PP2B), an enzyme critical for cerebellar plasticity in male and female mice, we examined the consequences of disrupted plasticity on the performance and adaptation of the vestibulo-ocular reflex (VOR). We find that, in contrast to Purkinje cell (PC)-specific deletion starting early postnatally, acute pharmacological as well as adult-onset genetic deletion of PP2B affects all forms of VOR adaptation but not the level of VOR itself. Next, we show that PC-specific genetic deletion of PP2B in juvenile mice leads to a progressive loss of the protein PP2B and a concurrent change in the VOR, in addition to the loss of adaptive abilities. Finally, re-expressing PP2B in adult mice that lack PP2B expression from early development rescues VOR adaptation but does not affect the performance of the reflex. Together, our results indicate that chronic or acute, genetic, or pharmacological block of PP2B disrupts the adaptation of the VOR. In contrast, only the absence of plasticity during cerebellar development affects the setpoint of VOR, an effect that cannot be corrected after maturation of the cerebellum. These findings suggest that PP2B-dependent cerebellar plasticity is required during a specific period to achieve the correct setpoint of the VOR.
Collapse
Affiliation(s)
- Bin Wu
- Department of Neurology & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
- Department of Neuroscience, Erasmus MC, Rotterdam 3015CN, The Netherlands
| | - Laura Post
- Department of Neuroscience, Erasmus MC, Rotterdam 3015CN, The Netherlands
| | - Zhanmin Lin
- Department of Neuroscience, Erasmus MC, Rotterdam 3015CN, The Netherlands
| | | |
Collapse
|
3
|
Zhong Y, Zhang N, Zhao F, Chang S, Chen W, Cao Q, Sun L, Wang Y, Gong Z, Lu L, Liu D, Yang L. RBFOX1 and Working Memory: From Genome to Transcriptome Revealed Posttranscriptional Mechanism Separate From Attention-Deficit/Hyperactivity Disorder. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2023; 3:1042-1052. [PMID: 37881587 PMCID: PMC10593897 DOI: 10.1016/j.bpsgos.2022.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 08/09/2022] [Accepted: 08/12/2022] [Indexed: 11/22/2022] Open
Abstract
Background Many psychiatric disorders share a working memory (WM) impairment phenotype, yet the genetic causes remain unclear. Here, we generated genetic profiles of WM deficits using attention-deficit/hyperactivity disorder samples and validated the results in zebrafish models. Methods We used 2 relatively large attention-deficit/hyperactivity disorder cohorts, 799 and 776 cases, respectively. WM impairment was assessed using the Rey Complex Figure Test. First, association analyses were conducted at single-variant, gene-based, and gene-set levels. Deeper insights into the biological mechanism were gained from further functional exploration by bioinformatic analyses and zebrafish models. Results Genomic analyses identified and replicated a locus with rs75885813 as the index single nucleotide polymorphism showing significant association with WM defects but not with attention-deficit/hyperactivity disorder. Functional feature exploration found that these single nucleotide polymorphisms may regulate the expression level of RBFOX1 through chromatin interaction. Further pathway enrichment analysis of potential associated single nucleotide polymorphisms revealed the involvement of posttranscription regulation that affects messenger RNA stability and/or alternative splicing. Zebrafish with functionally knocked down or genome-edited rbfox1 exhibited WM impairment but no hyperactivity. Transcriptome profiling of rbfox1-defective zebrafish indicated that alternative exon usages of snap25a might partially lead to reduced WM learning of larval zebrafish. Conclusions The locus with rs75885813 in RBFOX1 was identified as associated with WM. Rbfox1 regulates synaptic and long-term potentiation-related gene snap25a to adjust WM at the posttranscriptional level.
Collapse
Affiliation(s)
- Yuanxin Zhong
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health, National Clinical Research Center for Mental Disorders, Beijing, China
| | - Na Zhang
- School of Life Science, Southern University of Science and Technology, Shenzhen, China
- Department of Biological Science, National University of Singapore, Singapore
| | - Feng Zhao
- School of Life Science, Southern University of Science and Technology, Shenzhen, China
| | - Suhua Chang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health, National Clinical Research Center for Mental Disorders, Beijing, China
| | - Wei Chen
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health, National Clinical Research Center for Mental Disorders, Beijing, China
| | - Qingjiu Cao
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health, National Clinical Research Center for Mental Disorders, Beijing, China
| | - Li Sun
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health, National Clinical Research Center for Mental Disorders, Beijing, China
| | - Yufeng Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health, National Clinical Research Center for Mental Disorders, Beijing, China
| | - Zhiyuan Gong
- Department of Biological Science, National University of Singapore, Singapore
| | - Lin Lu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health, National Clinical Research Center for Mental Disorders, Beijing, China
- Peking-Tsinghua Center for Life Sciences, International Data Group, McGovern Institute for Brain Research at Peking University, Peking University, Beijing, China
| | - Dong Liu
- School of Life Science, Southern University of Science and Technology, Shenzhen, China
| | - Li Yang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health, National Clinical Research Center for Mental Disorders, Beijing, China
| |
Collapse
|
4
|
Wu Y, Xu D, He Y, Yan Z, Liu R, Liu Z, He C, Liu X, Yu Y, Yang X, Pan W. Dimethyl itaconate ameliorates the deficits of goal-directed behavior in Toxoplasma gondii infected mice. PLoS Negl Trop Dis 2023; 17:e0011350. [PMID: 37256871 DOI: 10.1371/journal.pntd.0011350] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 05/02/2023] [Indexed: 06/02/2023] Open
Abstract
BACKGROUND The neurotrophic parasite Toxoplasma gondii (T. gondii) has been implicated as a risk factor for neurodegenerative diseases. However, there is only limited information concerning its underlying mechanism and therapeutic strategy. Here, we investigated the effects of T. gondii chronic infection on the goal-directed cognitive behavior in mice. Moreover, we evaluated the preventive and therapeutic effect of dimethyl itaconate on the behavior deficits induced by the parasite. METHODS The infection model was established by orally infecting the cysts of T. gondii. Dimethyl itaconate was intraperitoneally administered before or after the infection. Y-maze and temporal order memory (TOM) tests were used to evaluate the prefrontal cortex-dependent behavior performance. Golgi staining, transmission electron microscopy, indirect immunofluorescence, western blot, and RNA sequencing were utilized to determine the pathological changes in the prefrontal cortex of mice. RESULTS We showed that T. gondii infection impaired the prefrontal cortex-dependent goal-directed behavior. The infection significantly downregulated the expression of the genes associated with synaptic transmission, plasticity, and cognitive behavior in the prefrontal cortex of mice. On the contrary, the infection robustly upregulated the expression of activation makers of microglia and astrocytes. In addition, the metabolic phenotype of the prefrontal cortex post infection was characterized by the enhancement of glycolysis and fatty acid oxidation, the blockage of the Krebs cycle, and the disorder of aconitate decarboxylase 1 (ACOD1)-itaconate axis. Notably, the administration of dimethyl itaconate significantly prevented and treated the cognitive impairment induced by T. gondii, which was evidenced by the improvement of behavioral deficits, synaptic ultrastructure lesion and neuroinflammation. CONCLUSION The present study demonstrates that T. gondii infection induces the deficits of the goal-directed behavior, which is associated with neuroinflammation, the impairment of synaptic ultrastructure, and the metabolic shifts in the prefrontal cortex of mice. Moreover, we report that dimethyl itaconate has the potential to prevent and treat the behavior deficits.
Collapse
Affiliation(s)
- Yongshuai Wu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, China
- National Experimental Teaching Demonstration Center of Basic Medicine (Xuzhou Medical University), Xuzhou, China
| | - Daxiang Xu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Yan He
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, China
- National Experimental Teaching Demonstration Center of Basic Medicine (Xuzhou Medical University), Xuzhou, China
| | - Ziyi Yan
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, China
- National Experimental Teaching Demonstration Center of Basic Medicine (Xuzhou Medical University), Xuzhou, China
| | - Rundong Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, China
- National Experimental Teaching Demonstration Center of Basic Medicine (Xuzhou Medical University), Xuzhou, China
| | - Zhuanzhuan Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- National Experimental Teaching Demonstration Center of Basic Medicine (Xuzhou Medical University), Xuzhou, China
| | - Cheng He
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- National Experimental Teaching Demonstration Center of Basic Medicine (Xuzhou Medical University), Xuzhou, China
| | - Xiaomei Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- National Experimental Teaching Demonstration Center of Basic Medicine (Xuzhou Medical University), Xuzhou, China
| | - Yinghua Yu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- National Experimental Teaching Demonstration Center of Basic Medicine (Xuzhou Medical University), Xuzhou, China
| | - Xiaoying Yang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- National Experimental Teaching Demonstration Center of Basic Medicine (Xuzhou Medical University), Xuzhou, China
| | - Wei Pan
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- National Experimental Teaching Demonstration Center of Basic Medicine (Xuzhou Medical University), Xuzhou, China
| |
Collapse
|
5
|
Malter JS. Pin1 and Alzheimer's disease. Transl Res 2023; 254:24-33. [PMID: 36162703 PMCID: PMC10111655 DOI: 10.1016/j.trsl.2022.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/29/2022] [Accepted: 09/19/2022] [Indexed: 10/14/2022]
Abstract
Alzheimer's disease (AD) is an immense and growing public health crisis. Despite over 100 years of investigation, the etiology remains elusive and therapy ineffective. Despite current gaps in knowledge, recent studies have identified dysfunction or loss-of-function of Pin1, a unique cis-trans peptidyl prolyl isomerase, as an important step in AD pathogenesis. Here I review the functionality of Pin1 and its role in neurodegeneration.
Collapse
Affiliation(s)
- James S Malter
- Department of Pathology, UT Southwestern Medical Center, 5333 Harry Hines Blvd, Dallas, TX 75390.
| |
Collapse
|
6
|
Pan W, Zhao J, Wu J, Xu D, Meng X, Jiang P, Shi H, Ge X, Yang X, Hu M, Zhang P, Tang R, Nagaratnam N, Zheng K, Huang XF, Yu Y. Dimethyl itaconate ameliorates cognitive impairment induced by a high-fat diet via the gut-brain axis in mice. MICROBIOME 2023; 11:30. [PMID: 36810115 PMCID: PMC9942412 DOI: 10.1186/s40168-023-01471-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 01/24/2023] [Indexed: 05/29/2023]
Abstract
BACKGROUND Gut homeostasis, including intestinal immunity and microbiome, is essential for cognitive function via the gut-brain axis. This axis is altered in high-fat diet (HFD)-induced cognitive impairment and is closely associated with neurodegenerative diseases. Dimethyl itaconate (DI) is an itaconate derivative and has recently attracted extensive interest due to its anti-inflammatory effect. This study investigated whether intraperitoneal administration of DI improves the gut-brain axis and prevents cognitive deficits in HF diet-fed mice. RESULTS DI effectively attenuated HFD-induced cognitive decline in behavioral tests of object location, novel object recognition, and nesting building, concurrent with the improvement of hippocampal RNA transcription profiles of genes associated with cognition and synaptic plasticity. In agreement, DI reduced the damage of synaptic ultrastructure and deficit of proteins (BDNF, SYN, and PSD95), the microglial activation, and neuroinflammation in the HFD-fed mice. In the colon, DI significantly lowered macrophage infiltration and the expression of pro-inflammatory cytokines (TNF-α, IL-1β, IL-6) in mice on the HF diet, while upregulating the expression of immune homeostasis-related cytokines (IL-22, IL-23) and antimicrobial peptide Reg3γ. Moreover, DI alleviated HFD-induced gut barrier impairments, including elevation of colonic mucus thickness and expression of tight junction proteins (zonula occludens-1, occludin). Notably, HFD-induced microbiome alteration was improved by DI supplementation, characterized by the increase of propionate- and butyrate-producing bacteria. Correspondingly, DI increased the levels of propionate and butyrate in the serum of HFD mice. Intriguingly, fecal microbiome transplantation from DI-treated HF mice facilitated cognitive variables compared with HF mice, including higher cognitive indexes in behavior tests and optimization of hippocampal synaptic ultrastructure. These results highlight the gut microbiota is necessary for the effects of DI in improving cognitive impairment. CONCLUSIONS The present study provides the first evidence that DI improves cognition and brain function with significant beneficial effects via the gut-brain axis, suggesting that DI may serve as a novel drug for treating obesity-associated neurodegenerative diseases. Video Abstract.
Collapse
Affiliation(s)
- Wei Pan
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Jinxiu Zhao
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Jiacheng Wu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- The Second School of Clinical Medicine, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Daxiang Xu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Xianran Meng
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Pengfei Jiang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Hongli Shi
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Xing Ge
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Xiaoying Yang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Minmin Hu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Peng Zhang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Renxian Tang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Nathan Nagaratnam
- Illawarra Health and Medical Research Institute (IHMRI) and School of Medicine, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Kuiyang Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| | - Xu-Feng Huang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- Illawarra Health and Medical Research Institute (IHMRI) and School of Medicine, University of Wollongong, Wollongong, NSW, 2522, Australia.
| | - Yinghua Yu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
7
|
Hagihara H, Shoji H, Kuroiwa M, Graef IA, Crabtree GR, Nishi A, Miyakawa T. Forebrain-specific conditional calcineurin deficiency induces dentate gyrus immaturity and hyper-dopaminergic signaling in mice. Mol Brain 2022; 15:94. [PMID: 36414974 PMCID: PMC9682671 DOI: 10.1186/s13041-022-00981-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 11/12/2022] [Indexed: 11/24/2022] Open
Abstract
Calcineurin (Cn), a phosphatase important for synaptic plasticity and neuronal development, has been implicated in the etiology and pathophysiology of neuropsychiatric disorders, including schizophrenia, intellectual disability, autism spectrum disorders, epilepsy, and Alzheimer's disease. Forebrain-specific conditional Cn knockout mice have been known to exhibit multiple behavioral phenotypes related to these disorders. In this study, we investigated whether Cn mutant mice show pseudo-immaturity of the dentate gyrus (iDG) in the hippocampus, which we have proposed as an endophenotype shared by these disorders. Expression of calbindin and GluA1, typical markers for mature DG granule cells (GCs), was decreased and that of doublecortin, calretinin, phospho-CREB, and dopamine D1 receptor (Drd1), markers for immature GC, was increased in Cn mutants. Phosphorylation of cAMP-dependent protein kinase (PKA) substrates (GluA1, ERK2, DARPP-32, PDE4) was increased and showed higher sensitivity to SKF81297, a Drd1-like agonist, in Cn mutants than in controls. While cAMP/PKA signaling is increased in the iDG of Cn mutants, chronic treatment with rolipram, a selective PDE4 inhibitor that increases intracellular cAMP, ameliorated the iDG phenotype significantly and nesting behavior deficits with nominal significance. Chronic rolipram administration also decreased the phosphorylation of CREB, but not the other four PKA substrates examined, in Cn mutants. These results suggest that Cn deficiency induces pseudo-immaturity of GCs and that cAMP signaling increases to compensate for this maturation abnormality. This study further supports the idea that iDG is an endophenotype shared by certain neuropsychiatric disorders.
Collapse
Affiliation(s)
- Hideo Hagihara
- Division of Systems Medical Science, Center for Medical Science, Fujita Health University, Toyoake, Aichi 470-1192 Japan
| | - Hirotaka Shoji
- Division of Systems Medical Science, Center for Medical Science, Fujita Health University, Toyoake, Aichi 470-1192 Japan
| | - Mahomi Kuroiwa
- Department of Pharmacology, Kurume University School of Medicine, Kurume, Fukuoka 830-0011 Japan
| | - Isabella A. Graef
- Department of Pathology, Stanford University of Medicine, Stanford, CA 94305 USA
| | - Gerald R. Crabtree
- Department of Pathology, Stanford University of Medicine, Stanford, CA 94305 USA
| | - Akinori Nishi
- Department of Pharmacology, Kurume University School of Medicine, Kurume, Fukuoka 830-0011 Japan
| | - Tsuyoshi Miyakawa
- Division of Systems Medical Science, Center for Medical Science, Fujita Health University, Toyoake, Aichi 470-1192 Japan
| |
Collapse
|
8
|
Sun W, Mei Y, Li X, Yang Y, An L. Maternal immune activation-induced proBDNF-mediated neural information processing dysfunction at hippocampal CA3-CA1 synapses associated with memory deficits in offspring. Front Cell Dev Biol 2022; 10:1018586. [PMID: 36438556 PMCID: PMC9691851 DOI: 10.3389/fcell.2022.1018586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 10/27/2022] [Indexed: 11/07/2023] Open
Abstract
Prenatal exposure to maternal infection increases the risk of offspring developing schizophrenia in adulthood. Current theories suggest that the consequences of MIA on mBDNF secretion may underlie the increased risk of cognitive disorder. There is little evidence for whether the expression of its precursor, proBDNF, is changed and how proBDNF-mediated signaling may involve in learning and memory. In this study, proBDNF levels were detected in the hippocampal CA1 and CA3 regions of male adult rats following MIA by prenatal polyI:C exposure. Behaviorally, learning and memory were assessed in contextual fear conditioning tasks. Local field potentials were recorded in the hippocampal CA3-CA1 pathway. The General Partial Directed Coherence approach was utilized to identify the directional alternation of neural information flow between CA3 and CA1 regions. EPSCs were recorded in CA1 pyramidal neurons to explore a possible mechanism involving the proBDNF-p75NTR signaling pathway. Results showed that the expression of proBDNF in the polyI:C-treated offspring was abnormally enhanced in both CA3 and CA1 regions. Meanwhile, the mBDNF expression was reduced in both hippocampal regions. Intra-hippocampal CA1 but not CA3 injection with anti-proBDNF antibody and p75NTR inhibitor TAT-Pep5 effectively mitigated the contextual memory deficits. Meanwhile, reductions in the phase synchronization between CA3 and CA1 and the coupling directional indexes from CA3 to CA1 were enhanced by the intra-CA1 infusions. Moreover, blocking proBDNF/p75NTR signaling could reverse the declined amplitude of EPSCs in CA1 pyramidal neurons, indicating the changes in postsynaptic information processing in the polyI:C-treated offspring. Therefore, the changes in hippocampal proBDNF activity in prenatal polyI:C exposure represent a potential mechanism involved in NIF disruption leading to contextual memory impairments.
Collapse
Affiliation(s)
- Wei Sun
- Department of Pediatric, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
- Behavioral Neuroscience Laboratory, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Yazi Mei
- Graduate School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoliang Li
- Department of Neurology, Jinan Geriatric/Rehabilitation Hospital, Jinan, China
| | - Yang Yang
- Department of Pediatric, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Lei An
- Department of Pediatric, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
- Behavioral Neuroscience Laboratory, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
- Department of Neurology, Jinan Geriatric/Rehabilitation Hospital, Jinan, China
- Department of Neurology, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| |
Collapse
|
9
|
Hou TY, Cai WP. What emotion dimensions can affect working memory performance in healthy adults? A review. World J Clin Cases 2022; 10:401-411. [PMID: 35097065 PMCID: PMC8771390 DOI: 10.12998/wjcc.v10.i2.401] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 08/28/2021] [Accepted: 11/30/2021] [Indexed: 02/06/2023] Open
Abstract
Due to the critical roles of emotion and working memory in our daily activities, a great deal of attention has been given to how emotion influences working memory performance. Although the association between emotion and working memory is relatively well established, whether mood enhances or impairs working memory performance remains controversial. The present review provides a relatively representative overview of the research on the effect of different dimensions of emotion on working memory among healthy adults spanning a 30-year period. The findings show that the valence, arousal and motivational dimensions of emotion could all exert an impact on working memory performance. The impact of emotion on working memory might be modulated by task relevance, emotion type, working memory paradigms and individual differences. The vast majority of the studies regarding the effect of emotion on working memory performance focused on the impact of negatively valenced affect and yielded highly contradictory findings. The impacts of arousal and motivation on working memory have been less explored, and inconsistent findings have also been reported. Possible explanations are discussed. Considerable research on the effect of certain dimensions of emotion on working memory has suffered from a lack of control of other emotional dimensions, and different aspects of working memory have been investigated by various paradigms. Directions for further studies should include the exploration of specific dimensions of emotion on different aspects of working memory, with the other dimensions being well controlled.
Collapse
Affiliation(s)
- Tian-Ya Hou
- Faculty of Psychology, The Second Military Medical University, Shanghai 200433, China
| | - Wen-Peng Cai
- Faculty of Psychology, The Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
10
|
Banki E, Fisi V, Moser S, Wengi A, Carrel M, Loffing-Cueni D, Penton D, Kratschmar DV, Rizzo L, Lienkamp S, Odermatt A, Rinschen MM, Loffing J. Specific disruption of calcineurin-signaling in the distal convoluted tubule impacts the transcriptome and proteome, and causes hypomagnesemia and metabolic acidosis. Kidney Int 2021; 100:850-869. [PMID: 34252449 DOI: 10.1016/j.kint.2021.06.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 06/11/2021] [Accepted: 06/18/2021] [Indexed: 12/27/2022]
Abstract
Adverse effects of calcineurin inhibitors (CNI), such as hypertension, hyperkalemia, acidosis, hypomagnesemia and hypercalciuria, have been linked to dysfunction of the distal convoluted tubule (DCT). To test this, we generated a mouse model with an inducible DCT-specific deletion of the calcineurin regulatory subunit B alpha (CnB1-KO). Three weeks after CnB1 deletion, these mice exhibited hypomagnesemia and acidosis, but no hypertension, hyperkalemia or hypercalciuria. Consistent with the hypomagnesemia, CnB1-KO mice showed a downregulation of proteins implicated in DCT magnesium transport, including TRPM6, CNNM2, SLC41A3 and parvalbumin but expression of calcium channel TRPV5 in the kidney was unchanged. The abundance of the chloride/bicarbonate exchanger pendrin was increased, likely explaining the acidosis. Plasma aldosterone levels, kidney renin expression, abundance of phosphorylated sodium chloride-cotransporter and abundance of the epithelial sodium channel were similar in control and CnB1-KO mice, consistent with a normal sodium balance. Long-term potassium homeostasis was maintained in CnB1-KO mice, but in-vivo and ex-vivo experiments indicated that CnB1 contributes to acute regulation of potassium balance and sodium chloride-cotransporter. Tacrolimus treatment of control and CnB1-KO mice demonstrated that CNI-related hypomagnesemia is linked to impaired calcineurin-signaling in DCT, while hypocalciuria and hyponatremia occur independently of CnB1 in DCT. Transcriptome and proteome analyses of isolated DCTs demonstrated that CnB1 deletion impacts the expression of several DCT-specific proteins and signaling pathways. Thus, our data support a critical role of calcineurin for DCT function and provide novel insights into the pathophysiology of CNI side-effects and involved molecular players in the DCT.
Collapse
Affiliation(s)
- Eszter Banki
- Institute of Anatomy, University of Zurich, Zurich, Switzerland; Swiss National Centre for Competence in Research "Kidney Control of Homeostasis," Zurich, Switzerland
| | - Viktoria Fisi
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Sandra Moser
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Agnieszka Wengi
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Monique Carrel
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | | | - David Penton
- Institute of Anatomy, University of Zurich, Zurich, Switzerland; Swiss National Centre for Competence in Research "Kidney Control of Homeostasis," Zurich, Switzerland
| | - Denise V Kratschmar
- Department of Pharmaceutical Sciences, Division of Molecular and Systems Toxicology, University of Basel, Basel, Switzerland
| | - Ludovica Rizzo
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Soeren Lienkamp
- Institute of Anatomy, University of Zurich, Zurich, Switzerland; Swiss National Centre for Competence in Research "Kidney Control of Homeostasis," Zurich, Switzerland
| | - Alex Odermatt
- Department of Pharmaceutical Sciences, Division of Molecular and Systems Toxicology, University of Basel, Basel, Switzerland
| | - Markus M Rinschen
- Kidney Research Center, University of Cologne, Köln, Germany; Department of Biomedicine, Aarhus University, Aarhus, Denmark; III Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Johannes Loffing
- Institute of Anatomy, University of Zurich, Zurich, Switzerland; Swiss National Centre for Competence in Research "Kidney Control of Homeostasis," Zurich, Switzerland.
| |
Collapse
|
11
|
Lin Z, Wu B, Paul MW, Li KW, Yao Y, Smal I, Proietti Onori M, Hasanbegovic H, Bezstarosti K, Demmers J, Houtsmuller AB, Meijering E, Hoebeek FE, Schonewille M, Smit AB, Gao Z, De Zeeuw CI. Protein Phosphatase 2B Dual Function Facilitates Synaptic Integrity and Motor Learning. J Neurosci 2021; 41:5579-5594. [PMID: 34021041 PMCID: PMC8244972 DOI: 10.1523/jneurosci.1741-20.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 04/01/2021] [Accepted: 04/23/2021] [Indexed: 11/21/2022] Open
Abstract
Protein phosphatase 2B (PP2B) is critical for synaptic plasticity and learning, but the molecular mechanisms involved remain unclear. Here we identified different types of proteins that interact with PP2B, including various structural proteins of the postsynaptic densities (PSDs) of Purkinje cells (PCs) in mice. Deleting PP2B reduced expression of PSD proteins and the relative thickness of PSD at the parallel fiber to PC synapses, whereas reexpression of inactive PP2B partly restored the impaired distribution of nanoclusters of PSD proteins, together indicating a structural role of PP2B. In contrast, lateral mobility of surface glutamate receptors solely depended on PP2B phosphatase activity. Finally, the level of motor learning covaried with both the enzymatic and nonenzymatic functions of PP2B. Thus, PP2B controls synaptic function and learning both through its action as a phosphatase and as a structural protein that facilitates synapse integrity.SIGNIFICANCE STATEMENT Phosphatases are generally considered to serve their critical role in learning and memory through their enzymatic operations. Here, we show that protein phosphatase 2B (PP2B) interacts with structural proteins at the synapses of cerebellar Purkinje cells. Differentially manipulating the enzymatic and structural domains of PP2B leads to different phenotypes in cerebellar learning. We propose that PP2B is crucial for cerebellar learning via two complementary actions, an enzymatic and a structural operation.
Collapse
Affiliation(s)
- Zhanmin Lin
- Department of Neuroscience, Erasmus MC, 3015 GE, Rotterdam, The Netherlands
| | - Bin Wu
- Department of Neuroscience, Erasmus MC, 3015 GE, Rotterdam, The Netherlands
- Department of Neurology and Institute of Neurology, Huashan Hospital, Fudan University, 200040, Shanghai, China
| | - Maarten W Paul
- Optical Imaging Center, Erasmus MC, 3015 GE, Rotterdam, The Netherlands
| | - Ka Wan Li
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, Amsterdam Neuroscience, 1081 HV, Amsterdam, The Netherlands
| | - Yao Yao
- Department of Medical informatics, Erasmus MC, 3015 GE, Rotterdam, The Netherlands
| | - Ihor Smal
- Department of Medical informatics, Erasmus MC, 3015 GE, Rotterdam, The Netherlands
| | | | - Hana Hasanbegovic
- Department of Neuroscience, Erasmus MC, 3015 GE, Rotterdam, The Netherlands
| | - Karel Bezstarosti
- Center for Proteomics, Erasmus MC, 3015 GE, Rotterdam, The Netherlands
| | - Jeroen Demmers
- Center for Proteomics, Erasmus MC, 3015 GE, Rotterdam, The Netherlands
| | | | - Erik Meijering
- School of Computer Science and Engineering & Graduate School of Biomedical Engineering, University of New South Wales, Sydney, 2052, New South Wales, Australia
| | - Freek E Hoebeek
- Department of Neuroscience, Erasmus MC, 3015 GE, Rotterdam, The Netherlands
- Department for Developmental Origins of Disease, Wilhelmina Children's Hospital and Brain Center, Utrecht Medical Center, 3584 EA, Utrecht, The Netherlands
| | | | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, Amsterdam Neuroscience, 1081 HV, Amsterdam, The Netherlands
| | - Zhenyu Gao
- Department of Neuroscience, Erasmus MC, 3015 GE, Rotterdam, The Netherlands
| | - Chris I De Zeeuw
- Department of Neuroscience, Erasmus MC, 3015 GE, Rotterdam, The Netherlands
- Netherlands Institute for Neuroscience, KNAW, 1105 BA, Amsterdam, The Netherlands
| |
Collapse
|
12
|
Wang JY, Li XY, Li HJ, Liu JW, Yao YG, Li M, Xiao X, Luo XJ. Integrative Analyses Followed by Functional Characterization Reveal TMEM180 as a Schizophrenia Risk Gene. Schizophr Bull 2021; 47:1364-1374. [PMID: 33768244 PMCID: PMC8379544 DOI: 10.1093/schbul/sbab032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Recent large-scale integrative analyses (including Transcriptome-Wide Association Study [TWAS] and Summary-data-based Mendelian Randomization [SMR]) have identified multiple genes whose cis-regulated expression changes may confer risk of schizophrenia. Nevertheless, expression quantitative trait loci (eQTL) data and genome-wide associations used for integrative analyses were mainly from populations of European ancestry, resulting in potential missing of pivotal biological insights in other continental populations due to population heterogeneity. Here we conducted TWAS and SMR integrative analyses using blood eQTL (from 162 subjects) and GWAS data (22 778 cases and 35 362 controls) of schizophrenia in East Asian (EAS) populations. Both TWAS (P = 2.89 × 10-14) and SMR (P = 6.04 × 10-5) analyses showed that decreased TMEM180 mRNA expression was significantly associated with risk of schizophrenia. We further found that TMEM180 was significantly down-regulated in the peripheral blood of schizophrenia cases compared with controls (P = 8.63 × 10-4 in EAS sample), and its expression was also significantly lower in the brain tissues of schizophrenia cases compared with controls (P = 1.87 × 10-5 in European sample from PsychENCODE). Functional explorations suggested that Tmem180 knockdown affected neurodevelopment, ie, proliferation and differentiation of neural stem cells. RNA sequencing showed that pathways regulated by Tmem180 were significantly enriched in brain development and synaptic transmission. In conclusion, our study provides convergent lines of evidence for the involvement of TMEM180 in schizophrenia, and highlights the potential and importance of resource integration and sharing at this big data era in bio-medical research.
Collapse
Affiliation(s)
- Jun-Yang Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Xiao-Yan Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Hui-Juan Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Jie-Wei Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Yong-Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China,KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China,CAS Center for Excellence in Brain Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China,KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China,CAS Center for Excellence in Brain Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiao Xiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Xiong-Jian Luo
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China,KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, Yunnan 650223, China,To whom correspondence should be addressed; Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; tel: +86-871-68125413, fax: +86-871-68125413, e-mail:
| |
Collapse
|
13
|
Xu H, Zhou W, Zhan L, Sui H, Zhang L, Zhao C, Lu X. The ZiBuPiYin recipe regulates proteomic alterations in brain mitochondria-associated ER membranes caused by chronic psychological stress exposure: Implications for cognitive decline in Zucker diabetic fatty rats. Aging (Albany NY) 2020; 12:23698-23726. [PMID: 33221746 PMCID: PMC7762487 DOI: 10.18632/aging.103894] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 07/23/2020] [Indexed: 01/10/2023]
Abstract
Chronic psychological stress (PS) cumulatively affects memory performance through the deleterious effects on hypothalamic-pituitary-adrenal axis regulation. Several functions damaged in cognitive impairment-related diseases are regulated by mitochondria-associated ER membranes (MAMs). To elucidate the role of ZiBuPiYin recipe (ZBPYR) in regulating the MAM proteome to improve PS-induced diabetes-associated cognitive decline (PSD), differentially expressed MAM proteins were identified among Zucker diabetic fatty rats, PSD rats, and PS combined with ZBPYR administration rats via iTRAQ with LC-MS/MS. Proteomic analysis revealed that the expressions of 85 and 33 proteins were altered by PS and ZBPYR treatment, respectively. Among these, 21 proteins were differentially expressed under both PS and ZBPYR treatments, whose functional categories included energy metabolism, lipid and protein metabolism, and synaptic dysfunction. Furthermore, calcium signaling and autophagy-related proteins may play roles in the pathogenesis of PSD and the mechanism of ZBPYR, respectively. Notably, KEGG pathway analysis suggested that ‘Alzheimer's disease’ and ‘oxidative phosphorylation’ pathways may be impaired in PSD pathogenesis, while ZBPYR could play a neuroprotective role through regulating the above pathways. Overall, exposure to chronic PS contributes to the evolution of diabetes-associated cognitive decline and ZBPYR might prevent and treat PSD by regulating the MAM proteome.
Collapse
Affiliation(s)
- Huiying Xu
- Modern Research Laboratory of Spleen Visceral Manifestations Theory, School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wen Zhou
- Modern Research Laboratory of Spleen Visceral Manifestations Theory, School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Libin Zhan
- Modern Research Laboratory of Spleen Visceral Manifestations Theory, School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Hua Sui
- Institute of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Lijing Zhang
- Modern Research Laboratory of Spleen Visceral Manifestations Theory, School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Chunyan Zhao
- Modern Research Laboratory of Spleen Visceral Manifestations Theory, School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiaoguang Lu
- Department of Emergency Medicine, Zhongshan Hospital, Dalian University, Dalian 116001, China
| |
Collapse
|
14
|
Systems biology reveals reprogramming of the S-nitroso-proteome in the cortical and striatal regions of mice during aging process. Sci Rep 2020; 10:13913. [PMID: 32807865 PMCID: PMC7431412 DOI: 10.1038/s41598-020-70383-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 07/28/2020] [Indexed: 12/26/2022] Open
Abstract
Cell aging depends on the rate of cumulative oxidative and nitrosative damage to DNA and proteins. Accumulated data indicate the involvement of protein S-nitrosylation (SNO), the nitric oxide (NO)-mediated posttranslational modification (PTM) of cysteine thiols, in different brain disorders. However, the changes and involvement of SNO in aging including the development of the organism from juvenile to adult state is still unknown. In this study, using the state-of-the-art mass spectrometry technology to identify S-nitrosylated proteins combined with large-scale computational biology, we tested the S-nitroso-proteome in juvenile and adult mice in both cortical and striatal regions. We found reprogramming of the S-nitroso-proteome in adult mice of both cortex and striatum regions. Significant biological processes and protein–protein clusters associated with synaptic and neuronal terms were enriched in adult mice. Extensive quantitative analysis revealed a large set of potentially pathological proteins that were significantly upregulated in adult mice. Our approach, combined with large scale computational biology allowed us to perform a system-level characterization and identification of the key proteins and biological processes that can serve as drug targets for aging and brain disorders in future studies.
Collapse
|
15
|
Speigel IA, Ma CM, Bichler EK, Gooch JL, García PS. Chronic Calcineurin Inhibition via Cyclosporine A Impairs Visuospatial Learning After Isoflurane Anesthesia. Anesth Analg 2020; 129:192-203. [PMID: 31082969 DOI: 10.1213/ane.0000000000004183] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Clinical studies implicate the perioperative period in cognitive complications, and increasing experimental evidence shows that the anesthetic agents can affect neuronal processes that underpin learning and memory. Calcineurin, a Ca-dependent phosphatase critically involved in synaptic plasticity, is activated after isoflurane exposure, but its role in the neurological response to anesthesia is unclear. METHODS We investigated the effect of chronic calcineurin inhibition on postanesthetic cognitive function. Mice were treated with 30 minutes of isoflurane anesthesia during a chronic cyclosporine A regimen. Behavioral end points during the perianesthesia period were quantified. Visuospatial learning was assessed with the water radial arm maze. Total and biotinylated surface protein expression of the α5β3γ2 γ-aminobutyric acid (GABA) type A receptors was measured. Expression of the GABA synthesis enzyme glutamate decarboxylase (GAD)-67 was also measured. RESULTS Mice treated with cyclosporine A before anesthesia showed significant deficits in visuospatial learning compared to sham and cyclosporine A-treated mice (n = 10 per group, P = .0152, Tukey post hoc test). Induction and emergence were unaltered by cyclosporine A. Analysis of hippocampal protein expression revealed an increased surface expression of the α5 GABA type A receptor subunit after isoflurane treatment (P = .019, Dunnett post hoc testing), as well as a decrease in GAD-67 expression. Cyclosporine A did not rescue either effect. CONCLUSIONS Our results confirm the work of others that isoflurane induces changes to inhibitory network function and exclude calcineurin inhibition via cyclosporine A as an intervention. Further, our studies suggest that calcineurin mediates a protective role in the neurological response to anesthesia, and patients receiving cyclosporine A may be an at-risk group for memory problems related to anesthesia.
Collapse
Affiliation(s)
- Iris A Speigel
- From the Neuroanesthesia Laboratory, Atlanta Veterans Affairs Medical Center/Emory University, Atlanta, Georgia.,Department of Anesthesiology, Emory University, Atlanta, Georgia
| | - Christopher M Ma
- Department of Anesthesiology, Emory University, Atlanta, Georgia.,Department of Nephrology, Emory University School of Medicine, Atlanta, Georgia
| | - Edyta K Bichler
- From the Neuroanesthesia Laboratory, Atlanta Veterans Affairs Medical Center/Emory University, Atlanta, Georgia.,Department of Anesthesiology, Emory University, Atlanta, Georgia
| | - Jennifer L Gooch
- Department of Nephrology, Emory University School of Medicine, Atlanta, Georgia
| | - Paul S García
- From the Neuroanesthesia Laboratory, Atlanta Veterans Affairs Medical Center/Emory University, Atlanta, Georgia.,Department of Anesthesiology, Emory University, Atlanta, Georgia
| |
Collapse
|
16
|
Reay WR, Cairns MJ. The role of the retinoids in schizophrenia: genomic and clinical perspectives. Mol Psychiatry 2020; 25:706-718. [PMID: 31666680 PMCID: PMC7156347 DOI: 10.1038/s41380-019-0566-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 09/23/2019] [Accepted: 10/17/2019] [Indexed: 12/13/2022]
Abstract
Signalling by retinoid compounds is vital for embryonic development, with particular importance for neurogenesis in the human brain. Retinoids, metabolites of vitamin A, exert influence over the expression of thousands of transcripts genome wide, and thus, act as master regulators of many important biological processes. A significant body of evidence in the literature now supports dysregulation of the retinoid system as being involved in the aetiology of schizophrenia. This includes mechanistic insights from large-scale genomic, transcriptomic and, proteomic studies, which implicate disruption of disparate aspects of retinoid biology such as transport, metabolism, and signalling. As a result, retinoids may present a valuable clinical opportunity in schizophrenia via novel pharmacotherapies and dietary intervention. Further work, however, is required to expand on the largely observational data collected thus far and confirm causality. This review will highlight the fundamentals of retinoid biology and examine the evidence for retinoid dysregulation in schizophrenia.
Collapse
Affiliation(s)
- William R. Reay
- 0000 0000 8831 109Xgrid.266842.cSchool of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW Australia ,grid.413648.cCentre for Brain and Mental Health Research, Hunter Medical Research Institute, Newcastle, NSW Australia
| | - Murray J. Cairns
- 0000 0000 8831 109Xgrid.266842.cSchool of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW Australia ,grid.413648.cCentre for Brain and Mental Health Research, Hunter Medical Research Institute, Newcastle, NSW Australia
| |
Collapse
|
17
|
Role of Actin Filament on Synaptic Vesicle Pooling in Cultured Hippocampal Neuron. Appl Microsc 2018. [DOI: 10.9729/am.2018.48.3.55] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
18
|
Tarasova EO, Gaydukov AE, Balezina OP. Calcineurin and Its Role in Synaptic Transmission. BIOCHEMISTRY (MOSCOW) 2018; 83:674-689. [PMID: 30195324 DOI: 10.1134/s0006297918060056] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Calcineurin (CaN) is a serine/threonine phosphatase widely expressed in different cell types and structures including neurons and synapses. The most studied role of CaN is its involvement in the functioning of postsynaptic structures of central synapses. The role of CaN in the presynaptic structures of central and peripheral synapses is less understood, although it has generated a considerable interest and is a subject of a growing number of studies. The regulatory role of CaN in synaptic vesicle endocytosis in the synapse terminals is actively studied. In recent years, new targets of CaN have been identified and its role in the regulation of enzymes and neurotransmitter secretion in peripheral neuromuscular junctions has been revealed. CaN is the only phosphatase that requires calcium and calmodulin for activation. In this review, we present details of CaN molecular structure and give a detailed description of possible mechanisms of CaN activation involving calcium, enzymes, and endogenous and exogenous inhibitors. Known and newly discovered CaN targets at pre- and postsynaptic levels are described. CaN activity in synaptic structures is discussed in terms of functional involvement of this phosphatase in synaptic transmission and neurotransmitter release.
Collapse
Affiliation(s)
- E O Tarasova
- Lomonosov Moscow State University, Faculty of Biology, Moscow, 119991, Russia
| | - A E Gaydukov
- Lomonosov Moscow State University, Faculty of Biology, Moscow, 119991, Russia. .,Pirogov Russian National Research Medical University, Moscow, 117997, Russia
| | - O P Balezina
- Lomonosov Moscow State University, Faculty of Biology, Moscow, 119991, Russia
| |
Collapse
|
19
|
Saraf J, Bhattacharya P, Kalia K, Borah A, Sarmah D, Kaur H, Dave KR, Yavagal DR. A Friend or Foe: Calcineurin across the Gamut of Neurological Disorders. ACS CENTRAL SCIENCE 2018; 4:805-819. [PMID: 30062109 PMCID: PMC6062828 DOI: 10.1021/acscentsci.8b00230] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Indexed: 05/24/2023]
Abstract
The serine/threonine phosphatase calcineurin (CaN) is a unique but confounding calcium/calmodulin-mediated enzyme. CaN has shown to play essential roles from regulating calcium homeostasis to being an intricate part of learning and memory formation. Neurological disorders, despite differing in their etiology, share similar pathological outcomes, such as mitochondrial dysfunction and apoptotic signaling brought about by excitotoxic elements. CaN, being deeply integrated in vital neuronal functions, may be implicated in various neurological disorders. Understanding the enzyme and its physiological niche in the nervous system is vital in uncovering its roles in the spectrum of brain disorders. By reviewing the crosstalk in different neurological pathologies, a possible grasp of CaN's complex signaling may lead to forming better neurotherapy. This Outlook attempts to explore the various neuronal functions of CaN and investigate its pervasive role through the gamut of neurological disorders.
Collapse
Affiliation(s)
- Jackson Saraf
- Department
of Pharmacology and Toxicology, National
Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Pallab Bhattacharya
- Department
of Pharmacology and Toxicology, National
Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Kiran Kalia
- Department
of Pharmacology and Toxicology, National
Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Anupom Borah
- Cellular
and Molecular Neurobiology Laboratory, Department of Life Science
and Bioinformatics, Assam University, Silchar, Assam 788011, India
| | - Deepaneeta Sarmah
- Department
of Pharmacology and Toxicology, National
Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Harpreet Kaur
- Department
of Pharmacology and Toxicology, National
Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Kunjan R Dave
- Department
of Neurology, University of Miami Miller
School of Medicine, Miami, Florida 33136, United States
| | - Dileep R Yavagal
- Department
of Neurology, University of Miami Miller
School of Medicine, Miami, Florida 33136, United States
| |
Collapse
|
20
|
Choi JE, Lee JJ, Kang W, Kim HJ, Cho JH, Han PL, Lee KJ. Proteomic Analysis of Hippocampus in a Mouse Model of Depression Reveals Neuroprotective Function of Ubiquitin C-terminal Hydrolase L1 (UCH-L1) via Stress-induced Cysteine Oxidative Modifications. Mol Cell Proteomics 2018; 17:1803-1823. [PMID: 29959188 PMCID: PMC6126396 DOI: 10.1074/mcp.ra118.000835] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 06/20/2018] [Indexed: 01/08/2023] Open
Abstract
Chronic physical restraint stress increases oxidative stress in the brain, and dysregulation of oxidative stress can be one of the causes of major depressive disorder. To understand the underlying mechanisms, we undertook a systematic proteomic analysis of hippocampus in a chronic restraint stress mouse model of depression. Combining two-dimensional gel electrophoresis (2D-PAGE) for protein separation with nanoUPLC-ESI-q-TOF tandem mass spectrometry, we identified sixty-three protein spots that changed in the hippocampus of mice subjected to chronic restraint stress. We identified and classified the proteins that changed after chronic stress, into three groups respectively functioning in neural plasticity, metabolic processes and protein aggregation. Of these, 5 proteins including ubiquitin C-terminal hydrolase L1 (UCH-L1), dihydropyrimidinase-related protein 2 (DPYL2), haloacid dehalogenase-like hydrolase domain-containing protein 2 (HDHD2), actin-related protein 2/3 complex subunit 5 (ARPC5) and peroxiredoxin-2 (PRDX2), showed pI shifts attributable to post-translational modifications. Further analysis indicated that UCH-L1 underwent differential oxidations of 2 cysteine residues following chronic stress. We investigated whether the oxidized form of UCH-L1 plays a role in stressed hippocampus, by comparing the effects of UCH-L1 and its Cys mutants on hippocampal cell line HT-22 in response to oxidative stress. This study demonstrated that UCH-L1 wild-type and cysteine to aspartic acid mutants, but not its cysteine to serine mutants, afforded neuroprotective effects against oxidative stress; there were no discernible differences between wild-type UCH-L1 and its mutants in the absence of oxidative stress. These findings suggest that cysteine oxidative modifications of UCH-L1 in the hippocampus play key roles in neuroprotection against oxidative stress caused in major depressive disorder.
Collapse
Affiliation(s)
- Jung-Eun Choi
- From the ‡College of Pharmacy and Graduate School of Pharmaceutical Sciences, and
| | - Jae-Jin Lee
- From the ‡College of Pharmacy and Graduate School of Pharmaceutical Sciences, and
| | - Wonmo Kang
- From the ‡College of Pharmacy and Graduate School of Pharmaceutical Sciences, and
| | - Hyun Jung Kim
- From the ‡College of Pharmacy and Graduate School of Pharmaceutical Sciences, and
| | - Jin-Hwan Cho
- From the ‡College of Pharmacy and Graduate School of Pharmaceutical Sciences, and
| | - Pyung-Lim Han
- §Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, Korea 03760
| | - Kong-Joo Lee
- From the ‡College of Pharmacy and Graduate School of Pharmaceutical Sciences, and
| |
Collapse
|
21
|
Balzani E, Falappa M, Balci F, Tucci V. An approach to monitoring home-cage behavior in mice that facilitates data sharing. Nat Protoc 2018; 13:1331-1347. [PMID: 29773907 DOI: 10.1038/nprot.2018.031] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Genetically modified mice are used as models for a variety of human behavioral conditions. However, behavioral phenotyping can be a major bottleneck in mouse genetics because many of the classic protocols are too long and/or are vulnerable to unaccountable sources of variance, leading to inconsistent results between centers. We developed a home-cage approach using a Chora feeder that is controlled by-and sends data to-software. In this approach, mice are tested in the standard cages in which they are held for husbandry, which removes confounding variables such as the stress induced by out-of-cage testing. This system increases the throughput of data gathering from individual animals and facilitates data mining by offering new opportunities for multimodal data comparisons. In this protocol, we use a simple work-for-food testing strategy as an example application, but the approach can be adapted for other experiments looking at, e.g., attention, decision-making or memory. The spontaneous behavioral activity of mice in performing the behavioral task can be monitored 24 h a day for several days, providing an integrated assessment of the circadian profiles of different behaviors. We developed a Python-based open-source analytical platform (Phenopy) that is accessible to scientists with no programming background and can be used to design and control such experiments, as well as to collect and share data. This approach is suitable for large-scale studies involving multiple laboratories.
Collapse
Affiliation(s)
- Edoardo Balzani
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genova, Italy
| | - Matteo Falappa
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genova, Italy.,Dipartimento di Neuroscienze, Riabilitazione, Oftalmologia, Genetica e Scienze Materno-Infantili (DINOGMI), Università degli Studi di Genova, Genova, Italy
| | - Fuat Balci
- Department of Psychology, Koç University, Istanbul, Turkey.,Research Center for Translational Medicine, Koç University, Istanbul, Turkey
| | - Valter Tucci
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genova, Italy
| |
Collapse
|
22
|
Hagihara H, Catts VS, Katayama Y, Shoji H, Takagi T, Huang FL, Nakao A, Mori Y, Huang KP, Ishii S, Graef IA, Nakayama KI, Shannon Weickert C, Miyakawa T. Decreased Brain pH as a Shared Endophenotype of Psychiatric Disorders. Neuropsychopharmacology 2018; 43:459-468. [PMID: 28776581 PMCID: PMC5770757 DOI: 10.1038/npp.2017.167] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 07/14/2017] [Accepted: 08/01/2017] [Indexed: 01/25/2023]
Abstract
Although the brains of patients with schizophrenia and bipolar disorder exhibit decreased brain pH relative to those of healthy controls upon postmortem examination, it remains controversial whether this finding reflects a primary feature of the diseases or is a result of confounding factors such as medication and agonal state. To date, systematic investigation of brain pH has not been undertaken using animal models that can be studied without confounds inherent in human studies. In the present study, we first reevaluated the pH of the postmortem brains of patients with schizophrenia and bipolar disorder by conducting a meta-analysis of existing data sets from 10 studies. We then measured pH, lactate levels, and related metabolite levels in brain homogenates from five neurodevelopmental mouse models of psychiatric disorders, including schizophrenia, bipolar disorder, and autism spectrum disorder. All mice were drug naive with the same agonal state, postmortem interval, and age within each strain. Our meta-analysis revealed that brain pH was significantly lower in patients with schizophrenia and bipolar disorder than in control participants, even when a few potential confounding factors (postmortem interval, age, and history of antipsychotic use) were considered. In animal experiments, we observed significantly lower pH and higher lactate levels in the brains of model mice relative to controls, as well as a significant negative correlation between pH and lactate levels. Our findings suggest that lower pH associated with increased lactate levels is not a mere artifact, but rather implicated in the underlying pathophysiology of schizophrenia and bipolar disorder.
Collapse
Affiliation(s)
- Hideo Hagihara
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan
| | - Vibeke S Catts
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, NSW, Australia,School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - Yuta Katayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Hirotaka Shoji
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan
| | - Tsuyoshi Takagi
- Institute for Developmental Research, Aichi Human Service Center, Kasugai, Japan,RIKEN Tsukuba Institute, Tsukuba, Japan
| | - Freesia L Huang
- Program of Developmental Neurobiology, National Institute of Child Health and Human Development, National Institute of Health, Bethesda, MD, USA
| | - Akito Nakao
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan
| | - Yasuo Mori
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Kuo-Ping Huang
- Program of Developmental Neurobiology, National Institute of Child Health and Human Development, National Institute of Health, Bethesda, MD, USA
| | | | - Isabella A Graef
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Keiichi I Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Cynthia Shannon Weickert
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, NSW, Australia,School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - Tsuyoshi Miyakawa
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan,Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Kutsukake-cho, Toyoake, Aichi 470-1192, Japan, Tel: +81 562 93 9376, Fax: +81 562 92 5382, E-mail:
| |
Collapse
|
23
|
A Functional Role for the Epigenetic Regulator ING1 in Activity-induced Gene Expression in Primary Cortical Neurons. Neuroscience 2017; 369:248-260. [PMID: 29158107 DOI: 10.1016/j.neuroscience.2017.11.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 11/09/2017] [Accepted: 11/11/2017] [Indexed: 12/19/2022]
Abstract
Epigenetic regulation of activity-induced gene expression involves multiple levels of molecular interaction, including histone and DNA modifications, as well as mechanisms of DNA repair. Here we demonstrate that the genome-wide deposition of inhibitor of growth family member 1 (ING1), which is a central epigenetic regulatory protein, is dynamically regulated in response to activity in primary cortical neurons. ING1 knockdown leads to decreased expression of genes related to synaptic plasticity, including the regulatory subunit of calcineurin, Ppp3r1. In addition, ING1 binding at a site upstream of the transcription start site (TSS) of Ppp3r1 depends on yet another group of neuroepigenetic regulatory proteins, the Piwi-like family, which are also involved in DNA repair. These findings provide new insight into a novel mode of activity-induced gene expression, which involves the interaction between different epigenetic regulatory mechanisms traditionally associated with gene repression and DNA repair.
Collapse
|
24
|
Cognitive impairments associated with alterations in synaptic proteins induced by the genetic loss of adenosine A 2A receptors in mice. Neuropharmacology 2017; 126:48-57. [DOI: 10.1016/j.neuropharm.2017.08.027] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 07/28/2017] [Accepted: 08/17/2017] [Indexed: 12/15/2022]
|
25
|
Zafar S, Younas N, Sheikh N, Tahir W, Shafiq M, Schmitz M, Ferrer I, Andréoletti O, Zerr I. Cytoskeleton-Associated Risk Modifiers Involved in Early and Rapid Progression of Sporadic Creutzfeldt-Jakob Disease. Mol Neurobiol 2017; 55:4009-4029. [PMID: 28573459 DOI: 10.1007/s12035-017-0589-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 04/28/2017] [Indexed: 10/19/2022]
Abstract
A high priority in the prion field is to identify pre-symptomatic events and associated profile of molecular changes. In this study, we demonstrate the pre-symptomatic dysregulation of cytoskeleton assembly and its associated cofilin-1 pathway in strain and brain region-specific manners in MM1 and VV2 subtype-specific Creutzfeldt-Jakob disease at clinical and pre-clinical stage. At physiological level, PrPC interaction with cofilin-1 and phosphorylated form of cofilin (p-cofilin(Ser3)) was investigated in primary cultures of mouse cortex neurons (PCNs) of PrPC wild-type and knockout mice (PrP-/-). Short-interfering RNA downregulation of active form of cofilin-1 resulted in the redistribution/downregulation of PrPC, increase of activated form of microglia, accumulation of dense form of F-actin, and upregulation of p-cofilin(Ser3). This upregulated p-cofilin(Ser3) showed redistribution of expression predominantly in the activated form of microglia in PCNs. At pathological level, cofilin-1 expression was significantly altered in cortex and cerebellum in both humans and mice at pre-clinical stage and at early symptomatic clinical stage of the disease. Further, to better understand the possible mechanism of dysregulation of cofilin-1, we also demonstrated alterations in upstream regulators; LIM kinase isoform 1 (LIMK1), slingshot phosphatase isoform 1 (SSH1), RhoA-associated kinase (Rock2), and amyloid precursor protein (APP) in sporadic Creutzfeldt-Jakob disease MM1 mice and in human MM1 and VV2 frontal cortex and cerebellum samples. In conclusion, our findings demonstrated for the first time a key pre-clinical response of cofilin-1 and the associated pathway in prion disease.
Collapse
Affiliation(s)
- Saima Zafar
- Department of Neurology, Clinical Dementia Center, and DZNE, Georg-August University, University Medical Center Goettingen (UMG), Robert-Koch-Str. 40, 37075, Goettingen, Germany.
| | - Neelam Younas
- Department of Neurology, Clinical Dementia Center, and DZNE, Georg-August University, University Medical Center Goettingen (UMG), Robert-Koch-Str. 40, 37075, Goettingen, Germany
| | - Nadeem Sheikh
- Department of Zoology, University of the Punjab, Lahore, Pakistan
| | - Waqas Tahir
- Department of Neurology, Clinical Dementia Center, and DZNE, Georg-August University, University Medical Center Goettingen (UMG), Robert-Koch-Str. 40, 37075, Goettingen, Germany
| | - Mohsin Shafiq
- Department of Neurology, Clinical Dementia Center, and DZNE, Georg-August University, University Medical Center Goettingen (UMG), Robert-Koch-Str. 40, 37075, Goettingen, Germany
| | - Matthias Schmitz
- Department of Neurology, Clinical Dementia Center, and DZNE, Georg-August University, University Medical Center Goettingen (UMG), Robert-Koch-Str. 40, 37075, Goettingen, Germany
| | - Isidre Ferrer
- Institute of Neuropathology, IDIBELL-University Hospital Bellvitge, University of Barcelona, Hospitalet de Llobregat, Spain.,Network Center for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Institute Carlos III, Ministry of Health, Madrid, Spain
| | - Olivier Andréoletti
- Institut National de la Recherche Agronomique/Ecole Nationale Vétérinaire, Toulouse, France
| | - Inga Zerr
- Department of Neurology, Clinical Dementia Center, and DZNE, Georg-August University, University Medical Center Goettingen (UMG), Robert-Koch-Str. 40, 37075, Goettingen, Germany
| |
Collapse
|
26
|
Li YC, Kavalali ET. Synaptic Vesicle-Recycling Machinery Components as Potential Therapeutic Targets. Pharmacol Rev 2017; 69:141-160. [PMID: 28265000 PMCID: PMC5394918 DOI: 10.1124/pr.116.013342] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Presynaptic nerve terminals are highly specialized vesicle-trafficking machines. Neurotransmitter release from these terminals is sustained by constant local recycling of synaptic vesicles independent from the neuronal cell body. This independence places significant constraints on maintenance of synaptic protein complexes and scaffolds. Key events during the synaptic vesicle cycle-such as exocytosis and endocytosis-require formation and disassembly of protein complexes. This extremely dynamic environment poses unique challenges for proteostasis at synaptic terminals. Therefore, it is not surprising that subtle alterations in synaptic vesicle cycle-associated proteins directly or indirectly contribute to pathophysiology seen in several neurologic and psychiatric diseases. In contrast to the increasing number of examples in which presynaptic dysfunction causes neurologic symptoms or cognitive deficits associated with multiple brain disorders, synaptic vesicle-recycling machinery remains an underexplored drug target. In addition, irrespective of the involvement of presynaptic function in the disease process, presynaptic machinery may also prove to be a viable therapeutic target because subtle alterations in the neurotransmitter release may counter disease mechanisms, correct, or compensate for synaptic communication deficits without the need to interfere with postsynaptic receptor signaling. In this article, we will overview critical properties of presynaptic release machinery to help elucidate novel presynaptic avenues for the development of therapeutic strategies against neurologic and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Ying C Li
- Departments of Neuroscience (Y.C.L., E.T.K.) and Physiology (E.T.K.), University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ege T Kavalali
- Departments of Neuroscience (Y.C.L., E.T.K.) and Physiology (E.T.K.), University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
27
|
Mitra S, Sameer Kumar GS, Tiwari V, Lakshmi BJ, Thakur SS, Kumar S. Implication of Genetic Deletion of Wdr13 in Mice: Mild Anxiety, Better Performance in Spatial Memory Task, with Upregulation of Multiple Synaptic Proteins. Front Mol Neurosci 2016; 9:73. [PMID: 27625594 PMCID: PMC5003927 DOI: 10.3389/fnmol.2016.00073] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Accepted: 08/08/2016] [Indexed: 11/29/2022] Open
Abstract
WDR13 expresses from the X chromosome and has a highly conserved coding sequence. There have been multiple associations of WDR13 with memory. However, its detailed function in context of brain and behavior remains unknown. We characterized the behavioral phenotype of 2 month old male mice lacking the homolog of WDR13 gene (Wdr13−/0). Taking cue from analysis of its expression in the brain, we chose hippocampus for molecular studies to delineate its function. Wdr13−/0 mice spent less time in the central area of the open field test (OFT) and with the novel object in novel object recognition test (NOR) as compared to the wild-type. However, these mice didn't show any significant changes in total time spent in arms or in frequency of arm entries in elevated plus maze (EPM). In the absence of Wdr13, there was a significant upregulation of synaptic proteins, viz., SYN1, RAB3A, CAMK2A etc. accompanied with increased spine density of hippocampal CA1 neurons and better spatial memory in mice as measured by increased time spent in the target quadrant of Morris water maze (MWM) during probe test. Parallel study from our lab has established c-JUN, ER α/β, and HDAC 1,3,7 as interacting partners of WDR13. WDR13 represses transcription from AP1 (c-JUN responsive) and Estrogen Receptor Element (ERE) promoters. We hypothesized that absence of Wdr13 would result in de-regulated expression of a number of genes including multiple synaptic genes leading to the observed phenotype. Knocking down Wdr13 in Neuro2a cell lines led to increased transcripts of Camk2a and Nrxn2 consistent with in-vivo results. Summarily, our data provides functional evidence for the role of Wdr13 in brain.
Collapse
Affiliation(s)
- Shiladitya Mitra
- Council of Scientific and Industrial Research - Centre for Cellular and Molecular Biology Hyderabad, India
| | - Ghantasala S Sameer Kumar
- Council of Scientific and Industrial Research - Centre for Cellular and Molecular Biology Hyderabad, India
| | - Vivek Tiwari
- Council of Scientific and Industrial Research - Centre for Cellular and Molecular Biology Hyderabad, India
| | - B Jyothi Lakshmi
- Council of Scientific and Industrial Research - Centre for Cellular and Molecular Biology Hyderabad, India
| | - Suman S Thakur
- Council of Scientific and Industrial Research - Centre for Cellular and Molecular Biology Hyderabad, India
| | - Satish Kumar
- Council of Scientific and Industrial Research - Centre for Cellular and Molecular Biology Hyderabad, India
| |
Collapse
|
28
|
Endosome-mediated endocytic mechanism replenishes the majority of synaptic vesicles at mature CNS synapses in an activity-dependent manner. Sci Rep 2016; 6:31807. [PMID: 27534442 PMCID: PMC4989163 DOI: 10.1038/srep31807] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 07/26/2016] [Indexed: 11/30/2022] Open
Abstract
Whether synaptic vesicles (SVs) are recovered via endosome-mediated pathways is a matter of debate; however, recent evidence suggests that clathrin-independent bulk endocytosis (CIE) via endosomes is functional and preferentially replenishes SV pools during strong stimulation. Here, using brefeldin-A (BFA) to block CIE, we found that CIE retrieved a minority of SVs at developing CNS synapses during strong stimulation, but its contribution increased up to 61% at mature CNS synapses. Contrary to previous views, BFA not only blocked SV formation from the endosome but also blocked the endosome formation at the plasma membrane. Adaptor protein 1 and 3 (AP-1/3) have key roles in SV reformation from endosomes during CIE, and AP-1 also affects bulk endosome formation from the plasma membrane. Finally, temporary blocking of chronic or acute neuronal activity with tetrodotoxin in mature neurons redirected most SV retrieval to endosome-independent pathways. These results show that during high neuronal activity, CIE becomes the major endocytic pathway at mature CNS synapses. Moreover, mature neurons use clathrin-mediated endocytosis and the CIE pathway to different extents depending on their previous activity; this may result in activity-dependent alterations of the SV composition which ultimately influence transmitter release and contribute to synaptic plasticity.
Collapse
|
29
|
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: 4.0] [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]
|
30
|
ADAR-mediated RNA editing suppresses sleep by acting as a brake on glutamatergic synaptic plasticity. Nat Commun 2016; 7:10512. [PMID: 26813350 PMCID: PMC4737855 DOI: 10.1038/ncomms10512] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 12/22/2015] [Indexed: 01/18/2023] Open
Abstract
It has been postulated that synaptic potentiation during waking is offset by a homoeostatic reduction in net synaptic strength during sleep. However, molecular mechanisms to support such a process are lacking. Here we demonstrate that deficiencies in the RNA-editing gene Adar increase sleep due to synaptic dysfunction in glutamatergic neurons in Drosophila. Specifically, the vesicular glutamate transporter is upregulated, leading to over-activation of NMDA receptors, and the reserve pool of glutamatergic synaptic vesicles is selectively expanded in Adar mutants. Collectively these changes lead to sustained neurotransmitter release under conditions that would otherwise result in synaptic depression. We propose that a shift in the balance from synaptic depression towards synaptic potentiation in sleep-promoting neurons underlies the increased sleep pressure of Adar-deficient animals. Our findings provide a plausible molecular mechanism linking sleep and synaptic plasticity. Sleep is postulated to offset buildup in net synaptic strength that occurs during waking experience. Here, the authors identify a role for the RNA editing gene Adar in regulating glutamatergic synaptic plasticity and show that disruption in Adar expression impairs normal waking in flies.
Collapse
|
31
|
Marland JRK, Smillie KJ, Cousin MA. Synaptic Vesicle Recycling Is Unaffected in the Ts65Dn Mouse Model of Down Syndrome. PLoS One 2016; 11:e0147974. [PMID: 26808141 PMCID: PMC4726538 DOI: 10.1371/journal.pone.0147974] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 01/11/2016] [Indexed: 01/08/2023] Open
Abstract
Down syndrome (DS) is the most common genetic cause of intellectual disability, and arises from trisomy of human chromosome 21. Accumulating evidence from studies of both DS patient tissue and mouse models has suggested that synaptic dysfunction is a key factor in the disorder. The presence of several genes within the DS trisomy that are either directly or indirectly linked to synaptic vesicle (SV) endocytosis suggested that presynaptic dysfunction could underlie some of these synaptic defects. Therefore we determined whether SV recycling was altered in neurons from the Ts65Dn mouse, the best characterised model of DS to date. We found that SV exocytosis, the size of the SV recycling pool, clathrin-mediated endocytosis, activity-dependent bulk endocytosis and SV generation from bulk endosomes were all unaffected by the presence of the Ts65Dn trisomy. These results were obtained using battery of complementary assays employing genetically-encoded fluorescent reporters of SV cargo trafficking, and fluorescent and morphological assays of fluid-phase uptake in primary neuronal culture. The absence of presynaptic dysfunction in central nerve terminals of the Ts65Dn mouse suggests that future research should focus on the established alterations in excitatory / inhibitory balance as a potential route for future pharmacotherapy.
Collapse
Affiliation(s)
- Jamie R. K. Marland
- Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, 15 George Square, Edinburgh, EH8 9XD, United Kingdom
| | - Karen J. Smillie
- Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, 15 George Square, Edinburgh, EH8 9XD, United Kingdom
| | - Michael A. Cousin
- Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, 15 George Square, Edinburgh, EH8 9XD, United Kingdom
- * E-mail:
| |
Collapse
|
32
|
Cottrell JR, Li B, Kyung JW, Ashford CJ, Mann JJ, Horvath TL, Ryan TA, Kim SH, Gerber DJ. Calcineurin Aγ is a Functional Phosphatase That Modulates Synaptic Vesicle Endocytosis. J Biol Chem 2015; 291:1948-1956. [PMID: 26627835 DOI: 10.1074/jbc.m115.705319] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Indexed: 12/11/2022] Open
Abstract
Variation in PPP3CC, the gene that encodes the γ isoform of the calcineurin catalytic subunit, has been reported to be associated with schizophrenia. Because of its low expression level in most tissues, there has been little research devoted to the specific function of the calcineurin Aγ (CNAγ) versus the calcineurin Aα (CNAα) and calcineurin Aβ (CNAβ) catalytic isoforms. Consequently, we have a limited understanding of the role of altered CNAγ function in psychiatric disease. In this study, we demonstrate that CNAγ is present in the rodent and human brain and dephosphorylates a presynaptic substrate of calcineurin. Through a combination of immunocytochemistry and immuno-EM, we further show that CNAγ is localized to presynaptic terminals in hippocampal neurons. Critically, we demonstrate that RNAi-mediated knockdown of CNAγ leads to a disruption of synaptic vesicle cycling in cultured rat hippocampal neurons. These data indicate that CNAγ regulates a critical aspect of synaptic vesicle cycling and suggest that variation in PPP3CC may contribute to psychiatric disease by altering presynaptic function.
Collapse
Affiliation(s)
| | - Bing Li
- From the Galenea Corporation, Wakefield, MA 01880
| | - Jae Won Kyung
- the Department of Physiology, Neurodegeneration Control Research Center, School of Medicine, Kyung Hee University, Seoul, 02447, South Korea
| | | | - James J Mann
- From the Galenea Corporation, Wakefield, MA 01880
| | - Tamas L Horvath
- the Program in Integrative Cell Signaling and Neurobiology of Metabolism, Section of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut 06511, and
| | - Timothy A Ryan
- the Department of Biochemistry, Weill Cornell Medical College, New York, New York 10021
| | - Sung Hyun Kim
- the Department of Physiology, Neurodegeneration Control Research Center, School of Medicine, Kyung Hee University, Seoul, 02447, South Korea.
| | | |
Collapse
|
33
|
Crabtree GW, Gogos JA. Synaptic plasticity, neural circuits, and the emerging role of altered short-term information processing in schizophrenia. Front Synaptic Neurosci 2014; 6:28. [PMID: 25505409 PMCID: PMC4243504 DOI: 10.3389/fnsyn.2014.00028] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 10/22/2014] [Indexed: 01/01/2023] Open
Abstract
Synaptic plasticity alters the strength of information flow between presynaptic and postsynaptic neurons and thus modifies the likelihood that action potentials in a presynaptic neuron will lead to an action potential in a postsynaptic neuron. As such, synaptic plasticity and pathological changes in synaptic plasticity impact the synaptic computation which controls the information flow through the neural microcircuits responsible for the complex information processing necessary to drive adaptive behaviors. As current theories of neuropsychiatric disease suggest that distinct dysfunctions in neural circuit performance may critically underlie the unique symptoms of these diseases, pathological alterations in synaptic plasticity mechanisms may be fundamental to the disease process. Here we consider mechanisms of both short-term and long-term plasticity of synaptic transmission and their possible roles in information processing by neural microcircuits in both health and disease. As paradigms of neuropsychiatric diseases with strongly implicated risk genes, we discuss the findings in schizophrenia and autism and consider the alterations in synaptic plasticity and network function observed in both human studies and genetic mouse models of these diseases. Together these studies have begun to point toward a likely dominant role of short-term synaptic plasticity alterations in schizophrenia while dysfunction in autism spectrum disorders (ASDs) may be due to a combination of both short-term and long-term synaptic plasticity alterations.
Collapse
Affiliation(s)
- Gregg W. Crabtree
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia UniversityNew York, NY, USA
| | - Joseph A. Gogos
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia UniversityNew York, NY, USA
- Department of Neuroscience, College of Physicians and Surgeons, Columbia UniversityNew York, NY, USA
| |
Collapse
|
34
|
Evstratova A, Chamberland S, Faundez V, Tóth K. Vesicles derived via AP-3-dependent recycling contribute to asynchronous release and influence information transfer. Nat Commun 2014; 5:5530. [PMID: 25410111 PMCID: PMC4239664 DOI: 10.1038/ncomms6530] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 10/10/2014] [Indexed: 12/21/2022] Open
Abstract
Action potentials trigger synchronous and asynchronous neurotransmitter release. Temporal properties of both types of release could be altered in an activity-dependent manner. While the effects of activity-dependent changes in synchronous release on postsynaptic signal integration have been studied, the contribution of asynchronous release to information transfer during natural stimulus patterns is unknown. Here we find that during trains of stimulations, asynchronous release contributes to the precision of action potential firing. Our data show that this form of release is selectively diminished in AP-3b2 KO animals, which lack functional neuronal AP-3, an adaptor protein regulating vesicle formation from endosomes generated during bulk endocytosis. We find that in the absence of neuronal AP-3, asynchronous release is attenuated and the activity-dependent increase in the precision of action potential timing is compromised. Lack of asynchronous release decreases the capacity of synaptic information transfer and renders synaptic communication less reliable in response to natural stimulus patterns.
Collapse
Affiliation(s)
- Alesya Evstratova
- Department of Psychiatry and Neuroscience, Quebec Mental Health Institute, Université Laval, Quebec City, Quebec, Canada G1J 2G3
| | - Simon Chamberland
- Department of Psychiatry and Neuroscience, Quebec Mental Health Institute, Université Laval, Quebec City, Quebec, Canada G1J 2G3
| | - Victor Faundez
- Department of Cell Biology, Emory University, Atlanta, Georgia 30322, USA
| | - Katalin Tóth
- Department of Psychiatry and Neuroscience, Quebec Mental Health Institute, Université Laval, Quebec City, Quebec, Canada G1J 2G3
| |
Collapse
|
35
|
Wu Y, Ly PTT, Song W. Aberrant expression of RCAN1 in Alzheimer's pathogenesis: a new molecular mechanism and a novel drug target. Mol Neurobiol 2014; 50:1085-97. [PMID: 24752590 DOI: 10.1007/s12035-014-8704-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 03/31/2014] [Indexed: 01/08/2023]
Abstract
AD, a devastating neurodegenerative disorder, is the most common cause of dementia in the elderly. Patients with AD are characterized by three hallmarks of neuropathology including neuritic plaque deposition, neurofibrillary tangle formation, and neuronal loss. Growing evidences indicate that dysregulation of regulator of calcineurin 1 (RCAN1) plays an important role in the pathogenesis of AD. Aberrant RCAN1 expression facilitates neuronal apoptosis and Tau hyperphosphorylation, leading to neuronal loss and neurofibrillary tangle formation. This review aims to describe the recent advances of the regulation of RCAN1 expression and its physiological functions. Moreover, the AD risk factors-induced RCAN1 dysregulation and its role in promoting neuronal loss, synaptic impairments and neurofibrillary tangle formation are summarized. Furthermore, we provide an outlook into the effects of RCAN1 dysregulation on APP processing, Aβ generation and neuritic plaque formation, and the possible underlying mechanisms, as well as the potential of targeting RCAN1 as a new therapeutic approach.
Collapse
Affiliation(s)
- Yili Wu
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, Graduate Program in Neuroscience, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, Canada, V6T 1Z3
| | | | | |
Collapse
|
36
|
Tokuoka H, Hatanaka T, Metzger D, Ichinose H. Nurr1 expression is regulated by voltage-dependent calcium channels and calcineurin in cultured hippocampal neurons. Neurosci Lett 2013; 559:50-5. [PMID: 24291696 DOI: 10.1016/j.neulet.2013.11.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 11/19/2013] [Indexed: 12/01/2022]
Abstract
Nurr1 is an orphan nuclear transcription factor expressed in the brain. While Nurr1 is assumed to be an immediate early gene, it is not fully understood how Nurr1 expression is regulated in an activity-dependent manner in the central nervous system. Here, we investigated the molecular mechanisms underlying the regulation of Nurr1 expression in cultured hippocampal and cortical neurons. We found that upregulation of neural activity by high KCl and bicuculline enhances Nurr1 levels, while blockade of its activity by tetrodotoxin reduces Nurr1 levels. The induction of Nurr1 expression was mediated by voltage-dependent calcium channels (VDCCs), as shown by cadmium and VDCC-specific inhibitors. Furthermore, calcineurin, but not calcium/calmodulin-dependent protein kinase (CaMK) was critical for the induction. Thus, Nurr1 expression is regulated by VDCC and calcineurin in a cell-autonomous, neural activity-dependent manner.
Collapse
Affiliation(s)
- Hirofumi Tokuoka
- Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Yokohama, Japan
| | - Takayuki Hatanaka
- Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Yokohama, Japan
| | - Daniel Metzger
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch F-67400, France; CNRS UMR7104, Illkirch, France; INSERM U964, Illkirch, France; Université de Strasbourg, Strasbourg, France
| | - Hiroshi Ichinose
- Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Yokohama, Japan.
| |
Collapse
|