1
|
Quach TT, Duchemin AM. Intelligence, brain structure, dendrites, and genes: Genetic, epigenetic and the underlying of the quadruple helix complexity. Neurosci Biobehav Rev 2025; 175:106212. [PMID: 40389043 DOI: 10.1016/j.neubiorev.2025.106212] [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: 01/20/2025] [Revised: 05/01/2025] [Accepted: 05/12/2025] [Indexed: 05/21/2025]
Abstract
Intelligence can be referred to as the mental ability to learn, comprehend abstract concepts, and solve complex problems. Twin and adoption studies have provided insights into the influence of the familial environment and highlighted the importance of heritability in the development of cognition. Detecting the relative contribution of brain areas, neuronal structures, and connectomes has brought some understanding on how various brain areas, white/gray matter structures and neuronal connectivity process information and contribute to intelligence. Using histological, anatomical, electrophysiological, neuropsychological, neuro-imaging and molecular biology methods, several key concepts have emerged: 1) the parietofrontal-hippocampal integrations probably constitute a substrate for smart behavior, 2) neuronal activity results in structural plasticity of dendritic branches responsible for information transfer, critical for learning and memory, 3) intelligent people process information efficiently, 4) the environment triggers mnemonic epigenomic programs (via dynamic regulation of chromatin accessibility, DNA methylation, loop interruption/formation and histone modification) conferring cognitive phenotypes throughout life, and 5) single/double DNA breaks are prominent in human brain disorders associated with cognitive impairment including Alzheimer's disease and schizophrenia. Along with these observations, molecular/cellular/biological studies have identified sets of specific genes associated with higher scores on intelligence tests. Interestingly, many of these genes are associated with dendritogenesis. Because dendrite structure/function is involved in cognition, the control of dendrite genesis/maintenance may be critical for understanding the landscape of general/specific cognitive ability and new pathways for therapeutic approaches.
Collapse
Affiliation(s)
- Tam T Quach
- Department of Neuroscience. The Ohio State University, Columbus, OH 43210, USA.
| | - Anne-Marie Duchemin
- Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
2
|
Xie Z, Song W, Meng Z, Ma A, Zhu J, Liang Y, Lin H, Lei C, Tan M. The interaction between KATNA1 and CRMP3 modulates microtubule dynamics and neurite outgrowth. Biochem Biophys Res Commun 2025; 752:151426. [PMID: 39938451 DOI: 10.1016/j.bbrc.2025.151426] [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: 10/13/2024] [Revised: 12/24/2024] [Accepted: 01/29/2025] [Indexed: 02/14/2025]
Abstract
The polymerization and severing of microtubules are fundamental to the growth and branching of neurites in hippocampal neurons. The catalytic ATPase-containing A-subunit of katanin p60 (p60, KATNA1) promotes growth and development of hippocampal neurites by severing microtubules, while collapsing response mediator protein 3 (CRMP3) assembles microtubules to regulate neurite outgrowth. However, whether microtubule severing and assembling proteins would work together to regulate neurite outgrowth, especially for KATNA1 and CRMP3 remains to be elucidated. In this study, we revealed the interaction between KATNA1 and CRMP3 through GST-pulldown and co-immunoprecipitation assays and identified the binding domains between KATNA1 and CRMP3 as the MIT of KATNA1 (residues 1-77) and the D region of CRMP3 (residues 64-413). Furthermore, we demonstrated that CRMP3 enhances the microtubule-severing efficiency of KATNA1. In cultured hippocampal neurons, overexpression of KATNA1 and CRMP3 increased neurite length and branch number, and co-expression of both proteins further enhanced the promoting effect. Moreover, genetic knockout of KATNA1 or/and CRMP3 significantly inhibited neurite outgrowth. Overall, our data suggest that the CRMP3 interaction enhances the severing activity of KATNA1, thereby promoting hippocampal neurite outgrowth.
Collapse
Affiliation(s)
- Zhiyao Xie
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, 999078, Macau
| | - Wei Song
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Zhichao Meng
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Ao Ma
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Jiehao Zhu
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Yaozhong Liang
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Hongsheng Lin
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Changbin Lei
- Department of Orthopedics, Affiliated Hospital of Xiangnan University (Clinical College), Chenzhou, Hunan, 423000, China.
| | - Minghui Tan
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China.
| |
Collapse
|
3
|
Liu J, Wang Q, Chen J. Case report: A de novo variant of CRMP1 in an individual with a neurodevelopmental disorder. Front Neurosci 2024; 18:1490731. [PMID: 39758889 PMCID: PMC11695366 DOI: 10.3389/fnins.2024.1490731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 11/25/2024] [Indexed: 01/07/2025] Open
Abstract
Background CRMP1 is a key protein involved in brain development. Methods We performed genetic testing through whole-exome sequencing (WES) in an individual with a neurodevelopmental disorder. Results We identified a de novo heterozygous CRMP1 NM_001014809.3:c.1755del (p.Lys586fs) variant in the affected individual. This mutation was submitted to ClinVar (SCV005196589). Conclusion Currently, the CRMP1 gene has no clear disease phenotype association in the Online Mendelian Inheritance in Man (OMIM) database. Our report may provide evidence for an association between the CRMP1 gene and neurodevelopmental disorders (NDDs).
Collapse
Affiliation(s)
- Juan Liu
- Department of Pediatrics, Mianyang Central Hospital, Mianyang, Sichuan, China
- Chengdu Medical College, Chengdu, Sichuan, China
| | - Qi Wang
- Department of Pediatrics, Mianyang Central Hospital, Mianyang, Sichuan, China
| | - Jia Chen
- Department of Pediatrics, Mianyang Central Hospital, Mianyang, Sichuan, China
| |
Collapse
|
4
|
Zheng X, Lin Y, Huang L, Lin X. Effect of lidocaine on cognitively impaired rats: Anti-inflammatory and antioxidant mechanisms in combination with CRMP2 antiphosphorylation. Immun Inflamm Dis 2023; 11:e1040. [PMID: 37904712 PMCID: PMC10566448 DOI: 10.1002/iid3.1040] [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: 04/29/2023] [Revised: 09/11/2023] [Accepted: 09/20/2023] [Indexed: 11/01/2023] Open
Abstract
OBJECTIVE Studies have shown that lidocaine has antioxidative stress, anti-inflammatory, and nerve-protective effects. The current study investigated the effects of lidocaine on cognitive function in rats with cognitive dysfunction. METHODS A total of 48 rats were randomly assigned to four groups of 12 rats each: control group; L (lidocaine) + D (d-galactose) group, d-galactose group (D group); and D + L group. We assessed cognitive function using a Morris water maze (MWM) and pathologic changes of hippocampal sections. An enzyme-linked immunosorbent assay (ELIZA) was used to detect serum malondialdehyde (MDA) and superoxide dismutase (SOD) levels in rats, and protein immunoblotting (western blot) was used to detect brain tissue proteins (collapsing response mediator protein-2 [CRMP2], phosphorylated-collapsing response mediator protein-2 [P-CRMP2], and β-amyloid protein [Aβ]). RESULTS The MWM showed that the d-gal group (284.09 ± 20.46, 5.20 ± 0.793) performed worse than the L + D (265.37 ± 22.34, 4.170 ± 0.577; p = .000) and D + L groups (254.72 ± 27.87, 3.750; p = .000) in escape latency and number of platform crossings, respectively. The L + D group (44.94 ± 2.92 pg/mL, 6.22 ± 0.50 pg/mL, and 460.02 ± 8.26 nmol/mL) and D + L group (46.88 ± 2.63 pg/mL, 5.90 ± 0.38 pg/mL, and 465.6 ± 16.07 nmol/mL) had significantly lower serum inflammatory levels of interleukin-6, tumor necrosis factor-α, and MDA than the d-gal group (57.79 ± 3.96 pg/mL, 11.25 ± 1.70 pg/mL, and 564.9 ± 15.90 nmol/mL), respectively. The L + D group (3.17 ± 0.41 μg/mL) and D + L group (3.08 ± 0.09 μg/mL) had significantly higher serum inflammatory levels of SOD than the d-gal group (2.20 ± 0.13 μg/mL) (all p = .000). The levels of CRMP2, P-CRMP2, and Aβ in the brain tissue homogenates of the L + D group (0.87 ± 0.04, 0.57 ± 0.0, and 0.16 ± 0.02) and the D + L group (0.82 ± 0.05, 0.58 ± 0.09, and 0.15 ± 0.02) were significantly different than the d-gal group (0.67 ± 0.03, 0.96 ± 0.040, and 0.29 ± 0.05). CONCLUSIONS Lidocaine was shown to reduce cognitive impairment in rats with cognitive dysfunction through anti-inflammatory and antioxidative stress mechanisms in combination with CRMP2 antiphosphorylation.
Collapse
Affiliation(s)
- Xiaohong Zheng
- Department of Anesthesiology, First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Anesthesiology, National Regional Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Yuerong Lin
- Department of Anesthesiology, First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Anesthesiology, National Regional Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Linshen Huang
- Department of Anesthesiology, First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Anesthesiology, National Regional Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Xianzhong Lin
- Department of Anesthesiology, First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Anesthesiology, National Regional Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| |
Collapse
|
5
|
Lukomska A, Theune WC, Xing J, Frost MP, Damania A, Gupta M, Trakhtenberg EF. Experimental gene expression of developmentally downregulated Crmp1, Crmp4, and Crmp5 promotes axon regeneration and retinal ganglion cell survival after optic nerve injury. Brain Res 2023; 1809:148368. [PMID: 37059258 PMCID: PMC10227878 DOI: 10.1016/j.brainres.2023.148368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/29/2023] [Accepted: 04/10/2023] [Indexed: 04/16/2023]
Abstract
Collapsin response mediator proteins (Crmps) play roles in neuronal development and axon growth. However, neuronal-specific roles of Crmp1, Crmp4, and Crmp5 in regeneration of injured central nervous system (CNS) axons in vivo are unclear. Here, we analyzed developmental and subtype-specific expression of Crmp genes in retinal ganglion cells (RGCs), tested whether overexpressing Crmp1, Crmp4, or Crmp5 in RGCs through localized intralocular AAV2 delivery promotes axon regeneration after optic nerve injury in vivo, and characterized developmental co-regulation of gene-concept networks associated with Crmps. We found that all Crmp genes are developmentally downregulated in RGCs during maturation. However, while Crmp1, Crmp2, and Crmp4 were expressed to a varying degree in most RGC subtypes, Crmp3 and Crmp5 were expressed only in a small subset of RGC subtypes. We then found that after optic nerve injury, Crmp1, Crmp4, and Crmp5 promote RGC axon regeneration to varying extents, with Crmp4 promoting the most axon regeneration and also localizing to axons. We also found that Crmp1 and Crmp4, but not Crmp5, promote RGC survival. Finally, we found that Crmp1, Crmp2, Crmp4, and Crmp5's ability to promote axon regeneration is associated with neurodevelopmental mechanisms, which control RGC's intrinsic axon growth capacity.
Collapse
Affiliation(s)
- Agnieszka Lukomska
- Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| | - William C Theune
- Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| | - Jian Xing
- Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| | - Matthew P Frost
- Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| | - Ashiti Damania
- Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| | - Mahit Gupta
- Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| | - Ephraim F Trakhtenberg
- Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA.
| |
Collapse
|
6
|
Qiu L, Sheng P, Wang X. Identification of Metabolic Syndrome-Related miRNA-mRNA Regulatory Networks and Key Genes Based on Bioinformatics Analysis. Biochem Genet 2023; 61:428-447. [PMID: 35877019 DOI: 10.1007/s10528-022-10257-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 05/18/2022] [Indexed: 01/24/2023]
Abstract
Metabolic syndrome, which affects approximately one-quarter of the world's population, is a combination of multiple traits and is associated with high all-cause mortality, increased cancer risk, and other hazards. It has been shown that the epigenetic functions of miRNAs are closely related to metabolic syndrome, but epigenetic studies have not yet fully elucidated the regulatory network and key genes associated with metabolic syndrome. To perform data analysis and screening of potential differentially expressed target miRNAs, mRNAs and genes based on a bioinformatics approach using a metabolic syndrome mRNA and miRNA gene microarray, leading to further analysis and identification of metabolic syndrome-related miRNA-mRNA regulatory networks and key genes. The miRNA gene set (GSE98896) and mRNA gene set (GSE98895) of peripheral blood samples from patients with metabolic syndrome from the GEO database were screened, and set|logFC|> 1 and adjusted P < 0.05 were used to identify the differentially expressed miRNAs and mRNAs. Differentially expressed miRNA transcription factors were predicted using FunRich software and subjected to GO and KEGG enrichment analysis. Next, biological process enrichment analysis of differentially expressed mRNAs was performed with Metascape. Differentially expressed miRNAs and mRNAs were identified and visualized as miRNA-mRNA regulatory networks based on the complementary pairing principle. Data analysis of genome-wide metabolic syndrome-related mRNAs was performed using the gene set enrichment analysis (GSEA) database. Finally, further WGCNA of the set of genes most closely associated with metabolic syndrome was performed to validate the findings. A total of 217 differentially expressed mRNAs and 158 differentially expressed miRNAs were identified by screening the metabolic syndrome miRNA and mRNA gene sets, and these molecules mainly included transcription factors, such as SP1, SP4, and EGR1, that function in the IL-17 signalling pathway; cytokine-cytokine receptor interaction; proteoglycan syndecan-mediated signalling events; and the glypican pathway, which is involved in the inflammatory response and glucose and lipid metabolism. miR-34C-5P, which was identified by constructing a miRNA-mRNA regulatory network, could regulate DPYSL4 expression to influence insulin β-cells, the inflammatory response and glucose oxidative catabolism. Based on GSEA, metabolic syndrome is known to be closely related to oxidative phosphorylation, DNA repair, neuronal damage, and glycolysis. Finally, RStudio and DAVID were used to perform WGCNA of the gene sets most closely associated with metabolic syndrome, and the results further validated the conclusions. Metabolic syndrome is a common metabolic disease worldwide, and its mechanism of action is closely related to the inflammatory response, glycolipid metabolism, and impaired mitochondrial function. miR-34C-5P can regulate DPYSL4 expression and can be a potential research target. In addition, UQCRQ and NDUFA8 are core genes of oxidative phosphorylation and have also been identified as potential targets for the future treatment of metabolic syndrome.
Collapse
Affiliation(s)
- Lingyan Qiu
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210029, China.,The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Pei Sheng
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210029, China.,The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Xu Wang
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210029, China. .,The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China.
| |
Collapse
|
7
|
Desprez F, Ung DC, Vourc’h P, Jeanne M, Laumonnier F. Contribution of the dihydropyrimidinase-like proteins family in synaptic physiology and in neurodevelopmental disorders. Front Neurosci 2023; 17:1154446. [PMID: 37144098 PMCID: PMC10153444 DOI: 10.3389/fnins.2023.1154446] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 03/15/2023] [Indexed: 05/06/2023] Open
Abstract
The dihydropyrimidinase-like (DPYSL) proteins, also designated as the collapsin response mediators (CRMP) proteins, constitute a family of five cytosolic phosphoproteins abundantly expressed in the developing nervous system but down-regulated in the adult mouse brain. The DPYSL proteins were initially identified as effectors of semaphorin 3A (Sema3A) signaling and consequently involved in regulation of growth cone collapse in young developing neurons. To date, it has been established that DPYSL proteins mediate signals for numerous intracellular/extracellular pathways and play major roles in variety of cellular process including cell migration, neurite extension, axonal guidance, dendritic spine development and synaptic plasticity through their phosphorylation status. The roles of DPYSL proteins at early stages of brain development have been described in the past years, particularly for DPYSL2 and DPYSL5 proteins. The recent characterization of pathogenic genetic variants in DPYSL2 and in DPYSL5 human genes associated with intellectual disability and brain malformations, such as agenesis of the corpus callosum and cerebellar dysplasia, highlighted the pivotal role of these actors in the fundamental processes of brain formation and organization. In this review, we sought to establish a detailed update on the knowledge regarding the functions of DPYSL genes and proteins in brain and to highlight their involvement in synaptic processing in later stages of neurodevelopment, as well as their particular contribution in human neurodevelopmental disorders (NDDs), such as autism spectrum disorders (ASD) and intellectual disability (ID).
Collapse
Affiliation(s)
| | - Dévina C. Ung
- UMR1253, iBrain, Inserm, University of Tours, Tours, France
| | - Patrick Vourc’h
- UMR1253, iBrain, Inserm, University of Tours, Tours, France
- Service de Génétique, Centre Hospitalier Régional Universitaire, Tours, France
- Laboratoire de Biochimie et de Biologie Moléculaire, Centre Hospitalier Régional Universitaire, Tours, France
| | - Médéric Jeanne
- UMR1253, iBrain, Inserm, University of Tours, Tours, France
- Service de Génétique, Centre Hospitalier Régional Universitaire, Tours, France
| | - Frédéric Laumonnier
- UMR1253, iBrain, Inserm, University of Tours, Tours, France
- Service de Génétique, Centre Hospitalier Régional Universitaire, Tours, France
- *Correspondence: Frédéric Laumonnier,
| |
Collapse
|
8
|
Quach TT, Stratton HJ, Khanna R, Mackey-Alfonso S, Deems N, Honnorat J, Meyer K, Duchemin AM. Neurodegenerative Diseases: From Dysproteostasis, Altered Calcium Signalosome to Selective Neuronal Vulnerability to AAV-Mediated Gene Therapy. Int J Mol Sci 2022; 23:ijms232214188. [PMID: 36430666 PMCID: PMC9694178 DOI: 10.3390/ijms232214188] [Citation(s) in RCA: 3] [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: 08/29/2022] [Revised: 11/01/2022] [Accepted: 11/04/2022] [Indexed: 11/18/2022] Open
Abstract
Despite intense research into the multifaceted etiology of neurodegenerative diseases (ND), they remain incurable. Here we provide a brief overview of several major ND and explore novel therapeutic approaches. Although the cause (s) of ND are not fully understood, the accumulation of misfolded/aggregated proteins in the brain is a common pathological feature. This aggregation may initiate disruption of Ca++ signaling, which is an early pathological event leading to altered dendritic structure, neuronal dysfunction, and cell death. Presently, ND gene therapies remain unidimensional, elusive, and limited to modifying one pathological feature while ignoring others. Considering the complexity of signaling cascades in ND, we discuss emerging therapeutic concepts and suggest that deciphering the molecular mechanisms involved in dendritic pathology may broaden the phenotypic spectrum of ND treatment. An innovative multiplexed gene transfer strategy that employs silencing and/or over-expressing multiple effectors could preserve vulnerable neurons before they are lost. Such therapeutic approaches may extend brain health span and ameliorate burdensome chronic disease states.
Collapse
Affiliation(s)
- Tam T. Quach
- Institute for Behavioral Medicine Research, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
- INSERM U1217/CNRS UMR5310, Université de Lyon, Université Claude Bernard Lyon 1, 69677 Lyon, France
| | | | - Rajesh Khanna
- Department of Molecular Pathobiology, New York University, New York, NY 10010, USA
| | - Sabrina Mackey-Alfonso
- Institute for Behavioral Medicine Research, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Nicolas Deems
- Institute for Behavioral Medicine Research, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Jérome Honnorat
- INSERM U1217/CNRS UMR5310, Université de Lyon, Université Claude Bernard Lyon 1, 69677 Lyon, France
- French Reference Center on Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon, 69677 Lyon, France
- SynatAc Team, Institut NeuroMyoGène, 69677 Lyon, France
| | - Kathrin Meyer
- The Research Institute of Nationwide Children Hospital, Columbus, OH 43205, USA
- Department of Pediatric, The Ohio State University, Columbus, OH 43210, USA
| | - Anne-Marie Duchemin
- Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, OH 43210, USA
- Correspondence: ; Tel.: +1-614-293-5517; Fax: +1-614-293-7599
| |
Collapse
|
9
|
Zhu Y, Zhou Z, Huang T, Zhang Z, Li W, Ling Z, Jiang T, Yang J, Yang S, Xiao Y, Charlier C, Georges M, Yang B, Huang L. Mapping and analysis of a spatiotemporal H3K27ac and gene expression spectrum in pigs. SCIENCE CHINA. LIFE SCIENCES 2022; 65:1517-1534. [PMID: 35122624 DOI: 10.1007/s11427-021-2034-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 10/29/2021] [Indexed: 12/12/2022]
Abstract
The limited knowledge of genomic noncoding and regulatory regions has restricted our ability to decipher the genetic mechanisms underlying complex traits in pigs. In this study, we characterized the spatiotemporal landscape of putative enhancers and promoters and their target genes by combining H3K27ac-targeted ChIP-Seq and RNA-Seq in fetal (prenatal days 74-75) and adult (postnatal days 132-150) tissues (brain, liver, heart, muscle and small intestine) sampled from Asian aboriginal Bama Xiang and European highly selected Large White pigs of both sexes. We identified 101,290 H3K27ac peaks, marking 18,521 promoters and 82,769 enhancers, including peaks that were active across all tissues and developmental stages (which could indicate safe harbor locus for exogenous gene insertion) and tissue- and developmental stage-specific peaks (which regulate gene pathways matching tissue- and developmental stage-specific physiological functions). We found that H3K27ac and DNA methylation in the promoter region of the XIST gene may be involved in X chromosome inactivation and demonstrated the utility of the present resource for revealing the regulatory patterns of known causal genes and prioritizing candidate causal variants for complex traits in pigs. In addition, we identified an average of 1,124 super-enhancers per sample and found that they were more likely to show tissue-specific activity than ordinary peaks. We have developed a web browser to improve the accessibility of the results ( http://segtp.jxau.edu.cn/pencode/?genome=susScr11 ).
Collapse
Affiliation(s)
- Yaling Zhu
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045, China
- Laboratory Animal Research Center, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Zhimin Zhou
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Tao Huang
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Zhen Zhang
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Wanbo Li
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Ziqi Ling
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Tao Jiang
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Jiawen Yang
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Siyu Yang
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Yanyuan Xiao
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Carole Charlier
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045, China
- Unit of Animal Genomics, GIGA-Institute and Faculty of Veterinary Medicine, University of Liege, 4000, Liege, Belgium
| | - Michel Georges
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045, China
- Unit of Animal Genomics, GIGA-Institute and Faculty of Veterinary Medicine, University of Liege, 4000, Liege, Belgium
| | - Bin Yang
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045, China.
| | - Lusheng Huang
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045, China.
| |
Collapse
|
10
|
Cuveillier C, Boulan B, Ravanello C, Denarier E, Deloulme JC, Gory-Fauré S, Delphin C, Bosc C, Arnal I, Andrieux A. Beyond Neuronal Microtubule Stabilization: MAP6 and CRMPS, Two Converging Stories. Front Mol Neurosci 2021; 14:665693. [PMID: 34025352 PMCID: PMC8131560 DOI: 10.3389/fnmol.2021.665693] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/09/2021] [Indexed: 12/21/2022] Open
Abstract
The development and function of the central nervous system rely on the microtubule (MT) and actin cytoskeletons and their respective effectors. Although the structural role of the cytoskeleton has long been acknowledged in neuronal morphology and activity, it was recently recognized to play the role of a signaling platform. Following this recognition, research into Microtubule Associated Proteins (MAPs) diversified. Indeed, historically, structural MAPs—including MAP1B, MAP2, Tau, and MAP6 (also known as STOP);—were identified and described as MT-binding and -stabilizing proteins. Extensive data obtained over the last 20 years indicated that these structural MAPs could also contribute to a variety of other molecular roles. Among multi-role MAPs, MAP6 provides a striking example illustrating the diverse molecular and cellular properties of MAPs and showing how their functional versatility contributes to the central nervous system. In this review, in addition to MAP6’s effect on microtubules, we describe its impact on the actin cytoskeleton, on neuroreceptor homeostasis, and its involvement in signaling pathways governing neuron development and maturation. We also discuss its roles in synaptic plasticity, brain connectivity, and cognitive abilities, as well as the potential relationships between the integrated brain functions of MAP6 and its molecular activities. In parallel, the Collapsin Response Mediator Proteins (CRMPs) are presented as examples of how other proteins, not initially identified as MAPs, fall into the broader MAP family. These proteins bind MTs as well as exhibiting molecular and cellular properties very similar to MAP6. Finally, we briefly summarize the multiple similarities between other classical structural MAPs and MAP6 or CRMPs.In summary, this review revisits the molecular properties and the cellular and neuronal roles of the classical MAPs, broadening our definition of what constitutes a MAP.
Collapse
|
11
|
Chen MX, Cheng S, Lei L, Zhang XF, Liu Q, Lin A, Wallis CU, Lukowicz MJ, Sham PC, Li Q, Ao LJ. The effects of maternal SSRI exposure on the serotonin system, prefrontal protein expression and behavioral development in male and female offspring rats. Neurochem Int 2021; 146:105041. [PMID: 33836218 DOI: 10.1016/j.neuint.2021.105041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 03/21/2021] [Accepted: 03/31/2021] [Indexed: 11/24/2022]
Abstract
Fluoxetine (FLX), a commonly used selective serotonin reuptake inhibitor, is often used to treat depression during pregnancy. However, prenatal exposure to FLX has been associated with a series of neuropsychiatric illnesses. The use of a rodent model can provide a clear indication as to whether prenatal exposure to SSRIs, independent of maternal psychiatric disorders or genetic syndromes, can cause long-term behavioral abnormalities in offspring. Thus, the present study aimed to explore whether prenatal FLX exposure causes long-term neurobehavioral effects, and identify the underlying mechanism between FLX and abnormal behaviors. In our study, 12/mg/kg/day of FLX or equal normal saline (NS) was administered to pregnant Sprague-Dawley (SD) rats (FLX = 30, NS = 27) on gestation day 11 till birth. We assessed the physical development and behavior of offspring, and in vivo magnetic resonance spectroscopy (MRS) was conducted to quantify biochemical alterations in the prefrontal cortex (PFC). Ex vivo measurements of brain serotonin level and a proteomic analysis were also undertaken. Our results showed that the offspring (male offspring in particular) of fluoxetine exposed mothers showed delayed physical development, increased anxiety-like behavior, and impaired social interaction. Moreover, down-regulation of 5-HT and SERT expression were identified in the PFC. We also found that prenatal FLX exposure significantly decreased NAA/tCr with 1H-MRS in the PFC of offspring. Finally, a proteomic study revealed sex-dependent differential protein expression. These findings may have translational importance suggesting that using SSRI medication alone in pregnant mothers may result in developmental delay in their offspring. Our results also help guide the choice of outcome measures in identifying of molecular and developmental mechanisms.
Collapse
Affiliation(s)
- Mo Xian Chen
- School of Rehabilitation, Kunming Medical University, Kunming, China
| | - Shu Cheng
- Department of Rehabilitation, China Resources & WISCO General Hospital, Wuhan, China
| | - Lei Lei
- Rehabilitation Medicine Department, The Affiliated Hospital of Southwest Medical University, Tai Ping Road, Luzhou, Sichuan, China
| | - Xiao Fan Zhang
- Department of Psychiatry, Tongji Hospital of Huazhong University of Science and Technology (HUST), China
| | - Qiang Liu
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Aijin Lin
- School of Rehabilitation, Kunming Medical University, Kunming, China
| | | | | | - Pak C Sham
- Department of Psychiatry, The University of Hong Kong, Hong Kong, SAR, China; State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, SAR, China; Centre for Genomic Sciences, The University of Hong Kong, Hong Kong, SAR, China
| | - Qi Li
- Department of Psychiatry, The University of Hong Kong, Hong Kong, SAR, China; State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, SAR, China.
| | - Li Juan Ao
- School of Rehabilitation, Kunming Medical University, Kunming, China.
| |
Collapse
|
12
|
Quach TT, Stratton HJ, Khanna R, Kolattukudy PE, Honnorat J, Meyer K, Duchemin AM. Intellectual disability: dendritic anomalies and emerging genetic perspectives. Acta Neuropathol 2021; 141:139-158. [PMID: 33226471 PMCID: PMC7855540 DOI: 10.1007/s00401-020-02244-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 12/12/2022]
Abstract
Intellectual disability (ID) corresponds to several neurodevelopmental disorders of heterogeneous origin in which cognitive deficits are commonly associated with abnormalities of dendrites and dendritic spines. These histological changes in the brain serve as a proxy for underlying deficits in neuronal network connectivity, mostly a result of genetic factors. Historically, chromosomal abnormalities have been reported by conventional karyotyping, targeted fluorescence in situ hybridization (FISH), and chromosomal microarray analysis. More recently, cytogenomic mapping, whole-exome sequencing, and bioinformatic mining have led to the identification of novel candidate genes, including genes involved in neuritogenesis, dendrite maintenance, and synaptic plasticity. Greater understanding of the roles of these putative ID genes and their functional interactions might boost investigations into determining the plausible link between cellular and behavioral alterations as well as the mechanisms contributing to the cognitive impairment observed in ID. Genetic data combined with histological abnormalities, clinical presentation, and transgenic animal models provide support for the primacy of dysregulation in dendrite structure and function as the basis for the cognitive deficits observed in ID. In this review, we highlight the importance of dendrite pathophysiology in the etiologies of four prototypical ID syndromes, namely Down Syndrome (DS), Rett Syndrome (RTT), Digeorge Syndrome (DGS) and Fragile X Syndrome (FXS). Clinical characteristics of ID have also been reported in individuals with deletions in the long arm of chromosome 10 (the q26.2/q26.3), a region containing the gene for the collapsin response mediator protein 3 (CRMP3), also known as dihydropyrimidinase-related protein-4 (DRP-4, DPYSL4), which is involved in dendritogenesis. Following a discussion of clinical and genetic findings in these syndromes and their preclinical animal models, we lionize CRMP3/DPYSL4 as a novel candidate gene for ID that may be ripe for therapeutic intervention.
Collapse
Affiliation(s)
- Tam T Quach
- Institute for Behavioral Medicine Research, Wexner Medical Center, The Ohio State University, Columbus, OH, 43210, USA
- INSERM U1217/CNRS, UMR5310, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | | | - Rajesh Khanna
- Department of Pharmacology, University of Arizona, Tucson, AZ, 85724, USA
| | | | - Jérome Honnorat
- INSERM U1217/CNRS, UMR5310, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- French Reference Center on Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon, Lyon, France
- SynatAc Team, Institut NeuroMyoGène, Lyon, France
| | - Kathrin Meyer
- The Research Institute of Nationwide Children Hospital, Columbus, OH, 43205, USA
- Department of Pediatric, The Ohio State University, Columbus, OH, 43210, USA
| | - Anne-Marie Duchemin
- Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
13
|
Hou ST. The regulatory and enzymatic functions of CRMPs in neuritogenesis, synaptic plasticity, and gene transcription. Neurochem Int 2020; 139:104795. [PMID: 32652266 DOI: 10.1016/j.neuint.2020.104795] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/18/2020] [Accepted: 06/22/2020] [Indexed: 12/13/2022]
Abstract
Collapsin response mediator proteins (CRMPs) are ubiquitously expressed in neurons from worms to humans. A cardinal feature of CRMPs is to mediate growth cone collapse in response to Semaphorin-3A signaling through interactions with cytoskeletal proteins. These are critical regulatory roles that CRMPs play during neuritogenesis and neural network formation. Through post-translational modifications, such as phosphorylation, O-GlcNAcylation, SUMOylation, and proteolytic cleavage, CRMPs participate in synaptic plasticity by modulating NMDA receptors, L- and N-type voltage-gated calcium channels (VGCCs), thus affecting neurotransmitter release. CRMPs also possess histone deacetylase (HDAC) activity, which deacetylates histone H4 during neuronal death. Calcium-dependent proteolytic cleavage of CRMPs results in the truncation of CRMPs, producing a large 54 kD fragment (p54). Translocation of the p54 fragment into the nucleus leads to deacetylation of nuclear histone H4 and de-repression of transcription factor E2F1 expression. Increased expression of E2F1 elevates the expression of genes in cell cycle and death. These new and exciting studies lead to the realization that CRMPs are multifunctional proteins with both regulatory and enzymatic functions. Increasing numbers of studies associate these functions of CRMPs with the development of mental and neurological disorders, such as schizophrenia, Alzheimer's diseases, brain trauma, and stroke. This review focuses on new evidence showing the regulatory and enzymatic functions of CRMPs and highlights recent understandings of CRMPs' roles in neurological diseases.
Collapse
Affiliation(s)
- Sheng-Tao Hou
- Brain Research Centre and Department of Biology, Southern University of Science and Technology, 1088 Xueyuan Blvd, Nanshan District, Shenzhen, Guangdong Province, 518055, PR China; Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada.
| |
Collapse
|
14
|
Quach TT, Moutal A, Khanna R, Deems NP, Duchemin AM, Barrientos RM. Collapsin Response Mediator Proteins: Novel Targets for Alzheimer's Disease. J Alzheimers Dis 2020; 77:949-960. [PMID: 32804096 PMCID: PMC7579750 DOI: 10.3233/jad-200721] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Numerous experimental and postmortem studies have increasingly reported dystrophic axons and dendrites, and alterations of dendritic spine morphology and density in the hippocampus as prominent changes in the early stages of Alzheimer's disease (AD). Furthermore, these alterations tend to correlate well with the progressive cognitive decline observed in AD. For these reasons, and because these neurite structures have a capacity to re-grow, re-establish lost connections, and are critical for learning and memory, there is compelling evidence to suggest that therapeutic interventions aimed at preventing their degradation or promoting their regrowth may hold tremendous promise in preventing the progression of AD. In this regard, collapsin response mediator proteins (CRMPs), a family of phosphoproteins playing a major role in axon guidance and dendritic growth, are especially interesting. The roles these proteins play in neurons and immune cells are reviewed here.
Collapse
Affiliation(s)
- Tam T. Quach
- Institute for Behavioral Medicine Research, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Aubin Moutal
- Department of Pharmacology, University of Arizona, Tucson, AZ, 85724, USA
| | - Rajesh Khanna
- Department of Pharmacology, University of Arizona, Tucson, AZ, 85724, USA
| | - Nicholas P. Deems
- Institute for Behavioral Medicine Research, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Anne-Marie Duchemin
- Department of Psychiatry and Behavioral Health, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Ruth M. Barrientos
- Institute for Behavioral Medicine Research, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
- Chronic Brain Injury Program, Discovery Themes Initiative, The Ohio State University, Columbus, OH, 43210, USA
- Department of Psychiatry and Behavioral Health, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
- Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
15
|
Han YY, Chen ZH, Shang YJ, Yan WW, Wu BY, Li CH. Cordycepin improves behavioral-LTP and dendritic structure in hippocampal CA1 area of rats. J Neurochem 2019; 151:79-90. [PMID: 31314908 DOI: 10.1111/jnc.14826] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 07/02/2019] [Accepted: 07/04/2019] [Indexed: 12/21/2022]
Abstract
Cordycepin, an adenosine analog, has been reported to improve cognitive function, but which seems to be inconsistent with the reports showing that cordycepin inhibited long-term potentiation (LTP). Behavioral-LTP is usually used to study long-term synaptic plasticity induced by learning tasks in freely moving animals. In order to investigate simultaneously the effects of cordycepin on LTP and behavior in rats, we applied the model of behavioral-LTP induced by Y-maze learning task through recording population spikes in hippocampal CA1 region. Golgi staining and Sholl analysis were employed to assess the morphological structure of dendrites in pyramidal cells of hippocampal CA1 area, and western blotting was used to examine the level of adenosine A1 receptors and A2A receptors (A2AR). We found that cordycepin significantly improved behavioral-LTP magnitude, accompanied by increases in the total length of dendrites, the number of intersections and spine density but did not affect Y-maze learning task. Furthermore, cordycepin obviously reduced A2AR level without altering adenosine A1 receptors level; and the agonist of A2AR (CGS 21680) rather than antagonist (SCH 58261) could reverse the potentiation of behavioral-LTP induced by cordycepin. These results suggested that cordycepin improved behavioral-LTP and morphological structure of dendrite in hippocampal CA1 but did not contribute to the improvement of learning and memory. And cordycepin improved behavioral-LTP may be through reducing the level of A2AR in hippocampus. Collectively, the effects of cordycepin on cognitive function and LTP were complex and involved multiple mechanisms.
Collapse
Affiliation(s)
- Yuan-Yuan Han
- School of Life Science, South China Normal University, Guangzhou, China
| | - Zhao-Hui Chen
- School of Life Science, South China Normal University, Guangzhou, China
| | - Ying-Jie Shang
- School of Life Science, South China Normal University, Guangzhou, China
| | - Wen-Wen Yan
- School of Life Science, South China Normal University, Guangzhou, China
| | - Bao-Yan Wu
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| | - Chu-Hua Li
- School of Life Science, South China Normal University, Guangzhou, China
| |
Collapse
|
16
|
Lin YS, Lin YF, Chen KC, Yang YK, Hsiao YH. Collapsin response mediator protein 5 (CRMP5) causes social deficits and accelerates memory loss in an animal model of Alzheimer's disease. Neuropharmacology 2019; 157:107673. [PMID: 31233825 DOI: 10.1016/j.neuropharm.2019.107673] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 06/10/2019] [Accepted: 06/13/2019] [Indexed: 02/02/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by several behavioral disturbances, especially cognitive decline and deficits in social competence. Previous studies revealed that decreased social activity would accelerate AD progression, whereas enhanced social interaction could rescue AD-induced memory impairment. Collapsin response mediator protein 5 (CRMP5), which belongs to a family of cytosolic proteins, is abundantly expressed in the brain and is involved in the regulation of neurodevelopment and the pathology of several neuropsychiatric diseases. However, the functions of CRMP5 in AD are still unclear. Here, we demonstrated that 9-month-old 3xTg-AD mice exhibited social behavioral deficits and increased hippocampal CRMP5 levels compared to control (B6129S) mice. Knockdown of CRMP5 reversed the social deficits in 9-month-old 3xTg-AD mice, whereas CRMP5 overexpression decreased social interaction in both 3xTg-AD and control mice at 6 months of age. Interestingly, decreased expression of CRMP5 rescued AD-induced memory impairment, but overexpression of CRMP5 accelerated memory loss only in 3xTg-AD mice. In addition, we found that CRMP5 could regulate surface GluA2 and GluA2 S880 phosphorylation. These results suggest that CRMP5 regulates social behavior via modulation of surface GluA2 trafficking and affects memory performance in 3xTg-AD mice.
Collapse
Affiliation(s)
- Yung-Shuen Lin
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Fen Lin
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Kao Chin Chen
- Department of Psychiatry, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yen Kuang Yang
- Department of Psychiatry, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Psychiatry, National Cheng Kung University Hospital, Dou-Liou Branch, Yunlin, Taiwan
| | - Ya-Hsin Hsiao
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Institute of Behavioral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
17
|
Opposing Morphogenetic Defects on Dendrites and Mossy Fibers of Dentate Granular Neurons in CRMP3-Deficient Mice. Brain Sci 2018; 8:brainsci8110196. [PMID: 30400291 PMCID: PMC6265834 DOI: 10.3390/brainsci8110196] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 10/30/2018] [Accepted: 10/31/2018] [Indexed: 12/27/2022] Open
Abstract
Collapsin response mediator proteins (CRMPs) are highly expressed in the brain during early postnatal development and continue to be present in specific regions into adulthood, especially in areas with extensive neuronal plasticity including the hippocampus. They are found in the axons and dendrites of neurons wherein they contribute to specific signaling mechanisms involved in the regulation of axonal and dendritic development/maintenance. We previously identified CRMP3’s role on the morphology of hippocampal CA1 pyramidal dendrites and hippocampus-dependent functions. Our focus here was to further analyze its role in the dentate gyrus where it is highly expressed during development and in adults. On the basis of our new findings, it appears that CRMP3 has critical roles both in axonal and dendritic morphogenesis of dentate granular neurons. In CRMP3-deficient mice, the dendrites become dystrophic while the infrapyramidal bundle of the mossy fiber shows aberrant extension into the stratum oriens of CA3. This axonal misguided projection of granular neurons suggests that the mossy fiber-CA3 synaptic transmission, important for the evoked propagation of the activity of the hippocampal trisynaptic circuitry, may be altered, whereas the dystrophic dendrites may impair the dynamic interactions with the entorhinal cortex, both expected to affect hippocampal function.
Collapse
|
18
|
|
19
|
Proteome and behavioral alterations in phosphorylation-deficient mutant Collapsin Response Mediator Protein2 knock-in mice. Neurochem Int 2018; 119:207-217. [PMID: 29758318 DOI: 10.1016/j.neuint.2018.04.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 03/08/2018] [Accepted: 04/19/2018] [Indexed: 02/02/2023]
Abstract
CRMP2, alternatively designated as DPYSL2, was the first CRMP family member to be identified as an intracellular molecule mediating the signaling of the axon guidance molecule Semaphorin 3A (Sema3A). In Sema3A signaling, cyclin-dependent kinase 5 (Cdk5) primarily phosphorylates CRMP2 at Ser522. Glycogen synthase kinase-3β (GSK-3β) subsequently phosphorylates the residues of Thr509 and Thr514 of CRMP2. Previous studies showed that CRMP2 is involved in pathogenesis of neurological disorders such as Alzheimer's disease. In Alzheimer's disease, hyper-phosphorylated forms of CRMP2 are accumulated in the paired helical filaments. To get insight into the possible involvement of the phosphorylation of CRMP2 in pathogenesis of neurological disorders, we previously created CRMP2 S522A knock-in (crmp2ki/ki) mice and demonstrated that the phosphorylation of CRMP2 at Ser522 is involved in normal dendrite patterning in cortical neurons. However, the behavioral impact and in vivo signaling network of the CRMP2 phosphorylation are not fully understood. In this study, we performed behavioral and proteomics analysis of crmp2ki/ki mice. The crmp2ki/ki mice appeared healthy and showed no obvious differences in physical characteristics compared to wild-type mice, but they showed impaired emotional behavior, reduced sociality, and low sensitivity to pain stimulation. Through mass-spectrometry-based proteomic analysis, we found that 59 proteins were increased and 77 proteins were decreased in the prefrontal cortex of crmp2ki/ki mice. Notably, CRMP3, CRMP4, and CRMP5, the other CRMP family proteins, were increased in crmp2ki/ki mice. KEGG (Kyoto Encyclopedia of Genes and Genomes) pathway analyses identified 14 pathways in increased total proteins and 13 pathways in decreased total proteins which are associated with the pathogenesis of Parkinson's, Alzheimer's, and Huntington's diseases. We also detected 20 pathways in increased phosphopeptides and 16 pathways in decreased phosphopeptides including "inflammatory mediator regulation of TRP channels" in crmp2ki/ki mice. Our study suggests that the phosphorylation of CRMP2 at Ser522 is involved in the signaling pathways that may be related to neuropsychiatric and neurodegenerative diseases and pain.
Collapse
|
20
|
Hong F, Ze Y, Zhou Y, Hong J, Yu X, Sheng L, Wang L. Nanoparticulate TiO 2 -mediated inhibition of the Wnt signaling pathway causes dendritic development disorder in cultured rat hippocampal neurons. J Biomed Mater Res A 2017; 105:2139-2149. [PMID: 28371053 DOI: 10.1002/jbm.a.36073] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Revised: 02/26/2017] [Accepted: 03/24/2017] [Indexed: 11/11/2022]
Abstract
Titanium dioxide nanoparticles (TiO2 NPs) are increasingly used in daily life, in industry, and in environmental clearing, but their potential neurodevelopmental toxicity has been highly debated. In this study, we explored whether TiO2 NPs inhibited development of dendritic morphology and identified possible molecular mechanisms associated with this inhibition in primary cultured rat hippocampal neurons. Results showed that TiO2 NPs decreased neurite length, the number of branches and the spine density, and impaired mitochondrial function in the developing neurons. Furthermore, TiO2 NPs significantly reduced the expression of several proteins involved in canonical Wnt3a/β-catenin signaling including Wnt3a, β-catenin, p-GSK-3β, and CyclinD1 and conversely, elevated GSK-3β expression. In addition to altering expression of proteins involved in canonical Wnt3a/β-catenin signaling, TiO2 NPs decreased expression of proteins invovled in non-canonical Wnt signaling, including, MKLP1, CRMP3, ErbB4, and KIF17. Taken together, these results indicate that suppression of dendritic development caused by TiO2 NPs is associated with inhibition of activation of the Wnt/β-catenin pathway or non-canonical Wnt pathway-induced expression of microtubule cytoskeletal components in the developing neurons. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 2139-2149, 2017.
Collapse
Affiliation(s)
- Fashui Hong
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University, Huaian, 223300, China.,Jiangsu Key Laboratory for Food Safety and Nutritional Function, Huaiyin Normal University, Huaian, 223300, China.,Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University, Huaian, 223300, China
| | - Yuguan Ze
- Medical College of Soochow University, Suzhou, 215123, China
| | - Yaoming Zhou
- Food Department, Jiangsu Food and Pharmaceutical Science College, Huaian, 223303, China
| | - Jie Hong
- Medical College of Soochow University, Suzhou, 215123, China
| | - Xiaohon Yu
- Medical College of Soochow University, Suzhou, 215123, China
| | - Lei Sheng
- Medical College of Soochow University, Suzhou, 215123, China
| | - Ling Wang
- Library of Soochow University, Suzhou, China, Suzhou, 215123, China
| |
Collapse
|
21
|
Nakamura H, Yamashita N, Kimura A, Kimura Y, Hirano H, Makihara H, Kawamoto Y, Jitsuki-Takahashi A, Yonezaki K, Takase K, Miyazaki T, Nakamura F, Tanaka F, Goshima Y. Comprehensive behavioral study and proteomic analyses of CRMP2-deficient mice. Genes Cells 2016; 21:1059-1079. [DOI: 10.1111/gtc.12403] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Accepted: 07/29/2016] [Indexed: 01/02/2023]
Affiliation(s)
- Haruko Nakamura
- Department of Molecular Pharmacology and Neurobiology; Yokohama City University Graduate School of Medicine; Yokohama 236-0004 Japan
- Department of Neurology and Stroke Medicine; Yokohama City University Graduate School of Medicine; Yokohama 236-0004 Japan
| | - Naoya Yamashita
- Department of Molecular Pharmacology and Neurobiology; Yokohama City University Graduate School of Medicine; Yokohama 236-0004 Japan
- JSPS Postdoctoral Fellowship for Research Abroad; Tokyo 102-0083 Japan
| | - Ayuko Kimura
- Advanced Medical Research Center; Yokohama City University; Yokohama 236-0004 Japan
| | - Yayoi Kimura
- Advanced Medical Research Center; Yokohama City University; Yokohama 236-0004 Japan
| | - Hisashi Hirano
- Advanced Medical Research Center; Yokohama City University; Yokohama 236-0004 Japan
| | - Hiroko Makihara
- Department of Molecular Pharmacology and Neurobiology; Yokohama City University Graduate School of Medicine; Yokohama 236-0004 Japan
| | - Yuko Kawamoto
- Department of Molecular Pharmacology and Neurobiology; Yokohama City University Graduate School of Medicine; Yokohama 236-0004 Japan
- Department of Neurology and Stroke Medicine; Yokohama City University Graduate School of Medicine; Yokohama 236-0004 Japan
| | - Aoi Jitsuki-Takahashi
- Department of Molecular Pharmacology and Neurobiology; Yokohama City University Graduate School of Medicine; Yokohama 236-0004 Japan
| | - Kumiko Yonezaki
- Department of Anesthesiology; Yokohama City University Graduate School of Medicine; Yokohama 236-0004 Japan
| | - Kenkichi Takase
- Department of Anesthesiology; Yokohama City University Graduate School of Medicine; Yokohama 236-0004 Japan
- Laboratory of Psychology; Jichi Medical University; Shimotsuke 329-0498 Japan
| | - Tomoyuki Miyazaki
- Department of Anesthesiology; Yokohama City University Graduate School of Medicine; Yokohama 236-0004 Japan
- Department of Physiology; Yokohama City University Graduate School of Medicine; Yokohama 236-0004 Japan
| | - Fumio Nakamura
- Department of Molecular Pharmacology and Neurobiology; Yokohama City University Graduate School of Medicine; Yokohama 236-0004 Japan
| | - Fumiaki Tanaka
- Department of Neurology and Stroke Medicine; Yokohama City University Graduate School of Medicine; Yokohama 236-0004 Japan
| | - Yoshio Goshima
- Department of Molecular Pharmacology and Neurobiology; Yokohama City University Graduate School of Medicine; Yokohama 236-0004 Japan
| |
Collapse
|
22
|
Nagai J, Baba R, Ohshima T. CRMPs Function in Neurons and Glial Cells: Potential Therapeutic Targets for Neurodegenerative Diseases and CNS Injury. Mol Neurobiol 2016; 54:4243-4256. [PMID: 27339876 DOI: 10.1007/s12035-016-0005-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Accepted: 06/14/2016] [Indexed: 12/19/2022]
Abstract
Neurodegeneration in the adult mammalian central nervous system (CNS) is fundamentally accelerated by its intrinsic neuronal mechanisms, including its poor regenerative capacity and potent extrinsic inhibitory factors. Thus, the treatment of neurodegenerative diseases faces many obstacles. The degenerative processes, consisting of axonal/dendritic structural disruption, abnormal axonal transport, release of extracellular factors, and inflammation, are often controlled by the cytoskeleton. From this perspective, regulators of the cytoskeleton could potentially be a therapeutic target for neurodegenerative diseases and CNS injury. Collapsin response mediator proteins (CRMPs) are known to regulate the assembly of cytoskeletal proteins in neurons, as well as control axonal growth and neural circuit formation. Recent studies have provided some novel insights into the roles of CRMPs in several inhibitory signaling pathways of neurodegeneration, in addition to its functions in neurological disorders and CNS repair. Here, we summarize the roles of CRMPs in axon regeneration and its emerging functions in non-neuronal cells, especially in inflammatory responses. We also discuss the direct and indirect targeting of CRMPs as a novel therapeutic strategy for neurological diseases.
Collapse
Affiliation(s)
- Jun Nagai
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, TWIns, 2-2 Wakamatsu-cho Shinjuku-ku, Tokyo, 162-8480, Japan.,Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
| | - Rina Baba
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, TWIns, 2-2 Wakamatsu-cho Shinjuku-ku, Tokyo, 162-8480, Japan
| | - Toshio Ohshima
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, TWIns, 2-2 Wakamatsu-cho Shinjuku-ku, Tokyo, 162-8480, Japan.
| |
Collapse
|
23
|
Tsutiya A, Watanabe H, Nakano Y, Nishihara M, Goshima Y, Ohtani‐Kaneko R. Deletion of collapsin response mediator protein 4 results in abnormal layer thickness and elongation of mitral cell apical dendrites in the neonatal olfactory bulb. J Anat 2016; 228:792-804. [PMID: 26739921 PMCID: PMC4831339 DOI: 10.1111/joa.12434] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2015] [Indexed: 11/28/2022] Open
Abstract
Collapsin response mediator protein 4 (CRMP4), a member of the CRMP family, is involved in the pathogenesis of neurodevelopmental disorders such as schizophrenia and autism. Here, we first compared layer thickness of the olfactory bulb between wild-type (WT) and CRMP4-knockout (KO) mice. The mitral cell layer (MCL) was significantly thinner, whereas the external plexiform layer (EPL) was significantly thicker in CRMP4-KO mice at postnatal day 0 (PD0) compared with WTs. However, differences in layer thickness disappeared by PD14. No apoptotic cells were found in the MCL, and the number of mitral cells (MCs) identified with a specific marker (i.e. Tbx21 antibody) did not change in CRMP4-KO neonates. However, DiI-tracing showed that the length of mitral cell apical dendrites was greater in CRMP4-KO neonates than in WTs. In addition, expression of CRMP4 mRNA in WT mice was most abundant in the MCL at PD0 and decreased afterward. These results suggest that CRMP4 contributes to dendritic elongation. Our in vitro studies showed that deletion or knockdown of CRMP4 resulted in enhanced growth of MAP2-positive neurites, whereas overexpression of CRMP4 reduced their growth, suggesting a new role for CRMP4 as a suppressor of dendritic elongation. Overall, our data suggest that disruption of CRMP4 produces a temporary alteration in EPL thickness, which is constituted mainly of mitral cell apical dendrites, through the enhanced growth of these dendrites.
Collapse
Affiliation(s)
| | - Hikaru Watanabe
- Graduate School of Life SciencesToyo UniversityOuraGunmaJapan
| | - Yui Nakano
- Graduate School of Life SciencesToyo UniversityOuraGunmaJapan
| | - Masugi Nishihara
- Department of Veterinary PhysiologyGraduate School of Agricultural and Life SciencesThe University of TokyoBunkyo‐kuTokyoJapan
| | - Yoshio Goshima
- Department of Molecular Pharmacology and NeurobiologyYokohama City University Graduate School of MedicineYokohamaKanazawa WardJapan
| | - Ritsuko Ohtani‐Kaneko
- Graduate School of Life SciencesToyo UniversityOuraGunmaJapan
- Bio‐Nano Electronic Research CentreToyo UniversityKawagoeSaitamaJapan
| |
Collapse
|
24
|
Quach TT, Lerch JK, Honnorat J, Khanna R, Duchemin AM. Neuronal networks in mental diseases and neuropathic pain: Beyond brain derived neurotrophic factor and collapsin response mediator proteins. World J Psychiatry 2016; 6:18-30. [PMID: 27014595 PMCID: PMC4804265 DOI: 10.5498/wjp.v6.i1.18] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 11/24/2015] [Accepted: 01/07/2016] [Indexed: 02/05/2023] Open
Abstract
The brain is a complex network system that has the capacity to support emotion, thought, action, learning and memory, and is characterized by constant activity, constant structural remodeling, and constant attempt to compensate for this remodeling. The basic insight that emerges from complex network organization is that substantively different networks can share common key organizational principles. Moreover, the interdependence of network organization and behavior has been successfully demonstrated for several specific tasks. From this viewpoint, increasing experimental/clinical observations suggest that mental disorders are neural network disorders. On one hand, single psychiatric disorders arise from multiple, multifactorial molecular and cellular structural/functional alterations spreading throughout local/global circuits leading to multifaceted and heterogeneous clinical symptoms. On the other hand, various mental diseases may share functional deficits across the same neural circuit as reflected in the overlap of symptoms throughout clinical diagnoses. An integrated framework including experimental measures and clinical observations will be necessary to formulate a coherent and comprehensive understanding of how neural connectivity mediates and constraints the phenotypic expression of psychiatric disorders.
Collapse
|
25
|
Menon S, Gupton SL. Building Blocks of Functioning Brain: Cytoskeletal Dynamics in Neuronal Development. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 322:183-245. [PMID: 26940519 PMCID: PMC4809367 DOI: 10.1016/bs.ircmb.2015.10.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neural connectivity requires proper polarization of neurons, guidance to appropriate target locations, and establishment of synaptic connections. From when neurons are born to when they finally reach their synaptic partners, neurons undergo constant rearrangment of the cytoskeleton to achieve appropriate shape and polarity. Of particular importance to neuronal guidance to target locations is the growth cone at the tip of the axon. Growth-cone steering is also dictated by the underlying cytoskeleton. All these changes require spatiotemporal control of the cytoskeletal machinery. This review summarizes the proteins that are involved in modulating the actin and microtubule cytoskeleton during the various stages of neuronal development.
Collapse
Affiliation(s)
- Shalini Menon
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, United States of America
| | - Stephanie L Gupton
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, United States of America; Neuroscience Center and Curriculum in Neurobiology, University of North Carolina, Chapel Hill, NC, United States of America; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, United States of America.
| |
Collapse
|
26
|
Tsutiya A, Nishihara M, Goshima Y, Ohtani-Kaneko R. Mouse pups lacking collapsin response mediator protein 4 manifest impaired olfactory function and hyperactivity in the olfactory bulb. Eur J Neurosci 2015; 42:2335-45. [PMID: 26118640 DOI: 10.1111/ejn.12999] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 06/11/2015] [Accepted: 06/22/2015] [Indexed: 01/07/2023]
Abstract
Members of the collapsin response mediator protein (CRMP) family are reported to be involved in the pathogenesis of various neuronal disorders, including schizophrenia and autism. One of them, CRMP4, is reported to participate in aspects of neuronal development, such as axonal guidance and dendritic development. However, no physiological or behavioral phenotypes in Crmp4 knockout (Crmp4-KO) mice have been identified, making it difficult to elucidate the in vivo roles of CRMP4. Focusing on the olfaction process because of the previous study showing strong expression of Crmp4 mRNA in the olfactory bulb (OB) during the early postnatal period, it was aimed to test the hypothesis that Crmp4-KO pups would exhibit abnormal olfaction. Based on measurements of their ultrasonic vocalizations, impaired olfactory ability in Crmp4-KO pups was found. In addition, c-Fos expression, a marker of neuron activity, revealed hyperactivity in the OB of Crmp4-KO pups compared with wild-types following exposure to an odorant. Moreover, the mRNA and protein expression levels of glutamate receptor 1 (GluR1) and 2 (GluR2) were exaggerated in Crmp4-KO pups relative to other excitatory and inhibitory receptors and transporters, raising the possibility that enhanced expression of these excitatory receptors contributes to the hyperactivity phenotype and impairs olfactory ability. This study provides evidence for an animal model for elucidating the roles of CRMP4 in the development of higher brain functions as well as for elucidating the developmental regulatory mechanisms controlling the activity of the neural circuitry.
Collapse
Affiliation(s)
- Atsuhiro Tsutiya
- Graduate School of Life Sciences, Toyo University, 1-1-1 Itakura, Oura, Gunma, 374-0193, Japan
| | - Masugi Nishihara
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Yoshio Goshima
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Ritsuko Ohtani-Kaneko
- Graduate School of Life Sciences, Toyo University, 1-1-1 Itakura, Oura, Gunma, 374-0193, Japan
- Bio-Nano Electronic Research Centre, Toyo University, Kawagoe, Saitama, Japan
| |
Collapse
|
27
|
CRMPs: critical molecules for neurite morphogenesis and neuropsychiatric diseases. Mol Psychiatry 2015; 20:1037-45. [PMID: 26077693 DOI: 10.1038/mp.2015.77] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 04/29/2015] [Accepted: 05/08/2015] [Indexed: 12/11/2022]
Abstract
Neuronal polarity and spatial rearrangement of neuronal processes are central to the development of all mature nervous systems. Recent studies have highlighted the dynamic expression of Collapsin-Response-Mediator Proteins (CRMPs) in neuronal dendritic/axonal compartments, described their interaction with cytoskeleton proteins, identified their ability to activate L- and N-type voltage-gated calcium channels (VGCCs) and delineated their crucial role as signaling molecules essential for neuron differentiation and neural network development and maintenance. In addition, evidence obtained from genome-wide/genetic linkage/proteomic/translational approaches revealed that CRMP expression is altered in human pathologies including mental (schizophrenia and mood disorders) and neurological (Alzheimer's, prion encephalopathy, epilepsy and others) disorders. Changes in CRMPs levels have been observed after psychotropic treatments, and disrupting CRMP2 binding to calcium channels blocked neuropathic pain. These observations, altogether with those obtained from genetically modified mice targeting individual CRMPs and RNA interference approaches, pave the way for considering CRMPs as potential early disease markers and modulation of their activity as therapeutic strategy for disorders associated with neurite abnormalities.
Collapse
|
28
|
Kadoyama K, Matsuura K, Nakamura-Hirota T, Takano M, Otani M, Matsuyama S. Changes in the expression of collapsin response mediator protein-2 during synaptic plasticity in the mouse hippocampus. J Neurosci Res 2015; 93:1684-92. [DOI: 10.1002/jnr.23626] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 07/23/2015] [Accepted: 07/23/2015] [Indexed: 11/09/2022]
Affiliation(s)
- Keiichi Kadoyama
- Department of Pharmaceutical Health Care; Faculty of Pharmaceutical Sciences; Himeji Dokkyo University; Himeji Japan
| | - Kenji Matsuura
- Department of Pharmaceutical Health Care; Faculty of Pharmaceutical Sciences; Himeji Dokkyo University; Himeji Japan
| | - Tooru Nakamura-Hirota
- Department of Pharmaceutical Health Care; Faculty of Pharmaceutical Sciences; Himeji Dokkyo University; Himeji Japan
| | - Masaoki Takano
- Department of Life Sciences Pharmacy; School of Pharmaceutical Sciences, Kobe Gakuin University; Kobe Japan
| | - Mieko Otani
- Department of Life Sciences Pharmacy; School of Pharmaceutical Sciences, Kobe Gakuin University; Kobe Japan
| | - Shogo Matsuyama
- Department of Pharmaceutical Health Care; Faculty of Pharmaceutical Sciences; Himeji Dokkyo University; Himeji Japan
| |
Collapse
|
29
|
Oei GTML, Heger M, van Golen RF, Alles LK, Flick M, van der Wal AC, van Gulik TM, Hollmann MW, Preckel B, Weber NC. Reduction of cardiac cell death after helium postconditioning in rats: transcriptional analysis of cell death and survival pathways. Mol Med 2015; 20:516-26. [PMID: 25171109 DOI: 10.2119/molmed.2014.00057] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 08/26/2014] [Indexed: 12/28/2022] Open
Abstract
Helium, a noble gas, has been used safely in humans. In animal models of regional myocardial ischemia/reperfusion (I/R) it was shown that helium conditioning reduces infarct size. Currently, it is not known how helium exerts its cytoprotective effects and which cell death/survival pathways are affected. The objective of this study, therefore, was to investigate the cell protective effects of helium postconditioning by PCR array analysis of genes involved in necrosis, apoptosis and autophagy. Male rats were subjected to 25 min of ischemia and 5, 15 or 30 min of reperfusion. Semiquantitative histological analysis revealed that 15 min of helium postconditioning reduced the extent of I/R-induced cell damage. This effect was not observed after 5 and 30 min of helium postconditioning. Analysis of the differential expression of genes showed that 15 min of helium postconditioning mainly caused upregulation of genes involved in autophagy and inhibition of apoptosis versus I/R alone. The results suggest that the cytoprotective effects of helium inhalation may be caused by a switch from pro-cell-death signaling to activation of cell survival mechanisms, which appears to affect a wide range of pathways.
Collapse
Affiliation(s)
- Gezina T M L Oei
- Department of Anesthesiology, Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), University of Amsterdam, Amsterdam, The Netherlands
| | - Michal Heger
- Department of Experimental Surgery, University of Amsterdam, Amsterdam, The Netherlands
| | - Rowan F van Golen
- Department of Experimental Surgery, University of Amsterdam, Amsterdam, The Netherlands
| | - Lindy K Alles
- Department of Experimental Surgery, University of Amsterdam, Amsterdam, The Netherlands
| | - Moritz Flick
- Department of Anesthesiology, Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), University of Amsterdam, Amsterdam, The Netherlands
| | - Allard C van der Wal
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Thomas M van Gulik
- Department of Experimental Surgery, University of Amsterdam, Amsterdam, The Netherlands
| | - Markus W Hollmann
- Department of Anesthesiology, Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), University of Amsterdam, Amsterdam, The Netherlands
| | - Benedikt Preckel
- Department of Anesthesiology, Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), University of Amsterdam, Amsterdam, The Netherlands
| | - Nina C Weber
- Department of Anesthesiology, Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
30
|
Molas S, Gener T, Güell J, Martín M, Ballesteros-Yáñez I, Sanchez-Vives MV, Dierssen M. Hippocampal changes produced by overexpression of the human CHRNA5/A3/B4 gene cluster may underlie cognitive deficits rescued by nicotine in transgenic mice. Acta Neuropathol Commun 2014; 2:147. [PMID: 25384568 PMCID: PMC4236452 DOI: 10.1186/s40478-014-0147-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 10/02/2014] [Indexed: 11/10/2022] Open
Abstract
Addiction involves long-lasting maladaptive changes including development of disruptive drug-stimuli associations. Nicotine-induced neuroplasticity underlies the development of tobacco addiction but also, in regions such as the hippocampus, the ability of this drug to enhance cognitive capabilities. Here, we propose that the genetic locus of susceptibility to nicotine addiction, the CHRNA5/A3/B4 gene cluster, encoding the α5, α3 and β4 subunits of the nicotinic acetylcholine receptors (nAChRs), may influence nicotine-induced neuroadaptations. We have used transgenic mice overexpressing the human cluster (TgCHRNA5/A3/B4) to investigate hippocampal structure and function in genetically susceptible individuals. TgCHRNA5/A3/B4 mice presented a marked reduction in the dendrite complexity of CA1 hippocampal pyramidal neurons along with an increased dendritic spine density. In addition, TgCHRNA5/A3/B4 exhibited increased VGLUT1/VGAT ratio in the CA1 region, suggesting an excitatory/inhibitory imbalance. These hippocampal alterations were accompanied by a significant impairment in short-term novelty recognition memory. Interestingly, chronic infusion of nicotine (3.25 mg/kg/d for 7 d) was able to rescue the reduced dendritic complexity, the excitatory/inhibitory imbalance and the cognitive impairment in TgCHRNA5/A3/B4. Our results suggest that chronic nicotine treatment may represent a compensatory strategy in individuals with altered expression of the CHRNA5/A3/B4 region.
Collapse
|
31
|
Brot S, Smaoune H, Youssef-Issa M, Malleval C, Benetollo C, Besançon R, Auger C, Moradi-Améli M, Honnorat J. Collapsin response-mediator protein 5 (CRMP5) phosphorylation at threonine 516 regulates neurite outgrowth inhibition. Eur J Neurosci 2014; 40:3010-20. [DOI: 10.1111/ejn.12674] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 06/16/2014] [Indexed: 12/28/2022]
Affiliation(s)
- Sébastien Brot
- Lyon Neuroscience Research Center; INSERM; UMR-S1028; CNRS UMR5292; Neuro-Oncology & Neuro-Inflammation Team; Lyon F-69372 France
- Université de Lyon; Université Claude Bernard Lyon 1; Lyon F-69000 France
| | - Hinda Smaoune
- Lyon Neuroscience Research Center; INSERM; UMR-S1028; CNRS UMR5292; Neuro-Oncology & Neuro-Inflammation Team; Lyon F-69372 France
- Université de Lyon; Université Claude Bernard Lyon 1; Lyon F-69000 France
| | - Mina Youssef-Issa
- Lyon Neuroscience Research Center; INSERM; UMR-S1028; CNRS UMR5292; Neuro-Oncology & Neuro-Inflammation Team; Lyon F-69372 France
- Université de Lyon; Université Claude Bernard Lyon 1; Lyon F-69000 France
| | - Céline Malleval
- Lyon Neuroscience Research Center; INSERM; UMR-S1028; CNRS UMR5292; Neuro-Oncology & Neuro-Inflammation Team; Lyon F-69372 France
- Université de Lyon; Université Claude Bernard Lyon 1; Lyon F-69000 France
| | - Claire Benetollo
- Neurogenetic and Optogenetic Platform; Lyon Neuroscience Research Center; Lyon Cedex 08 F-69372 France
| | - Roger Besançon
- Lyon Neuroscience Research Center; INSERM; UMR-S1028; CNRS UMR5292; Neuro-Oncology & Neuro-Inflammation Team; Lyon F-69372 France
- Université de Lyon; Université Claude Bernard Lyon 1; Lyon F-69000 France
| | - Carole Auger
- Lyon Neuroscience Research Center; INSERM; UMR-S1028; CNRS UMR5292; Neuro-Oncology & Neuro-Inflammation Team; Lyon F-69372 France
- Université de Lyon; Université Claude Bernard Lyon 1; Lyon F-69000 France
| | - Mahnaz Moradi-Améli
- Lyon Neuroscience Research Center; INSERM; UMR-S1028; CNRS UMR5292; Neuro-Oncology & Neuro-Inflammation Team; Lyon F-69372 France
- Université de Lyon; Université Claude Bernard Lyon 1; Lyon F-69000 France
| | - Jérôme Honnorat
- Lyon Neuroscience Research Center; INSERM; UMR-S1028; CNRS UMR5292; Neuro-Oncology & Neuro-Inflammation Team; Lyon F-69372 France
- Université de Lyon; Université Claude Bernard Lyon 1; Lyon F-69000 France
- Hospices Civils de Lyon; Neuro-Oncologie; Bron F-69677 France
| |
Collapse
|
32
|
Imperlini E, Orrù S, Corbo C, Daniele A, Salvatore F. Altered brain protein expression profiles are associated with molecular neurological dysfunction in the PKU mouse model. J Neurochem 2014; 129:1002-12. [PMID: 24548049 PMCID: PMC4286000 DOI: 10.1111/jnc.12683] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 01/07/2014] [Accepted: 02/02/2014] [Indexed: 12/14/2022]
Abstract
Phenylketonuria (PKU), if not detected and treated in newborns, causes severe neurological dysfunction and cognitive and behavioral deficiencies. Despite the biochemical characterization of PKU, the molecular mechanisms underlying PKU-associated brain dysfunction remain poorly understood. The aim of this study was to gain insights into the pathogenesis of this neurological damage by analyzing protein expression profiles in brain tissue of Black and Tan BRachyury-PahEnu2 mice (a mouse model of PKU). We compared the cerebral protein expression of homozygous PKU mice with that of their heterozygous counterparts using two-dimensional difference gel electrophoresis analysis, and identified 21 differentially expressed proteins, four of which were over-expressed and 17 under-expressed. An in silico bioinformatic approach indicated that protein under-expression was related to neuronal differentiation and dendritic growth, and to such neurological disorders as progressive motor neuropathy and movement disorders. Moreover, functional annotation analyses showed that some identified proteins were involved in oxidative metabolism. To further investigate the proteins involved in the neurological damage, we validated two of the proteins that were most strikingly under-expressed, namely, Syn2 and Dpysl2, which are involved in synaptic function and neurotransmission. We found that Glu2/3 and NR1 receptor subunits were over-expressed in PKU mouse brain. Our results indicate that differential expression of these proteins may be associated with the processes underlying PKU brain dysfunction, namely, decreased synaptic plasticity and impaired neurotransmission.
Collapse
|
33
|
Tonosaki M, Itoh K, Umekage M, Kishimoto T, Yaoi T, Lemmon VP, Fushiki S. L1cam is crucial for cell locomotion and terminal translocation of the Soma in radial migration during murine corticogenesis. PLoS One 2014; 9:e86186. [PMID: 24489698 PMCID: PMC3904877 DOI: 10.1371/journal.pone.0086186] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 12/08/2013] [Indexed: 11/27/2022] Open
Abstract
L1cam (L1) is a cell adhesion molecule associated with a spectrum of human neurological diseases, the most well-known being X-linked hydrocephalus. Although we recently demonstrated that L1 plays an important role in neuronal migration during cortical histogenesis, the mechanisms of delayed migration have still not been clarified. In this study, we found that cell locomotion in the intermediate zone and terminal translocation in the primitive cortical zone (PCZ) were affected by L1-knockdown (L1-KD). Time-lapse analyses revealed that L1-KD neurons produced by in utero electroporation of shRNA targeting L1 (L1-shRNAs) molecules showed decreased locomotion velocity in the intermediate zone, compared with control neurons. Furthermore, L1-KD neurons showed longer and more undulated leading processes during translocation through the primitive cortical zone. The curvature index, a quantitative index for curvilinearity, as well as the length of the leading process, were increased, whereas the somal movement was decreased in L1-KD neurons during terminal translocation in the PCZ. These results suggest that L1 has a role in radial migration of cortical neurons.
Collapse
Affiliation(s)
- Madoka Tonosaki
- Department of Pathology and Applied Neurobiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kyoko Itoh
- Department of Pathology and Applied Neurobiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
- * E-mail:
| | - Masafumi Umekage
- Department of Pathology and Applied Neurobiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tomokazu Kishimoto
- Department of Pathology and Applied Neurobiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Department of Psychiatry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takeshi Yaoi
- Department of Pathology and Applied Neurobiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Vance P. Lemmon
- The Miami Project to Cure Paralysis, University of Miami School of Medicine, Lois Pope LIFE Center, Miami, Florida, United States of America
| | - Shinji Fushiki
- Department of Pathology and Applied Neurobiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
34
|
Zhang Y, Pan YH, Yin Q, Yang T, Dong D, Liao CC, Zhang S. Critical roles of mitochondria in brain activities of torpid Myotis ricketti bats revealed by a proteomic approach. J Proteomics 2014; 105:266-84. [PMID: 24434588 DOI: 10.1016/j.jprot.2014.01.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 12/31/2013] [Accepted: 01/04/2014] [Indexed: 01/21/2023]
Abstract
UNLABELLED Bats are the only mammals that fly and hibernate. Little is known about their overall metabolism in the brain during hibernation. In this study, brain proteins of torpid and active Myotis ricketti bats were fractionated and compared using a proteomic approach. Results showed that 21% (23 proteins) of identified proteins with significant expression changes were associated with amino acid metabolism and proteostasis. The expression levels of proteins involved in energy metabolism (15 proteins), cytoskeletal structure (18 proteins), and stress response (13 proteins) were also significantly altered in torpid bats. Over 30% (34 proteins) of differentially expressed proteins were associated with mitochondrial functions. Various post-translational modifications (PTMs) on PDHB, DLD, and ARG1 were detected, suggesting that bats use PTMs to regulate protein functions during torpor. Antioxidation and stress responses in torpid bats were similar to those of hibernated squirrels, suggesting a common strategy adopted by small hibernators against brain dysfunction. Since many amino acids that metabolize in mitochondria modulate neuronal transmissions, results of this study reveal pivotal roles of mitochondria in neural communication, metabolic regulation, and brain cell survival during bat hibernation. This article is part of a Special Issue entitled: Proteomics of non-model organisms. BIOLOGICAL SIGNIFICANCE This study reveals the mechanisms used by bats to regulate brain activities during torpor. These mechanisms include post-translational modifications and differential expression of proteins involved in mitochondrial electron transport, anaerobic glycolysis, TCA cycle efflux, cytoskeletal plasticity, amino acid metabolism, vesicle structure, antioxidation defense, stress response, and proteostasis. Our study provides insights in metabolic regulation of flying mammals during torpor and common strategies used by small hibernators in response to hibernation. This article is part of a Special Issue entitled: Proteomics of non-model organisms.
Collapse
Affiliation(s)
- Yijian Zhang
- Laboratory of Molecular Ecology and Evolution, Institute for Advanced Studies in Multidisciplinary Science and Technology, East China Normal University, Shanghai 200062, China
| | - Yi-Hsuan Pan
- Laboratory of Molecular Ecology and Evolution, Institute for Advanced Studies in Multidisciplinary Science and Technology, East China Normal University, Shanghai 200062, China
| | - Qiuyuan Yin
- Laboratory of Molecular Ecology and Evolution, Institute for Advanced Studies in Multidisciplinary Science and Technology, East China Normal University, Shanghai 200062, China
| | - Tianxiao Yang
- Laboratory of Molecular Ecology and Evolution, Institute for Advanced Studies in Multidisciplinary Science and Technology, East China Normal University, Shanghai 200062, China
| | - Dong Dong
- Laboratory of Molecular Ecology and Evolution, Institute for Advanced Studies in Multidisciplinary Science and Technology, East China Normal University, Shanghai 200062, China
| | - Chen-Chung Liao
- Proteomic Research Center, National Yang-Ming University, Taipei 11221, Taiwan.
| | - Shuyi Zhang
- Laboratory of Molecular Ecology and Evolution, Institute for Advanced Studies in Multidisciplinary Science and Technology, East China Normal University, Shanghai 200062, China.
| |
Collapse
|
35
|
Ip JPK, Fu AKY, Ip NY. CRMP2: functional roles in neural development and therapeutic potential in neurological diseases. Neuroscientist 2014; 20:589-98. [PMID: 24402611 DOI: 10.1177/1073858413514278] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cytoskeletal restructuring is essential for nearly all cellular processes in the developing brain. After cell fate determination, newborn cortical neurons must migrate to their final positions while establishing proper axon-dendrite polarity. Significant progress has recently been made towards understanding the cellular and molecular mechanisms underlying neuronal polarization in vivo. Collapsin response mediator protein 2 (CRMP2) has long been identified as a microtubule-binding protein that regulates neuronal polarity in vitro. Recent studies provide new insights into the roles of CRMP2 in neuronal migration and subsequent neuronal differentiation. Both the expression and activity of CRMP2 are tightly regulated during cortex development. CRMP2 is suggested to be important in the multipolar-bipolar transition in radial migration. The increasing number of known interaction partners indicates that CRMP2 has functions beyond cytoskeletal regulation, including axonal transport, vesicle trafficking, and neurotransmitter release. This review discusses the current knowledge about CRMP2 in the context of neuronal development and highlights a recent emerging theme regarding its potential therapeutic applications.
Collapse
Affiliation(s)
- Jacque P K Ip
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Amy K Y Fu
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Nancy Y Ip
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, China
| |
Collapse
|
36
|
Yamashita N, Takahashi A, Takao K, Yamamoto T, Kolattukudy P, Miyakawa T, Goshima Y. Mice lacking collapsin response mediator protein 1 manifest hyperactivity, impaired learning and memory, and impaired prepulse inhibition. Front Behav Neurosci 2013; 7:216. [PMID: 24409129 PMCID: PMC3873514 DOI: 10.3389/fnbeh.2013.00216] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 12/16/2013] [Indexed: 11/13/2022] Open
Abstract
Collapsin response mediator protein 1 (CRMP1) is one of the CRMP family members that are involved in various aspects of neuronal development such as axonal guidance and neuronal migration. Here we provide evidence that crmp1 (-/-) mice exhibited behavioral abnormalities related to schizophrenia. The crmp1 (-/-) mice exhibited hyperactivity and/or impaired emotional behavioral phenotype. These mice also exhibited impaired context-dependent memory and long-term memory retention. Furthermore, crmp1 (-/-) mice exhibited decreased prepulse inhibition, and this phenotype was rescued by administration of chlorpromazine, a typical antipsychotic drug. In addition, in vivo microdialysis revealed that the methamphetamine-induced release of dopamine in prefrontal cortex was exaggerated in crmp1 (-/-) mice, suggesting that enhanced mesocortical dopaminergic transmission contributes to their hyperactivity phenotype. These observations suggest that impairment of CRMP1 function may be involved in the pathogenesis of schizophrenia. We propose that crmp1 (-/-) mouse may model endophenotypes present in this neuropsychiatric disorder.
Collapse
Affiliation(s)
- Naoya Yamashita
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine Yokohama, Japan
| | - Aoi Takahashi
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine Yokohama, Japan
| | - Keizo Takao
- Section of Behavior Patterns, Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences Okazaki, Japan ; Genetic Engineering and Functional Genomics Group, Frontier Technology Center, Graduate School of Medicine, Kyoto University Kyoto, Japan ; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency Kawaguchi, Japan
| | - Toshifumi Yamamoto
- Laboratory of Molecular Psychopharmacology, Graduate School of Nanobiosciences, Yokohama City University Yokohama, Japan
| | - Pappachan Kolattukudy
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida Orlando, FL, USA
| | - Tsuyoshi Miyakawa
- Section of Behavior Patterns, Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences Okazaki, Japan ; Genetic Engineering and Functional Genomics Group, Frontier Technology Center, Graduate School of Medicine, Kyoto University Kyoto, Japan ; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency Kawaguchi, Japan ; Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University Toyoake, Japan
| | - Yoshio Goshima
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine Yokohama, Japan
| |
Collapse
|
37
|
Simforoosh N, Basiri A, Shakhssalim N, Shahram G, Tabibi A, Khosdel A, Ziaee SAM. Warm ischemia is not a risk factor for delayed graft function in a living-donor kidney transplant. EXP CLIN TRANSPLANT 2013; 11:575-6. [PMID: 24344951 DOI: 10.6002/ect.2013.0192r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Nasser Simforoosh
- Shahid Labbafinejad Hospital, Urology Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | | | | | | | | |
Collapse
|
38
|
Feldman P, Khanna R. Challenging the catechism of therapeutics for chronic neuropathic pain: Targeting CaV2.2 interactions with CRMP2 peptides. Neurosci Lett 2013; 557 Pt A:27-36. [PMID: 23831344 PMCID: PMC3849117 DOI: 10.1016/j.neulet.2013.06.057] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Revised: 06/24/2013] [Accepted: 06/24/2013] [Indexed: 11/25/2022]
Abstract
Chronic neuropathic pain management is a worldwide concern. Pharmaceutical companies globally have historically targeted ion channels as the therapeutic catechism with many blockbuster successes. Remarkably, no new pain therapeutic has been approved by European or American regulatory agencies over the last decade. This article will provide an overview of an alternative approach to ion channel drug discovery: targeting regulators of ion channels, specifically focusing on voltage-gated calcium channels. We will highlight the discovery of an anti-nociceptive peptide derived from a novel calcium channel interacting partner - the collapsin response mediator protein 2 (CRMP2). In vivo administration of this peptide reduces pain behavior in a number of models of neuropathic pain without affecting sympathetic-associated cardiovascular activity, memory retrieval, sensorimotor function, or depression. A CRMP2-derived peptide analgesic, with restricted access to the CNS, represents a completely novel approach to the treatment of severe pain with an improved safety profile. As peptides now represent one of the fastest growing classes of new drugs, it is expected that peptide targeting of protein interactions within the calcium channel complex may be a paradigm shift in ion channel drug discovery.
Collapse
Affiliation(s)
- Polina Feldman
- Sophia Therapeutics LLC, 351 West 10th Street, Indianapolis, IN 46202, USA
| | - Rajesh Khanna
- Sophia Therapeutics LLC, 351 West 10th Street, Indianapolis, IN 46202, USA
- Department of Pharmacology and Toxicology, 635 Barnhill Drive, Indianapolis, IN 46202, USA
- Department of Biochemistry and Molecular Biology, 635 Barnhill Drive, Indianapolis, IN 46202, USA
- Program in Medical Neurosciences, Paul and Carole Stark Neurosciences Research Institute, 950 West Walnut Street, Indianapolis, IN 46202, USA
| |
Collapse
|
39
|
Collapsin response mediator protein 3 deacetylates histone H4 to mediate nuclear condensation and neuronal death. Sci Rep 2013; 3:1350. [PMID: 23443259 PMCID: PMC3583001 DOI: 10.1038/srep01350] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Accepted: 02/12/2013] [Indexed: 12/29/2022] Open
Abstract
CRMP proteins play critical regulatory roles during semaphorin-mediated neurite outgrowth, neuronal differentiation and death. Albeit having a high degree of structure and sequence resemblance to that of liver dihydropyrimidinase, purified rodent brain CRMPs do not hydrolyze dihydropyrimidinase substrates. Here we found that mouse CRMP3 has robust histone H4 deacetylase activity. During excitotoxicity-induced mouse neuronal death, calpain-cleaved, N-terminally truncated CRMP3 undergoes nuclear translocation to cause nuclear condensation through deacetylation of histone H4. CRMP3-mediated deacetylation of H4 leads to de-repression of the E2F1 gene transcription and E2F1-dependent neuronal death. These studies revealed a novel mechanism of CRMP3 in neuronal death. Together with previous well established bodies of literature that inhibition of histone deacetylase activity provides neuroprotection, we envisage that inhibition of CRMP3 may represent a novel therapeutic approach towards excitotoxicity-induced neuronal death.
Collapse
|
40
|
Quach TT, Wilson SM, Rogemond V, Chounlamountri N, Kolattukudy PE, Martinez S, Khanna M, Belin MF, Khanna R, Honnorat J, Duchemin AM. Mapping CRMP3 domains involved in dendrite morphogenesis and voltage-gated calcium channel regulation. J Cell Sci 2013; 126:4262-73. [PMID: 23868973 DOI: 10.1242/jcs.131409] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Although hippocampal neurons are well-distinguished by the morphological characteristics of their dendrites and their structural plasticity, the mechanisms involved in regulating their neurite initiation, dendrite growth, network formation and remodeling are still largely unknown, in part because the key molecules involved remain elusive. Identifying new dendrite-active cues could uncover unknown molecular mechanisms that would add significant understanding to the field and possibly lead to the development of novel neuroprotective therapy because these neurons are impaired in many neuropsychiatric disorders. In our previous studies, we deleted the gene encoding CRMP3 in mice and identified the protein as a new endogenous signaling molecule that shapes diverse features of the hippocampal pyramidal dendrites without affecting axon morphology. We also found that CRMP3 protects dendrites against dystrophy induced by prion peptide PrP(106-126). Here, we report that CRMP3 has a profound influence on neurite initiation and dendrite growth of hippocampal neurons in vitro. Our deletional mapping revealed that the C-terminus of CRMP3 probably harbors its dendritogenic capacity and supports an active transport mechanism. By contrast, overexpression of the C-terminal truncated CRMP3 phenocopied the effect of CRMP3 gene deletion with inhibition of neurite initiation or decrease in dendrite complexity, depending on the stage of cell development. In addition, this mutant inhibited the activity of CRMP3, in a similar manner to siRNA. Voltage-gated calcium channel inhibitors prevented CRMP3-induced dendritic growth and somatic Ca(2+) influx in CRMP3-overexpressing neurons was augmented largely via L-type channels. These results support a link between CRMP3-mediated Ca(2+) influx and CRMP3-mediated dendritic growth in hippocampal neurons.
Collapse
Affiliation(s)
- Tam T Quach
- Lyon Neuroscience Research Center INSERM U1028/CNRS UMR 5292, F-69372 Lyon, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Iwakura T, Sakoh M, Tsutiya A, Yamashita N, Ohtani A, Tsuda MC, Ogawa S, Tsukahara S, Nishihara M, Shiga T, Goshima Y, Kato T, Ohtani-Kaneko R. Collapsin response mediator protein 4 affects the number of tyrosine hydroxylase-immunoreactive neurons in the sexually dimorphic nucleus in female mice. Dev Neurobiol 2013; 73:502-17. [PMID: 23420586 DOI: 10.1002/dneu.22076] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 12/25/2012] [Accepted: 02/12/2013] [Indexed: 01/04/2023]
Abstract
In the sexually dimorphic anteroventral periventricular nucleus (AVPV) of the hypothalamus, females have a greater number of tyrosine hydroxylase-immunoreactive (TH-ir) and kisspeptin-immunoreactive (kisspeptin-ir) neurons than males. In this study, we used proteomics analysis and gene-deficient mice to identify proteins that regulate the number of TH-ir and kisspeptin-ir neurons in the AVPV. Analysis of protein expressions in the rat AVPV on postnatal day 1 (PD1; the early phase of sex differentiation) using two-dimensional fluorescence difference gel electrophoresis followed by MALDI-TOF-MS identified collapsin response mediator protein 4 (CRMP4) as a protein exhibiting sexually dimorphic expression. Interestingly, this sexually differential expressions of CRMP4 protein and mRNA in the AVPV was not detected on PD6. Prenatal testosterone exposure canceled the sexual difference in the expression of Crmp4 mRNA in the rat AVPV. Next, we used CRMP4-knockout (CRMP4-KO) mice to determine the in vivo function of CRMP4 in the AVPV. Crmp4 knockout did not change the number of kisspeptin-ir neurons in the adult AVPV in either sex. However, the number of TH-ir neurons was increased in the AVPV of adult female CRMP4-KO mice as compared with the adult female wild-type mice. During development, no significant difference in the number of TH-ir neurons was detected between sexes or genotypes on embryonic day 15, but a female-specific increase in TH-ir neurons was observed in CRMP4-KO mice on PD1, when the sex difference was not yet apparent in wild-type mice. These results indicate that CRMP4 regulates the number of TH-ir cell number in the female AVPV.
Collapse
Affiliation(s)
- Takashi Iwakura
- Doctoral Program in Kansei, Behavioral and Brain Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki, 305-8577, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Yasukawa M, Ishida K, Yuge Y, Hanaoka M, Minami Y, Ogawa M, Sasaki T, Saito M, Tsuji T. Dpysl4 is involved in tooth germ morphogenesis through growth regulation, polarization and differentiation of dental epithelial cells. Int J Biol Sci 2013; 9:382-90. [PMID: 23630450 PMCID: PMC3638293 DOI: 10.7150/ijbs.5510] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Accepted: 04/09/2013] [Indexed: 11/05/2022] Open
Abstract
Dihydropyrimidinase-related protein 4 (Dpysl4) is a known regulator of hippocampal neuron development. Here, we report that Dpysl4 is involved in growth regulation, polarization and differentiation of dental epithelial cells during tooth germ morphogenesis. A reduction in Dpysl4 gene expression in the tooth germ produced a loss of ameloblasts, resulting in the decrease of synthesis and secretion of enamel. The inhibition of Dpysl4 gene expression led to promotion of cell proliferation of inner enamel epithelial cells and inhibition of the differentiation of these cells into pre-ameloblasts, which was confirmed by analyzing cell polarization, columnar cell structure formation and the expression of ameloblast marker genes. By contrast, overexpression of Dpysl4 in dental epithelial cells induces inhibition of growth and increases the expression of the inner enamel epithelial cell marker gene, Msx2. These findings suggest that Dpysl4 plays essential roles in tooth germ morphogenesis through the regulation of dental epithelial cell proliferation, cell polarization and differentiation.
Collapse
Affiliation(s)
- Masato Yasukawa
- Department of Biological Science and Technology, Graduate school of Industrial Science and Technology, Tokyo University of Science, Chiba 278-8510, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Niisato E, Nagai J, Yamashita N, Abe T, Kiyonari H, Goshima Y, Ohshima T. CRMP4 suppresses apical dendrite bifurcation of CA1 pyramidal neurons in the mouse hippocampus. Dev Neurobiol 2012; 72:1447-57. [PMID: 22234963 DOI: 10.1002/dneu.22007] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Revised: 12/25/2011] [Accepted: 12/29/2011] [Indexed: 02/03/2023]
Abstract
Collapsin response mediator proteins (CRMPs) are a family of cytosolic phosphoproteins that consist of 5 members (CRMP 1-5). CRMP2 and CRMP4 regulate neurite outgrowth by binding to tubulin heterodimers, resulting in the assembly of microtubules. CRMP2 also mediates the growth cone collapse response to the repulsive guidance molecule semaphorin-3A (Sema3A). However, the role of CRMP4 in Sema3A signaling and its function in the developing mouse brain remain unclear. We generated CRMP4-/- mice in order to study the in vivo function of CRMP4 and identified a phenotype of proximal bifurcation of apical dendrites in the CA1 pyramidal neurons of CRMP4-/- mice. We also observed increased dendritic branching in cultured CRMP4-/- hippocampal neurons as well as in cultured cortical neurons treated with CRMP4 shRNA. Sema3A induces extension and branching of the dendrites of hippocampal neurons; however, these inductions were compromised in the CRMP4-/- hippocampal neurons. These results suggest that CRMP4 suppresses apical dendrite bifurcation of CA1 pyramidal neurons in the mouse hippocampus and that this is partly dependent on Sema3A signaling.
Collapse
Affiliation(s)
- Emi Niisato
- Department of Life Science and Medical Bioscience, Waseda University, Tokyo 162-8480, Japan
| | | | | | | | | | | | | |
Collapse
|
44
|
Aberrant expression of collapsin response mediator proteins‐1, ‐2 and ‐5 in the brain of intrauterine growth restricted rats. Int J Dev Neurosci 2012; 31:53-60. [DOI: 10.1016/j.ijdevneu.2012.10.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Revised: 09/15/2012] [Accepted: 10/08/2012] [Indexed: 12/29/2022] Open
|
45
|
Tsutiya A, Ohtani-Kaneko R. Postnatal alteration of collapsin response mediator protein 4 mRNA expression in the mouse brain. J Anat 2012; 221:341-51. [PMID: 22816653 PMCID: PMC3458253 DOI: 10.1111/j.1469-7580.2012.01544.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2012] [Indexed: 11/30/2022] Open
Abstract
Collapsin response mediator protein 4 (CRMP4) is a molecular marker for immature neurons but only limited information is available on the spatiotemporal gene expression changes of Crmp4 in the developing rodent. In the present study, the variation of CRMP4 mRNA expression in the mouse brain was investigated from postnatal day (PD) 0 (the day of birth) to adulthood by in situ hybridization. The hybridization signals were broadly detected on PD0 and regional changes in expression during development were noted. Expression patterns of CRMP4 mRNA were classified into the following three types: (i) signals that were strongest on PD0 or PD7, weak or undetectable on PD14, and absent in adulthood: this pattern was observed in most brain areas; (ii) signals that were first detected on PD0 or PD7 and persisted into adulthood: this pattern was seen in the dentate gyrus and subventricular zone of the olfactory bulb (OB); and (iii) signals that were strongest on PD0 and decreased gradually with age but were still detectable in adulthood: this pattern was identified for the first time in the mitral cell layer of the OB. Analysis using quantitative real-time RT-PCR confirmed higher expression of CRMP4 mRNA in the OB than in other adult brain regions. The persistence of CRMP4 mRNA in the adult OB, including the mitral cell layer, suggests the possibility of both neurogenetic and non-neurogenetic functional roles of CRMP4 in this region.
Collapse
Affiliation(s)
- Atsuhiro Tsutiya
- Graduate School of Life Sciences, Toyo UniversityOura, Gunnma, Japan
| | - Ritsuko Ohtani-Kaneko
- Graduate School of Life Sciences, Toyo UniversityOura, Gunnma, Japan
- Bio-Nano Electronic Research Center, Toyo UniversityKawagoe, Saitama, Japan
| |
Collapse
|
46
|
Chen Y, Sheng H, Xu Y, Zhang Y, Ni X. Activation of CRHR2 exerts an inhibitory effect on the expression of collapsin response mediator protein 3 in hippocampal neurons. Neuropeptides 2012; 46:93-8. [PMID: 22245585 DOI: 10.1016/j.npep.2011.12.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Revised: 12/20/2011] [Accepted: 12/21/2011] [Indexed: 12/19/2022]
Abstract
Corticotropin-releasing hormone (CRH) family peptides as well as their receptors have been shown to exhibit various functions in hippocampus. However, effects of CRH receptors activation on collapsin response mediator protein 3 (CRMP3), the key protein for dendrite outgrowth and cell apoptosis, remain unclear. In the present study, we determined the effects of CRHR1 and CRHR2 on CRMP3 expression in cultured hippocampal neurons. CRH and urocortin II (UCNII) dose-dependently suppressed CRMP3 mRNA and protein expression. The inhibitory effect on CRMP3 expression was completely reversed by CRHR2 antagonist but not by CRHR1 antagonist. Investigations on the signaling pathways of UCNII showed that CRHR2 mediated UCNII-induced increase in phosphorylated phospholipase C (PLC)-β3 expression. Blocking PLC activity with U73122 and PKC with Gö6976 completely prevented UCNII-inhibited CRMP3 expression. Our results suggest that CRHR2 activation decrease CRMP3 expression in hippocampal neurons via a mechanism that is dependent on PLC/PKC signaling pathways.
Collapse
Affiliation(s)
- Yanming Chen
- Department of Physiology and The Key Laboratory of Molecular Neurobiology of Ministry of Education, Second Military Medical University, Shanghai 200433, PR China
| | | | | | | | | |
Collapse
|
47
|
Collapsin response mediator proteins regulate neuronal development and plasticity by switching their phosphorylation status. Mol Neurobiol 2012; 45:234-46. [PMID: 22351471 DOI: 10.1007/s12035-012-8242-4] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Accepted: 01/29/2012] [Indexed: 12/21/2022]
Abstract
Collapsin response mediator protein (CRMP) was originally identified as a molecule involved in semaphorin3A signaling. CRMPs are now known to consist of five homologous cytosolic proteins, CRMP1-5. All of them are phosphorylated and highly expressed in the developing and adult nervous system. In vitro experiments have clearly demonstrated that CRMPs play important roles in neuronal development and maturation through the regulation of their phosphorylation. Several recent knockout mice studies have revealed in vivo roles of CRMPs in neuronal migration, neuronal network formation, synapse formation, synaptic plasticity, and neuronal diseases. Dynamic spatiotemporal regulation of phosphorylation status of CRMPs is involved in many aspects of neuronal development.
Collapse
|
48
|
Lin PC, Chan PM, Hall C, Manser E. Collapsin response mediator proteins (CRMPs) are a new class of microtubule-associated protein (MAP) that selectively interacts with assembled microtubules via a taxol-sensitive binding interaction. J Biol Chem 2011; 286:41466-41478. [PMID: 21953449 DOI: 10.1074/jbc.m111.283580] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Collapsin response mediator proteins are ubiquitously expressed from multiple genes (CRMPs 1-5) and play important roles in dividing cells and during semaphorin 3A (Sema3A) signaling. Nonetheless, their mode of action remains opaque. Here we carried out in vivo and in vitro assays that demonstrate that CRMPs are a new class of microtubule-associated protein (MAP). In experiments with CRMP1 or CRMP2 and their derivatives, only the C-terminal region (residues 490-572) mediated microtubule binding. The in vivo microtubule association of CRMPs was abolished by taxol or epothilone B, which is highly unusual. CRMP2-depleted cells exhibited destabilized anaphase astral microtubules and altered spindle position. In a cell-based assay, all CRMPs stabilized interphase microtubules against nocodazole-mediated depolymerization, with CRMP1 being the most potent. Remarkably, a 82-residue C-terminal region of CRMP1 or CRMP2, unrelated to other microtubule binding motifs, is sufficient to stabilize microtubules. In cells, we demonstrate that glycogen synthase kinase-3β (GSK3β) inhibition potentiates this activity. Thus, CRMPs are a new class of MAP that binds through a unique motif, but in common with others such as Tau, is antagonized by GSK3β. This regulation is consistent with such kinases being critical for the Sema3A (collapsin) pathway. These findings have implications for cancer and neurodegeneration.
Collapse
Affiliation(s)
- Pao-Chun Lin
- Institute of Medical Biology, 8A Biomedical Grove, Singapore 138648
| | - Perry M Chan
- Small G-protein Signaling and Kinases (sGSK-NRP) Group, Neuroscience Research Partnership, 61 Biopolis Drive, Singapore 138673
| | - Christine Hall
- Institute of Neurology, University College London, 1 Wakefield Street, London WC1N 1PJ, United Kingdom
| | - Ed Manser
- Small G-protein Signaling and Kinases (sGSK-NRP) Group, Neuroscience Research Partnership, 61 Biopolis Drive, Singapore 138673; Institute of Medical Biology, 8A Biomedical Grove, Singapore 138648.
| |
Collapse
|
49
|
Quach TT, Wang Y, Khanna R, Chounlamountri N, Auvergnon N, Honnorat J, Duchemin AM. Effect of CRMP3 expression on dystrophic dendrites of hippocampal neurons. Mol Psychiatry 2011; 16:689-91. [PMID: 21339751 DOI: 10.1038/mp.2011.6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
50
|
CRMP5 (collapsin response mediator protein 5) regulates dendritic development and synaptic plasticity in the cerebellar Purkinje cells. J Neurosci 2011; 31:1773-9. [PMID: 21289187 DOI: 10.1523/jneurosci.5337-10.2011] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Collapsin response mediator protein 5 (CRMP5) is one of the CRMP members that expresses abundantly in the developing brain. To examine the in vivo function of CRMP5, we generated crmp5-deficient (crmp5(-/-)) mice. Anti-calbindin immunofluorescence studies of crmp5(-/-) mice revealed aberrant dendrite morphology; specifically, a decrease in the size of soma and diameter of primary dendrite of the cerebellar Purkinje cells at postnatal day 21 (P21) and P28, but not at P14. Coincidentally, CRMP5 is detected in Purkinje cells at P21 and P28 from crmp5(+/-) mice. In cerebellar slices of crmp5(-/-) mice, the induction of long-term depression of excitatory synaptic transmission between parallel fibers and Purkinje cells was deficient. Given that brain-derived neurotrophic factor (BDNF) plays major roles in dendritic development, we tried to elucidate the possible roles of CRMP5 in BDNF signaling. The effect of BDNF to induce dendritic branching was markedly attenuated in cultured crmp5(-/-) neurons. Furthermore, CRMP5 was tyrosine phosphorylated when coexpressed with neurotrophic tyrosine kinase receptor type 2 (TrkB), a receptor for BDNF, in HEK293T cells. These findings suggest that CRMP5 is involved in the development, maintenance and synaptic plasticity of Purkinje cells.
Collapse
|