1
|
Xiao H, Xu Y, Cui S, Wang JH. Neuroligin-3-Mediated Synapse Formation Strengthens Interactions between Hippocampus and Barrel Cortex in Associative Memory. Int J Mol Sci 2024; 25:711. [PMID: 38255783 PMCID: PMC10815421 DOI: 10.3390/ijms25020711] [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: 11/28/2023] [Revised: 12/16/2023] [Accepted: 12/17/2023] [Indexed: 01/24/2024] Open
Abstract
Memory traces are believed to be broadly allocated in cerebral cortices and the hippocampus. Mutual synapse innervations among these brain areas are presumably formed in associative memory. In the present study, we have used neuronal tracing by pAAV-carried fluorescent proteins and neuroligin-3 mRNA knockdown by shRNAs to examine the role of neuroligin-3-mediated synapse formation in the interconnection between primary associative memory cells in the sensory cortices and secondary associative memory cells in the hippocampus during the acquisition and memory of associated signals. Our studies show that mutual synapse innervations between the barrel cortex and the hippocampal CA3 region emerge and are upregulated after the memories of associated whisker and odor signals come into view. These synapse interconnections are downregulated by a knockdown of neuroligin-3-mediated synapse linkages. New synapse interconnections and the strengthening of these interconnections appear to endorse the belief in an interaction between the hippocampus and sensory cortices for memory consolidation.
Collapse
Affiliation(s)
- Huajuan Xiao
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing 100049, China;
| | - Yang Xu
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China;
| | - Shan Cui
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China;
| | - Jin-Hui Wang
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China;
| |
Collapse
|
2
|
Notaras M, Lodhi A, Fang H, Greening D, Colak D. The proteomic architecture of schizophrenia iPSC-derived cerebral organoids reveals alterations in GWAS and neuronal development factors. Transl Psychiatry 2021; 11:541. [PMID: 34667143 PMCID: PMC8526592 DOI: 10.1038/s41398-021-01664-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/24/2021] [Accepted: 09/30/2021] [Indexed: 12/21/2022] Open
Abstract
Schizophrenia (Scz) is a brain disorder that has a typical onset in early adulthood but otherwise maintains unknown disease origins. Unfortunately, little progress has been made in understanding the molecular mechanisms underlying neurodevelopment of Scz due to ethical and technical limitations in accessing developing human brain tissue. To overcome this challenge, we have previously utilized patient-derived Induced Pluripotent Stem Cells (iPSCs) to generate self-developing, self-maturating, and self-organizing 3D brain-like tissue known as cerebral organoids. As a continuation of this prior work, here we provide an architectural map of the developing Scz organoid proteome. Utilizing iPSCs from n = 25 human donors (n = 8 healthy Ctrl donors, and n = 17 Scz patients), we generated 3D cerebral organoids, employed 16-plex isobaric sample-barcoding chemistry, and simultaneously subjected samples to comprehensive high-throughput liquid-chromatography/mass-spectrometry (LC/MS) quantitative proteomics. Of 3,705 proteins identified by high-throughput proteomic profiling, we identified that just ~2.62% of the organoid global proteomic landscape was differentially regulated in Scz organoids. In sum, just 43 proteins were up-regulated and 54 were down-regulated in Scz patient-derived organoids. Notably, a range of neuronal factors were depleted in Scz organoids (e.g., MAP2, TUBB3, SV2A, GAP43, CRABP1, NCAM1 etc.). Based on global enrichment analysis, alterations in key pathways that regulate nervous system development (e.g., axonogenesis, axon development, axon guidance, morphogenesis pathways regulating neuronal differentiation, as well as substantia nigra development) were perturbed in Scz patient-derived organoids. We also identified prominent alterations in two novel GWAS factors, Pleiotrophin (PTN) and Podocalyxin (PODXL), in Scz organoids. In sum, this work serves as both a report and a resource that researchers can leverage to compare, contrast, or orthogonally validate Scz factors and pathways identified in observational clinical studies and other model systems.
Collapse
Affiliation(s)
- Michael Notaras
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Aiman Lodhi
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Haoyun Fang
- Baker Institute for Heart and Diabetes, Melbourne, VIC, Australia
| | - David Greening
- Baker Institute for Heart and Diabetes, Melbourne, VIC, Australia.
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia.
- Central Clinical School, Monash University, Melbourne, VIC, Australia.
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia.
| | - Dilek Colak
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, Cornell University, New York, NY, USA.
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medical College, Cornell University, New York, NY, USA.
| |
Collapse
|
3
|
Positive association between PTN polymorphisms and schizophrenia in Northeast Chinese Han population. Psychiatr Genet 2020; 30:141-149. [PMID: 32868733 DOI: 10.1097/ypg.0000000000000262] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
As a severely and highly heritable psychotic disorder, schizophrenia has become a serious public health problem in modern society. Pleiotrophin (PTN) is a secreted cell cytokine associated with the extracellular matrix and acts as a growth factor. PTN is mainly expressed in neuroectodermal and mesodermal tissues, indicating its effect in neuron migration and epithelium-mesenchyme interactions. Whereas PTN is associated with some neurodegenerative diseases and has modulating effects on them. In this study, we aimed to investigate the association between PTN polymorphisms and schizophrenia in an independent case-control sample-set including 738 schizophrenia patients and 1085 healthy controls. Of the 13 selected single nucleotide polymorphisms (SNPs), five showed significant differences in allele or/and genotype frequencies between patients and controls: rs3959914 (genotype: χ = 11.5217, P = 0.0032); rs11765480 (genotype: χ = 10.6620, P = 0.0049); rs1473355 (genotype: χ = 8.3902, P = 0.0151); rs322246 (allele: χ = 5.5954, P = 0.0180); rs322240 (genotype: χ = 8.8429, P = 0.0121; allele: χ = 8.7802, P = 0.0031). The haplotype analysis of the selected SNPs showed different haplotype frequencies for one block (rs322240, rs322246) between cases and controls (global: χ = 9.0290, P = 0.0110; A-G: χ = 8.985, P = 0.0027; C-A: χ = 5.814, P = 0.0159). Our present results indicate PTN as a susceptibility gene for schizophrenia.
Collapse
|
4
|
Chang X, Lima LDA, Liu Y, Li J, Li Q, Sleiman PMA, Hakonarson H. Common and Rare Genetic Risk Factors Converge in Protein Interaction Networks Underlying Schizophrenia. Front Genet 2018; 9:434. [PMID: 30323833 PMCID: PMC6172705 DOI: 10.3389/fgene.2018.00434] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Accepted: 09/12/2018] [Indexed: 11/25/2022] Open
Abstract
Hundreds of genomic loci have been identified with the recent advances of schizophrenia in genome-wide association studies (GWAS) and sequencing studies. However, the functional interactions among those genes remain largely unknown. We developed a network-based approach to integrate multiple genetic risk factors, which lead to the discovery of new susceptibility genes and causal sub-networks, or pathways in schizophrenia. We identified significantly and consistently over-represented pathways in the largest schizophrenia GWA studies, which are highly relevant to synaptic plasticity, neural development and signaling transduction, such as long-term potentiation, neurotrophin signaling pathway, and the ERBB signaling pathway. We also demonstrated that genes targeted by common SNPs are more likely to interact with genes harboring de novo mutations (DNMs) in the protein-protein interaction (PPI) network, suggesting a mutual interplay of both common and rare variants in schizophrenia. We further developed an edge-based search algorithm to identify the top-ranked gene modules associated with schizophrenia risk. Our results suggest that the N-methyl-D-aspartate receptor (NMDAR) interactome may play a leading role in the pathology of schizophrenia, as it is highly targeted by multiple types of genetic risk factors.
Collapse
Affiliation(s)
- Xiao Chang
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Leandro de Araujo Lima
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Yichuan Liu
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Jin Li
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, United States.,Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Qingqin Li
- Janssen Research & Development, LLC, Titusville, NJ, United States
| | - Patrick M A Sleiman
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, United States.,Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Hakon Hakonarson
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, United States.,Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| |
Collapse
|
5
|
Fujikawa A, Chow JPH, Matsumoto M, Suzuki R, Kuboyama K, Yamamoto N, Noda M. Identification of novel splicing variants of protein tyrosine phosphatase receptor type Z. J Biochem 2017; 162:381-390. [PMID: 28992190 DOI: 10.1093/jb/mvx042] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 06/14/2017] [Indexed: 11/13/2022] Open
Abstract
Protein tyrosine phosphatase receptor type Z (PTPRZ, also known as PTPζ or RPTPβ) is preferentially expressed in the central nervous system (CNS). PTPRZ plays important roles during development and adulthood in CNS myelination, learning and memory. Three splicing isoforms for PTPRZ have been identified to date: two receptor type isoforms, PTPRZ-A and PTPRZ-B, and one secretory isoform, PTPRZ-S. We herein identified novel PTPRZ receptor sub-isoforms without a seven-amino acid sequence encoded by exon 16. This sequence forms a part of the helix-turn-helix segment called the 'wedge' structure, which is located at the N-terminal region in the membrane-proximal protein tyrosine phosphatase domain. In contrast to conventional receptor isoforms with uniform expression, the deleted isoforms were expressed in the brain, but not in the retina, indicating the tissue-specific splicing of exon 16. Biochemical analyses of PTPRZ intracellular regions revealed differences in the characteristics of the deleted form, namely, stronger binding activity to postsynaptic density protein 95 (PSD95) and greater enrichment in the postsynaptic density fraction than the full-length form. Furthermore, the exon 16-deleted form exhibited higher catalytic efficiency in vitro. These results suggest that sub-isoforms of PTPRZ have different functions because of variations in the wedge structure.
Collapse
Affiliation(s)
- Akihiro Fujikawa
- Division of Molecular Neurobiology, National Institute for Basic Biology
| | - Jeremy Pak Hong Chow
- Division of Molecular Neurobiology, National Institute for Basic Biology.,School of Life Science, Graduate University for Advanced Studies, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan
| | - Masahito Matsumoto
- Division of Molecular Neurobiology, National Institute for Basic Biology
| | - Ryoko Suzuki
- Division of Molecular Neurobiology, National Institute for Basic Biology
| | - Kazuya Kuboyama
- Division of Molecular Neurobiology, National Institute for Basic Biology
| | - Naoki Yamamoto
- Laboratory of Molecular Biology & Histochemistry, Fujita Health University, 1-98, Toyoake, Aichi 470-1192, Japan
| | - Masaharu Noda
- Division of Molecular Neurobiology, National Institute for Basic Biology.,School of Life Science, Graduate University for Advanced Studies, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan
| |
Collapse
|
6
|
Qin EY, Cooper DD, Abbott KL, Lennon J, Nagaraja S, Mackay A, Jones C, Vogel H, Jackson PK, Monje M. Neural Precursor-Derived Pleiotrophin Mediates Subventricular Zone Invasion by Glioma. Cell 2017; 170:845-859.e19. [PMID: 28823557 PMCID: PMC5587159 DOI: 10.1016/j.cell.2017.07.016] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Revised: 06/15/2017] [Accepted: 07/13/2017] [Indexed: 12/26/2022]
Abstract
The lateral ventricle subventricular zone (SVZ) is a frequent and consequential site of pediatric and adult glioma spread, but the cellular and molecular mechanisms mediating this are poorly understood. We demonstrate that neural precursor cell (NPC):glioma cell communication underpins this propensity of glioma to colonize the SVZ through secretion of chemoattractant signals toward which glioma cells home. Biochemical, proteomic, and functional analyses of SVZ NPC-secreted factors revealed the neurite outgrowth-promoting factor pleiotrophin, along with required binding partners SPARC/SPARCL1 and HSP90B, as key mediators of this chemoattractant effect. Pleiotrophin expression is strongly enriched in the SVZ, and pleiotrophin knock down starkly reduced glioma invasion of the SVZ in the murine brain. Pleiotrophin, in complex with the binding partners, activated glioma Rho/ROCK signaling, and ROCK inhibition decreased invasion toward SVZ NPC-secreted factors. These findings demonstrate a pathogenic role for NPC:glioma interactions and potential therapeutic targets to limit glioma invasion. PAPERCLIP.
Collapse
Affiliation(s)
- Elizabeth Y Qin
- Department of Neurology, Stanford University, Palo Alto, CA 94305, USA
| | | | - Keene L Abbott
- Baxter Laboratory, Department of Microbiology & Immunology, Stanford University, Palo Alto, CA 94305, USA
| | - James Lennon
- Department of Neurology, Stanford University, Palo Alto, CA 94305, USA
| | - Surya Nagaraja
- Department of Neurology, Stanford University, Palo Alto, CA 94305, USA
| | - Alan Mackay
- Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London SM2 5NG, UK
| | - Chris Jones
- Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London SM2 5NG, UK
| | - Hannes Vogel
- Department of Neurology, Stanford University, Palo Alto, CA 94305, USA; Department of Pathology, Stanford University, Palo Alto, CA 94305, USA; Department of Neurosurgery, Stanford University, Palo Alto, CA 94305, USA; Department of Pediatrics, Stanford University, Palo Alto, CA 94305, USA
| | - Peter K Jackson
- Baxter Laboratory, Department of Microbiology & Immunology, Stanford University, Palo Alto, CA 94305, USA; Department of Pathology, Stanford University, Palo Alto, CA 94305, USA
| | - Michelle Monje
- Department of Neurology, Stanford University, Palo Alto, CA 94305, USA; Department of Pathology, Stanford University, Palo Alto, CA 94305, USA; Department of Neurosurgery, Stanford University, Palo Alto, CA 94305, USA; Department of Pediatrics, Stanford University, Palo Alto, CA 94305, USA.
| |
Collapse
|
7
|
Minge D, Senkov O, Kaushik R, Herde MK, Tikhobrazova O, Wulff AB, Mironov A, van Kuppevelt TH, Oosterhof A, Kochlamazashvili G, Dityatev A, Henneberger C. Heparan Sulfates Support Pyramidal Cell Excitability, Synaptic Plasticity, and Context Discrimination. Cereb Cortex 2017; 27:903-918. [PMID: 28119345 PMCID: PMC5390399 DOI: 10.1093/cercor/bhx003] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 12/04/2016] [Accepted: 01/04/2017] [Indexed: 02/06/2023] Open
Abstract
Heparan sulfate (HS) proteoglycans represent a major component of the extracellular matrix and are critical for brain development. However, their function in the mature brain remains to be characterized. Here, acute enzymatic digestion of HS side chains was used to uncover how HSs support hippocampal function in vitro and in vivo. We found that long-term potentiation (LTP) of synaptic transmission at CA3-CA1 Schaffer collateral synapses was impaired after removal of highly sulfated HSs with heparinase 1. This reduction was associated with decreased Ca2+ influx during LTP induction, which was the consequence of a reduced excitability of CA1 pyramidal neurons. At the subcellular level, heparinase treatment resulted in reorganization of the distal axon initial segment, as detected by a reduction in ankyrin G expression. In vivo, digestion of HSs impaired context discrimination in a fear conditioning paradigm and oscillatory network activity in the low theta band after fear conditioning. Thus, HSs maintain neuronal excitability and, as a consequence, support synaptic plasticity and learning.
Collapse
Affiliation(s)
- Daniel Minge
- Institute of Cellular Neurosciences, University of Bonn Medical School, 53105 Bonn, Germany
| | - Oleg Senkov
- German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
| | - Rahul Kaushik
- German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
| | - Michel K. Herde
- Institute of Cellular Neurosciences, University of Bonn Medical School, 53105 Bonn, Germany
| | - Olga Tikhobrazova
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, 16163 Genova, Italy
- Department of Neurotechnology, Lobachevsky State University of Nizhny Novgorod, 603950 Nizhny Novgorod, Russia
| | - Andreas B. Wulff
- Institute of Cellular Neurosciences, University of Bonn Medical School, 53105 Bonn, Germany
| | - Andrey Mironov
- Department of Neurotechnology, Lobachevsky State University of Nizhny Novgorod, 603950 Nizhny Novgorod, Russia
- Central Research Laboratory, Nizhny Novgorod State Medical Academy, 603005 Nizhny Novgorod, Russia
| | - Toin H. van Kuppevelt
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Arie Oosterhof
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Gaga Kochlamazashvili
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, 16163 Genova, Italy
- Department of Molecular Pharmacology and Cell Biology, Leibniz-Institut für Molekulare Pharmakologie (FMP), 13125 Berlin, Germany
| | - Alexander Dityatev
- German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, 16163 Genova, Italy
- Department of Neurotechnology, Lobachevsky State University of Nizhny Novgorod, 603950 Nizhny Novgorod, Russia
- Medizinische Fakultät, Otto-von-Guericke-Universität Magdeburg, 39120 Magdeburg, Germany
| | - Christian Henneberger
- Institute of Cellular Neurosciences, University of Bonn Medical School, 53105 Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), 53175 Bonn, Germany
- Institute of Neurology, University College London, London WC1N 3BG, UK
| |
Collapse
|
8
|
Rauvala H, Paveliev M, Kuja-Panula J, Kulesskaya N. Inhibition and enhancement of neural regeneration by chondroitin sulfate proteoglycans. Neural Regen Res 2017; 12:687-691. [PMID: 28616017 PMCID: PMC5461598 DOI: 10.4103/1673-5374.206630] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The current dogma in neural regeneration research implies that chondroitin sulfate proteoglycans (CSPGs) inhibit plasticity and regeneration in the adult central nervous system (CNS). We argue that the role of the CSPGs can be reversed from inhibition to activation by developmentally expressed CSPG-binding factors. Heparin-binding growth-associated molecule (HB-GAM; also designated as pleiotrophin) has been studied as a candidate molecule that might modulate the role of CSPG matrices in plasticity and regeneration. Studies in vitro show that in the presence of soluble HB-GAM chondroitin sulfate (CS) chains of CSPGs display an enhancing effect on neurite outgrowth. Based on the in vitro studies, we suggest a model according to which the HB-GAM/CS complex binds to the neuron surface receptor glypican-2, which induces neurite growth. Furthermore, HB-GAM masks the CS binding sites of the neurite outgrowth inhibiting receptor protein tyrosine phosphatase sigma (PTPσ), which may contribute to the HB-GAM-induced regenerative effect. In vivo studies using two-photon imaging after local HB-GAM injection into prick-injury of the cerebral cortex reveal regeneration of dendrites that has not been previously demonstrated after injuries of the mammalian nervous system. In the spinal cord, two-photon imaging displays HB-GAM-induced axonal regeneration. Studies on the HB-GAM/CS mechanism in vitro and in vivo are expected to pave the way for drug development for injuries of brain and spinal cord.
Collapse
Affiliation(s)
- Heikki Rauvala
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | | | | | - Natalia Kulesskaya
- Neuroscience Center, University of Helsinki, Helsinki, Finland.,Department of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| |
Collapse
|
9
|
Lizen B, Hutlet B, Bissen D, Sauvegarde D, Hermant M, Ahn MT, Gofflot F. HOXA5 localization in postnatal and adult mouse brain is suggestive of regulatory roles in postmitotic neurons. J Comp Neurol 2016; 525:1155-1175. [PMID: 27650319 DOI: 10.1002/cne.24123] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 09/08/2016] [Accepted: 09/15/2016] [Indexed: 01/13/2023]
Abstract
Hoxa5 is a member of the Hox gene family, which plays critical roles in successive steps of the central nervous system formation during embryonic and fetal development. Hoxa5 expression in the adult mouse brain has been reported, suggesting that this gene may be functionally required in the brain after birth. To provide further insight into the Hoxa5 expression pattern and potential functions in the brain, we have characterized its neuroanatomical profile from embryonic stages to adulthood. While most Hox mapping studies have been based solely on transcript analysis, we extended our analysis to HOXA5 protein localization in adulthood using specific antibodies. Our results show that Hoxa5 expression appears in the most caudal part of the hindbrain at fetal stages, where it is maintained until adulthood. In the medulla oblongata and pons, we detected Hoxa5 expression in many precerebellar neurons and in several nuclei implicated in the control of autonomic functions. In these territories, the HOXA5 protein is present solely in neurons, specifically in γ-aminobutyric acid (GABA)ergic, glutamatergic, and catecholaminergic neurons. Finally, we also detected Hoxa5 transcripts, but not the HOXA5 protein, in the thalamus and the cortex, from postnatal stages to adult stages, and in the cerebellum at adulthood. We provide evidence that some larger variants of Hoxa5 transcripts are present in these territories. Our mapping analysis allowed us to build hypotheses regarding HOXA5 functions in the nervous system after birth, such as a potential role in the establishment and refinement/plasticity of precerebellar circuits during postnatal and adult life. J. Comp. Neurol. 525:1155-1175, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Benoit Lizen
- Institute of Life Sciences, Catholic University of Louvain, 1348, Louvain-la-Neuve, Belgium
| | - Bertrand Hutlet
- Institute of Life Sciences, Catholic University of Louvain, 1348, Louvain-la-Neuve, Belgium
| | - Diane Bissen
- Institute of Life Sciences, Catholic University of Louvain, 1348, Louvain-la-Neuve, Belgium
| | - Deborah Sauvegarde
- Institute of Life Sciences, Catholic University of Louvain, 1348, Louvain-la-Neuve, Belgium
| | - Maryse Hermant
- Institute of Life Sciences, Catholic University of Louvain, 1348, Louvain-la-Neuve, Belgium
| | - Marie-Thérèse Ahn
- Institute of Life Sciences, Catholic University of Louvain, 1348, Louvain-la-Neuve, Belgium
| | - Françoise Gofflot
- Institute of Life Sciences, Catholic University of Louvain, 1348, Louvain-la-Neuve, Belgium
| |
Collapse
|
10
|
González-Castillo C, Ortuño-Sahagún D, Guzmán-Brambila C, Márquez-Aguirre AL, Raisman-Vozari R, Pallás M, Rojas-Mayorquín AE. The absence of pleiotrophin modulates gene expression in the hippocampus in vivo and in cerebellar granule cells in vitro. Mol Cell Neurosci 2016; 75:113-21. [PMID: 27468976 DOI: 10.1016/j.mcn.2016.07.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 07/04/2016] [Accepted: 07/25/2016] [Indexed: 12/28/2022] Open
Abstract
Pleiotrophin (PTN) is a secreted growth factor recently proposed to act as a neuromodulatory peptide in the Central Nervous System. PTN appears to be involved in neurodegenerative diseases and neural disorders, and it has also been implicated in learning and memory. Specifically, PTN-deficient mice exhibit a lower threshold for LTP induction in the hippocampus, which is attenuated in mice overexpressing PTN. However, there is little information about the signaling systems recruited by PTN to modulate neural activity. To address this issue, the gene expression profile in hippocampus of mice lacking PTN was analyzed using microarrays of 22,000 genes. In addition, we corroborated the effect of the absence of PTN on the expression of these genes by silencing this growth factor in primary neuronal cultures in vitro. The microarray analysis identified 102 genes that are differentially expressed (z-score>3.0) in PTN null mice, and the expression of eight of those modified in the hippocampus of KO mice was also modified in vitro after silencing PTN in cultured neurons with siRNAs. The data obtained indicate that the absence of PTN affects AKT pathway response and modulates the expression of genes related with neuroprotection (Mgst3 and Estrogen receptor 1, Ers 1) and cell differentiation (Caspase 6, Nestin, and Odz4), both in vivo and in vitro.
Collapse
Affiliation(s)
- Celia González-Castillo
- Doctorado en Ciencias en Biología Molecular en Medicina (DCBMM), CUCS, Universidad de Guadalajara, Jalisco, Mexico
| | - Daniel Ortuño-Sahagún
- Instituto de Investigación en Ciencias Biomédicas (IICB), CUCS, Universidad de Guadalajara, Jalisco, Mexico.
| | - Carolina Guzmán-Brambila
- Tecnológico de Monterrey, División de Biotecnología y Salud, Escuela de Medicina, Campus Guadalajara, Jalisco, Mexico
| | - Ana Laura Márquez-Aguirre
- Unidad de Biotecnología Médica y Farmacéutica, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco A.C., 44270 Guadalajara, Jalisco, Mexico
| | - Rita Raisman-Vozari
- Sorbonne Université UPMC UM75 INSERM U1127, CNRS UMR 7225, Institut du Cerveau et de la Moelle Epinière, Paris, France
| | - Mercé Pallás
- Department of Pharmacology and Medical Chemistry, Faculty of Pharmacy, Institute of Neuroscience (INUB), Centros de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), University of Barcelona, Spain
| | - Argelia E Rojas-Mayorquín
- Departamento de Ciencias Ambientales, Instituto de Neurociencias, CUCBA, Universidad de Guadalajara, Jalisco, Mexico.
| |
Collapse
|
11
|
Viggiano A, Cacciola G, Widmer DAJ, Viggiano D. Anxiety as a neurodevelopmental disorder in a neuronal subpopulation: Evidence from gene expression data. Psychiatry Res 2015; 228:729-40. [PMID: 26089015 DOI: 10.1016/j.psychres.2015.05.032] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Revised: 05/14/2015] [Accepted: 05/26/2015] [Indexed: 12/20/2022]
Abstract
The relationship between genes and anxious behavior, is nor linear nor monotonic. To address this problem, we analyzed with a meta-analytic method the literature data of the behavior of knockout mice, retrieving 33 genes whose deletion was accompanied by increased anxious behavior, 34 genes related to decreased anxious behavior and 48 genes not involved in anxiety. We correlated the anxious behavior resulting from the deletion of these genes to their brain expression, using the Allen Brain Atlas and Gene Expression Omnibus (GEO) database. The main finding is that the genes accompanied, after deletion, by a modification of the anxious behavior, have lower expression in the cerebral cortex, the amygdala and the ventral striatum. The lower expression level was putatively due to their selective presence in a neuronal subpopulation. This difference was replicated also using a database of human gene expression, further showing that the differential expression pertained, in humans, a temporal window of young postnatal age (4 months up to 4 years) but was not evident at fetal or adult human stages. Finally, using gene enrichment analysis we also show that presynaptic genes are involved in the emergence of anxiety and postsynaptic genes in the reduction of anxiety after gene deletion.
Collapse
Affiliation(s)
- Adela Viggiano
- Department of Health Sciences, University of Molise, Campobasso 86100, Italy
| | - Giovanna Cacciola
- Department of Health Sciences, University of Molise, Campobasso 86100, Italy
| | | | - Davide Viggiano
- Department of Health Sciences, University of Molise, Campobasso 86100, Italy; Department of Cardio-Thoracic and Respiratory Science, Second University of Naples, Naples, Italy.
| |
Collapse
|
12
|
Smith PD, Coulson-Thomas VJ, Foscarin S, Kwok JCF, Fawcett JW. "GAG-ing with the neuron": The role of glycosaminoglycan patterning in the central nervous system. Exp Neurol 2015; 274:100-14. [PMID: 26277685 DOI: 10.1016/j.expneurol.2015.08.004] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 07/17/2015] [Accepted: 08/06/2015] [Indexed: 01/17/2023]
Abstract
Proteoglycans (PGs) are a diverse family of proteins that consist of one or more glycosaminoglycan (GAG) chains, covalently linked to a core protein. PGs are major components of the extracellular matrix (ECM) and play critical roles in development, normal function and damage-response of the central nervous system (CNS). GAGs are classified based on their disaccharide subunits, into the following major groups: chondroitin sulfate (CS), heparan sulfate (HS), heparin (HEP), dermatan sulfate (DS), keratan sulfate (KS) and hyaluronic acid (HA). All except HA are modified by sulfation, giving GAG chains specific charged structures and binding properties. While significant neuroscience research has focused on the role of one PG family member, chondroitin sulfate proteoglycan (CSPG), there is ample evidence in support of a role for the other PGs in regulating CNS function in normal and pathological conditions. This review discusses the role of all the identified PG family members (CS, HS, HEP, DS, KS and HA) in normal CNS function and in the context of pathology. Understanding the pleiotropic roles of these molecules in the CNS may open the door to novel therapeutic strategies for a number of neurological conditions.
Collapse
Affiliation(s)
- Patrice D Smith
- John van Geest Centre for Brain Repair, University of Cambridge, Forvie Site, Robinson Way, Cambridge, UK; Department of Neuroscience, Carleton University, Ottawa, ON, Canada.
| | - Vivien J Coulson-Thomas
- John van Geest Centre for Brain Repair, University of Cambridge, Forvie Site, Robinson Way, Cambridge, UK
| | - Simona Foscarin
- John van Geest Centre for Brain Repair, University of Cambridge, Forvie Site, Robinson Way, Cambridge, UK
| | - Jessica C F Kwok
- John van Geest Centre for Brain Repair, University of Cambridge, Forvie Site, Robinson Way, Cambridge, UK
| | - James W Fawcett
- John van Geest Centre for Brain Repair, University of Cambridge, Forvie Site, Robinson Way, Cambridge, UK.
| |
Collapse
|
13
|
González-Castillo C, Ortuño-Sahagún D, Guzmán-Brambila C, Pallàs M, Rojas-Mayorquín AE. Pleiotrophin as a central nervous system neuromodulator, evidences from the hippocampus. Front Cell Neurosci 2015; 8:443. [PMID: 25620911 PMCID: PMC4287103 DOI: 10.3389/fncel.2014.00443] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 12/10/2014] [Indexed: 02/04/2023] Open
Abstract
Pleiotrophin (PTN) is a secreted growth factor, and also a cytokine, associated with the extracellular matrix, which has recently starting to attract attention as a significant neuromodulator with multiple neuronal functions during development. PTN is expressed in several tissues, where its signals are generally related with cell proliferation, growth, and differentiation by acting through different receptors. In Central Nervous System (CNS), PTN exerts post-developmental neurotrophic and -protective effects, and additionally has been involved in neurodegenerative diseases and neural disorders. Studies in Drosophila shed light on some aspects of the different levels of regulatory control of PTN invertebrate homologs. Specifically in hippocampus, recent evidence from PTN Knock-out (KO) mice involves PTN functioning in learning and memory. In this paper, we summarize, discuss, and contrast the most recent advances and results that lead to proposing a PTN as a neuromodulatory molecule in the CNS, particularly in hippocampus.
Collapse
Affiliation(s)
- Celia González-Castillo
- Doctorwado en Ciencias en Biología Molecular en Medicina (DCBMM), CUCS, Universidad de Guadalajara Guadalajara, Jalisco, México
| | - Daniel Ortuño-Sahagún
- Instituto de Investigación en Ciencias Biomédicas (IICB), CUCS, Universidad de Guadalajara, Guadalajara Jalisco, México
| | - Carolina Guzmán-Brambila
- Tecnológico de Monterrey, División de Biotecnología y Salud, Escuela de Medicina, Campus Guadalajara Guadalajara, Jalisco, México
| | - Mercè Pallàs
- Department of Pharmacology and Medical Chemistry, Faculty of Pharmacy School of Pharmacy, Institute of Biomedicine (IBUB), Centros de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), University of Barcelona Barcelona, Spain
| | | |
Collapse
|
14
|
Behavioral and neuroanatomical abnormalities in pleiotrophin knockout mice. PLoS One 2014; 9:e100597. [PMID: 25000129 PMCID: PMC4085064 DOI: 10.1371/journal.pone.0100597] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 05/28/2014] [Indexed: 11/30/2022] Open
Abstract
Pleiotrophin (PTN) is an extracellular matrix-associated protein with neurotrophic and neuroprotective effects that is involved in a variety of neurodevelopmental processes. Data regarding the cognitive-behavioral and neuroanatomical phenotype of pleiotrophin knockout (KO) mice is limited. The purpose of this study was to more fully characterize this phenotype, with emphasis on the domains of learning and memory, cognitive-behavioral flexibility, exploratory behavior and anxiety, social behavior, and the neuronal and vascular microstructure of the lateral entorhinal cortex (EC). PTN KOs exhibited cognitive rigidity, heightened anxiety, behavioral reticence in novel contexts and novel social interactions suggestive of neophobia, and lamina-specific decreases in neuronal area and increases in neuronal density in the lateral EC. Initial learning of spatial and other associative tasks, as well as vascular density in the lateral EC, was normal in the KOs. These data suggest that the absence of PTN in vivo is associated with disruption of specific cognitive and affective processes, raising the possibility that further study of PTN KOs might have implications for the study of human disorders with similar features.
Collapse
|
15
|
Lee YS. Genes and signaling pathways involved in memory enhancement in mutant mice. Mol Brain 2014; 7:43. [PMID: 24894914 PMCID: PMC4050447 DOI: 10.1186/1756-6606-7-43] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 05/27/2014] [Indexed: 11/10/2022] Open
Abstract
Mutant mice have been used successfully as a tool for investigating the mechanisms of memory at multiple levels, from genes to behavior. In most cases, manipulating a gene expressed in the brain impairs cognitive functions such as memory and their underlying cellular mechanisms, including synaptic plasticity. However, a remarkable number of mutations have been shown to enhance memory in mice. Understanding how to improve a system provides valuable insights into how the system works under normal conditions, because this involves understanding what the crucial components are. Therefore, more can be learned about the basic mechanisms of memory by studying mutant mice with enhanced memory. This review will summarize the genes and signaling pathways that are altered in the mutants with enhanced memory, as well as their roles in synaptic plasticity. Finally, I will discuss how knowledge of memory-enhancing mechanisms could be used to develop treatments for cognitive disorders associated with impaired plasticity.
Collapse
Affiliation(s)
- Yong-Seok Lee
- Department of Life Science, College of Natural Science, Chung-Ang University, Seoul 156-756, Republic of Korea.
| |
Collapse
|
16
|
Neural ECM molecules in synaptic plasticity, learning, and memory. PROGRESS IN BRAIN RESEARCH 2014; 214:53-80. [DOI: 10.1016/b978-0-444-63486-3.00003-7] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
17
|
Shetty GA, Hattiangady B, Shetty AK. Neural stem cell- and neurogenesis-related gene expression profiles in the young and aged dentate gyrus. AGE (DORDRECHT, NETHERLANDS) 2013; 35:2165-2176. [PMID: 23322452 PMCID: PMC3824978 DOI: 10.1007/s11357-012-9507-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Accepted: 12/27/2012] [Indexed: 06/01/2023]
Abstract
Hippocampal neurogenesis, important for memory and mood function, wanes greatly in old age. Studies in rat models have implied that this decrease is not due to loss of neural stem cells (NSCs) in the subgranular zone of the dentate gyrus (DG) but rather due to an increased quiescence of NSCs. Additional studies have suggested that changes in the microenvironment, particularly declines in the concentrations of neurotrophic factors, underlie this change. In this study, we compared the expression of 84 genes that are important for NSC proliferation and neurogenesis between the DG of young (4 months old) and aged (24 months old) Fischer 344 rats, using a quantitative real-time polymerase chain reaction array. Interestingly, the expression of a vast majority of genes that have been reported previously to positively or negatively regulate NSC proliferation was unaltered with aging. Furthermore, most genes important for cell cycle arrest, regulation of cell differentiation, growth factors and cytokine levels, synaptic functions, apoptosis, cell adhesion and cell signaling, and regulation of transcription displayed stable expression in the DG with aging. The exceptions included increased expression of genes important for NSC proliferation and neurogenesis (Stat3 and Shh), DNA damage response and NF-kappaB signaling (Cdk5rap3), neuromodulation (Adora1), and decreased expression of a gene important for neuronal differentiation (HeyL). Thus, age-related decrease in hippocampal neurogenesis is not associated with a decline in the expression of selected genes important for NSC proliferation and neurogenesis in the DG.
Collapse
Affiliation(s)
- Geetha A. Shetty
- />Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine at Scott & White, 5701 Airport Road, Module C, Temple, 76502 TX USA
- />Research Service, Olin E. Teague Veterans’ Medical Center, CTVHCS, Temple, TX USA
- />Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX USA
| | - Bharathi Hattiangady
- />Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine at Scott & White, 5701 Airport Road, Module C, Temple, 76502 TX USA
- />Research Service, Olin E. Teague Veterans’ Medical Center, CTVHCS, Temple, TX USA
- />Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX USA
- />Division of Neurosurgery, Duke University Medical Center, Durham, NC USA
- />Research and Surgery Services, Durham Veterans Affairs Medical Center, Durham, NC USA
| | - Ashok K. Shetty
- />Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine at Scott & White, 5701 Airport Road, Module C, Temple, 76502 TX USA
- />Research Service, Olin E. Teague Veterans’ Medical Center, CTVHCS, Temple, TX USA
- />Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX USA
- />Division of Neurosurgery, Duke University Medical Center, Durham, NC USA
- />Research and Surgery Services, Durham Veterans Affairs Medical Center, Durham, NC USA
| |
Collapse
|
18
|
Expression and localization of receptor protein tyrosine phosphatase β and its ligand pleiotrophin in the submandibular gland of mice. Arch Oral Biol 2013; 58:181-91. [DOI: 10.1016/j.archoralbio.2012.09.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Revised: 07/31/2012] [Accepted: 09/20/2012] [Indexed: 01/06/2023]
|
19
|
Abstract
Astrocytosis, a process in which astrocytes undergo proliferation and enhancement of glial fibrillary acidic protein (GFAP) expression, has been suggested to play important roles in the maintenance of dependence to amphetamine and its derivatives. It was previously shown that mice with genetic deletion of pleiotrophin (PTN), a neurotrophic factor upregulated in different brain areas after administration of amphetamine, show a longer lasting amphetamine-induced conditioned place preference (CPP) when compared to wild type mice. In this work, we aimed to pursue the possibility of a different astrocytic response induced by amphetamine in PTN-/- and PTN+/+ mice, which could underlie the higher vulnerability of PTN-/- mice to maintain amphetamine CPP. In confirmation of previous studies, we found that PTN-/- mice significantly maintained amphetamine (3mg/kg)-induced CPP 5 days after the last drug administration compared to PTN+/+ mice. Interestingly, the number of astrocytes in nucleus accumbens (NAcc), cingulate cortex (CG) and caudate putamen (CPu) did not differ between mice that maintained and did not maintain amphetamine-induced CPP independently of the genotype considered. However, we found that PTN-/- mice showed significantly decreased numbers of astrocytes in CG and CPu compared to PTN+/+ mice independently of whether they maintained amphetamine-induced CPP 5 days after the last drug administration or not. The data demonstrate that maintenance of amphetamine-induced CPP depends on the endogenous expression of PTN. The data tend to discard a correlation between activated astrocytes and maintenance of amphetamine conditioning effects and suggest PTN as a potential modulator of activation of astrocytes after amphetamine treatment.
Collapse
|
20
|
Rosenfield SM, Bowden ET, Cohen-Missner S, Gibby KA, Ory V, Henke RT, Riegel AT, Wellstein A. Pleiotrophin (PTN) expression and function and in the mouse mammary gland and mammary epithelial cells. PLoS One 2012; 7:e47876. [PMID: 23077670 PMCID: PMC3471873 DOI: 10.1371/journal.pone.0047876] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Accepted: 09/24/2012] [Indexed: 11/19/2022] Open
Abstract
Expression of the heparin-binding growth factor, pleiotrophin (PTN) in the mammary gland has been reported but its function during mammary gland development is not known. We examined the expression of PTN and its receptor ALK (Anaplastic Lymphoma Kinase) at various stages of mouse mammary gland development and found that their expression in epithelial cells is regulated in parallel during pregnancy. A 30-fold downregulation of PTN mRNA expression was observed during mid-pregnancy when the mammary gland undergoes lobular-alveolar differentiation. After weaning of pups, PTN expression was restored although baseline expression of PTN was reduced significantly in mammary glands of mice that had undergone multiple pregnancies. We found PTN expressed in epithelial cells of the mammary gland and thus used a monoclonal anti-PTN blocking antibody to elucidate its function in cultured mammary epithelial cells (MECs) as well as during gland development. Real-time impedance monitoring of MECs growth, migration and invasion during anti-PTN blocking antibody treatment showed that MECs motility and invasion but not proliferation depend on the activity of endogenous PTN. Increased number of mammospheres with laminin deposition after anti-PTN blocking antibody treatment of MECs in 3D culture and expression of progenitor markers suggest that the endogenously expressed PTN inhibits the expansion and differentiation of epithelial progenitor cells by disrupting cell-matrix adhesion. In vivo, PTN activity was found to inhibit ductal outgrowth and branching via the inhibition of phospho ERK1/2 signaling in the mammary epithelial cells. We conclude that PTN delays the maturation of the mammary gland by maintaining mammary epithelial cells in a progenitor phenotype and by inhibiting their differentiation during mammary gland development.
Collapse
Affiliation(s)
- Sonia M. Rosenfield
- Lombardi Cancer Center, Georgetown University, Washington, District of Columbia, United States of America
| | - Emma T. Bowden
- MedImmune, Gaithersburg, Maryland, United States of America
| | - Shani Cohen-Missner
- Lombardi Cancer Center, Georgetown University, Washington, District of Columbia, United States of America
| | - Krissa A. Gibby
- Lombardi Cancer Center, Georgetown University, Washington, District of Columbia, United States of America
| | - Virginie Ory
- Lombardi Cancer Center, Georgetown University, Washington, District of Columbia, United States of America
| | - Ralf T. Henke
- Lombardi Cancer Center, Georgetown University, Washington, District of Columbia, United States of America
| | - Anna T. Riegel
- Lombardi Cancer Center, Georgetown University, Washington, District of Columbia, United States of America
| | - Anton Wellstein
- Lombardi Cancer Center, Georgetown University, Washington, District of Columbia, United States of America
- * E-mail:
| |
Collapse
|
21
|
Wlodarczyk J, Mukhina I, Kaczmarek L, Dityatev A. Extracellular matrix molecules, their receptors, and secreted proteases in synaptic plasticity. Dev Neurobiol 2012; 71:1040-53. [PMID: 21793226 DOI: 10.1002/dneu.20958] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Neural cells secrete diverse molecules, which accumulate in the extracellular space and form the extracellular matrix (ECM). Interactions between cells and the ECM are well recognized to play the crucial role in cell migration and guidance of growing axons, whereas formation of mature neural ECM in the form of perineuronal nets is believed to restrict certain forms of developmental plasticity. On the other hand, major components of perineuronal nets and other ECM molecules support induction of functional plasticity, the most studied form of which is long-term potentiation. Here, we review the underlying mechanisms by which ECM molecules, their receptors and remodeling proteases regulate the induction and maintenance of synaptic modifications. In particular, we highlight that activity-dependent secretion and activation of proteases leads to a local cleavage of the ECM and release of signaling proteolytic fragments. These molecules regulate transmitter receptor trafficking, actin cytoskeleton, growth of dendritic spines, and formation of dendritic filopodia.
Collapse
|
22
|
Mukhina IV, Korotchenko SA, Dityatev AE. Extracellular matrix molecules, their receptors, and extracellular proteases as synaptic plasticity modulators. NEUROCHEM J+ 2012. [DOI: 10.1134/s1819712412020055] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
23
|
Sone M, Muramatsu H, Muramatsu T, Nakashima T. Morphological observation of the stria vascularis in midkine and pleiotrophin knockout mice. Auris Nasus Larynx 2011; 38:41-5. [DOI: 10.1016/j.anl.2010.05.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2010] [Revised: 04/26/2010] [Accepted: 05/06/2010] [Indexed: 02/04/2023]
|
24
|
Neunaber C, Catala-Lehnen P, Beil FT, Marshall RP, Kanbach V, Baranowsky A, Lehmann W, Streichert T, Ignatius A, Muramatsu T, Schinke T, Amling M. Increased trabecular bone formation in mice lacking the growth factor midkine. J Bone Miner Res 2010; 25:1724-35. [PMID: 20200993 DOI: 10.1002/jbmr.75] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Midkine (Mdk) and pleiotrophin (Ptn) comprise a family of heparin-binding growth factors known primarily for their effects on neuronal cells. Since transgenic mice overexpressing Ptn have been reported to display increased bone density, we have previously analyzed Ptn-deficient mice but failed to detect any abnormality of skeletal development and remodeling. Together with the finding that Mdk expression increases in the course of primary osteoblast differentiation, we reasoned that Mdk, rather than Ptn, could play a physiologic role in bone formation. Here, we show that Mdk-deficient mice display an increased trabecular bone volume at 12 and 18 months of age, accompanied by cortical porosity. Histomorphometric quantification demonstrated an increased bone-formation rate compared with wild-type littermates, whereas bone resorption was differentially affected in trabecular and cortical bone of Mdk-deficient mice. To understand the effect of Mdk on bone formation at the molecular level, we performed a genome-wide expression analysis of primary osteoblasts and identified Ank and Enpp1 as Mdk-induced genes whose decreased expression in Mdk-deficient osteoblasts may explain, at least in part, the observed skeletal phenotype. Finally, we performed ovariectomy and observed bone loss only in wild-type but not in Mdk-deficient animals. Taken together, our data demonstrate that Mdk deficiency, at least in mice, results in an increased trabecular bone formation, thereby raising the possibility that Mdk-specific antagonists might prove beneficial in osteoporosis therapy.
Collapse
Affiliation(s)
- Claudia Neunaber
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Kalus I, Salmen B, Viebahn C, von Figura K, Schmitz D, D'Hooge R, Dierks T. Differential involvement of the extracellular 6-O-endosulfatases Sulf1 and Sulf2 in brain development and neuronal and behavioural plasticity. J Cell Mol Med 2010; 13:4505-21. [PMID: 20394677 PMCID: PMC4515066 DOI: 10.1111/j.1582-4934.2008.00558.x] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The extracellular sulfatases Sulf1 and Sulf2 remove specific 6-O-sulfate groups from heparan sulfate, thereby modulating numerous signalling pathways underlying development and homeostasis. In vitro data have suggested that the two enzymes show functional redundancy. To elucidate their in vivo functions and to further address the question of a putative redundancy, we have generated Sulf1- and Sulf2-deficient mice. Phenotypic analysis of these animals revealed higher embryonic lethality of Sulf2 knockout mice, which can be associated with neuroanatomical malformations during embryogenesis. Sulf1 seems not to be essential for developmental or postnatal viability, as mice deficient in this sulfatase show no overt phenotype. However, neurite outgrowth deficits were observed in hippocampal and cerebellar neurons of both mutant mouse lines, suggesting that not only Sulf2 but also Sulf1 function plays a role in the developing nervous system. Behavioural analysis revealed differential deficits with regard to cage activity and spatial learning for Sulf1- and Sulf2-deficient mouse lines. In addition, Sulf1-specific deficits were shown for synaptic plasticity in the CA1 region of the hippocampus, associated with a reduced spine density. These results reveal that Sulf1 and Sulf2 fulfil non-redundant functions in vivo in the development and maintenance of the murine nervous system.
Collapse
Affiliation(s)
- Ina Kalus
- Department of Chemistry, Biochemistry I, Bielefeld University, Bielefeld, Germany
| | | | | | | | | | | | | |
Collapse
|
26
|
Weng T, Liu L. The role of pleiotrophin and beta-catenin in fetal lung development. Respir Res 2010; 11:80. [PMID: 20565841 PMCID: PMC2901351 DOI: 10.1186/1465-9921-11-80] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Accepted: 06/18/2010] [Indexed: 12/21/2022] Open
Abstract
Mammalian lung development is a complex biological process, which is temporally and spatially regulated by growth factors, hormones, and extracellular matrix proteins. Abnormal changes of these molecules often lead to impaired lung development, and thus pulmonary diseases. Epithelial-mesenchymal interactions are crucial for fetal lung development. This paper reviews two interconnected pathways, pleiotrophin and Wnt/β-catenin, which are involved in fibroblast and epithelial cell communication during fetal lung development.
Collapse
Affiliation(s)
- Tingting Weng
- Department of Physiological Sciences, Oklahoma State University, Stillwater, Oklahoma 74078, USA
| | | |
Collapse
|
27
|
An integrated systems analysis implicates EGR1 downregulation in simian immunodeficiency virus encephalitis-induced neural dysfunction. J Neurosci 2009; 29:12467-76. [PMID: 19812322 DOI: 10.1523/jneurosci.3180-09.2009] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Human immunodeficiency virus (HIV)-associated dementia (HAD) is a syndrome occurring in HIV-infected patients with advanced disease that likely develops as a result of macrophage and microglial activation as well as other immune events triggered by virus in the central nervous system. The most relevant experimental model of HAD, rhesus macaques exhibiting simian immunodeficiency virus (SIV) encephalitis (SIVE), closely reproduces the human disease and has been successfully used to advance our understanding of mechanisms underlying HAD. In this study we integrate gene expression data from uninfected and SIV-infected hippocampus with a human protein interaction network and discover modules of genes whose expression patterns distinguish these two states, to facilitate identification of neuronal genes that may contribute to SIVE/HIV cognitive deficits. Using this approach we identify several downregulated candidate genes and select one, EGR1, a key molecule in hippocampus-related learning and memory, for further study. We show that EGR1 is downregulated in SIV-infected hippocampus and that it can be downregulated in differentiated human neuroblastoma cells by treatment with CCL8, a product of activated microglia. Integration of expression data with protein interaction data to discover discriminatory modules of interacting proteins can be usefully used to prioritize differentially expressed genes for further study. Investigation of EGR1, selected in this manner, indicates that its downregulation in SIVE may occur as a consequence of the host response to infection, leading to deficits in cognition.
Collapse
|
28
|
del Olmo N, Gramage E, Alguacil LF, Pérez-Pinera P, Deuel TF, Herradón G. Pleiotrophin inhibits hippocampal long-term potentiation: a role of pleiotrophin in learning and memory. Growth Factors 2009; 27:189-94. [PMID: 19384682 DOI: 10.1080/08977190902906859] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Pleiotrophin (PTN) is a growth factor that has been shown to be involved in hippocampal synaptic plasticity and learning. To further understand the involvement of PTN in memory processes, we performed in vitro electrophysiological studies in PTN-stimulated CA1 from rat hippocampal slices combined with the behavioural testing of PTN deficient (PTN - / - ) mice. We found that PTN inhibited hippocampal long-term potentiation (LTP) induced by high-frequency stimulation (HFS) consisted in three trains of 100 Hz separated by 20 s. To test the possibility that PTN might be involved in behavioural memory processes, we tested the learning behaviour of PTN - / - mice using the Y-maze test. We did not observe significant differences in recognition memory between PTN - / - and Wild Type (WT) mice when a 30 min-interval intertrial (ITI) was used in the Y-maze test. However, whereas WT mice showed disruption of recognition memory using a 60 min-ITI, PTN - / - mice maintained the recognition memory. The data demonstrate that PTN inhibits hippocampal LTP in vitro and might play a role in memory processes in vivo.
Collapse
Affiliation(s)
- Nuria del Olmo
- Laboratory of Pharmacology and Toxicology, Universidad San Pablo CEU, Madrid, Spain
| | | | | | | | | | | |
Collapse
|
29
|
Imai S, Heino TJ, Hienola A, Kurata K, Büki K, Matsusue Y, Väänänen HK, Rauvala H. Osteocyte-derived HB-GAM (pleiotrophin) is associated with bone formation and mechanical loading. Bone 2009; 44:785-94. [PMID: 19442624 DOI: 10.1016/j.bone.2009.01.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2008] [Revised: 12/19/2008] [Accepted: 01/02/2009] [Indexed: 02/04/2023]
Abstract
HB-GAM (also known as pleiotrophin) is a cell matrix-associated protein that is highly expressed in bone. It affects osteoblast function, and might therefore play a role in bone development and remodeling. We aimed to investigate the role of HB-GAM in bone in vivo and in vitro. The bones of HB-GAM deficient mice with an inbred mouse background were studied by histological, histomorphometrical, radiological, biomechanical and mu-CT analyses and the effect of immobilization was evaluated. HB-GAM localization in vivo was studied. MLO-Y4 osteocytes were subjected to fluid shear stress in vitro, and gene and protein expression were studied by subtractive hybridization, quantitative PCR and Western blot. Human osteoclasts were cultured in the presence of rhHB-GAM and their formation and resorption activities were assayed. In agreement with previous reports, the skeletal structure of the HB-GAM knockout mice developed normally. However, a growth retardation of the weight-bearing bones was observed by 2 months of age, suggesting a link to physical activity. Adult HB-GAM deficient mice were characterized by low bone formation and osteopenia, as well as resistance to immobilization-dependent bone remodeling. HB-GAM was localized around osteocytes and their processes in vivo and furthermore, osteocytic HB-GAM expression was upregulated by mechanical loading in vitro. HB-GAM did not affect on human osteoclast formation or resorption in vitro. Taken together, our results suggest that HB-GAM is an osteocyte-derived factor that could participate in mediating the osteogenic effects of mechanical loading on bone.
Collapse
Affiliation(s)
- S Imai
- Department of Orthopaedic Surgery, Shiga University of Medical Science, Setatsukinowa, Otsu, Japan
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
Most molecular and cellular studies of cognitive function have focused on either normal or pathological states, but recent research with transgenic mice has started to address the mechanisms of enhanced cognition. These results point to key synaptic and nuclear signalling events that can be manipulated to facilitate the induction or increase the stability of synaptic plasticity, and therefore enhance the acquisition or retention of information. Here, we review these surprising findings and explore their implications to both mechanisms of learning and memory and to ongoing efforts to develop treatments for cognitive disorders. These findings represent the beginning of a fundamental new approach in the study of enhanced cognition.
Collapse
Affiliation(s)
- Yong-Seok Lee
- Department of Neurobiology, Brain Research Institute, University of California, Los Angeles, California 90095, USA
| | | |
Collapse
|
31
|
Riekki R, Pavlov I, Tornberg J, Lauri SE, Airaksinen MS, Taira T. Altered synaptic dynamics and hippocampal excitability but normal long-term plasticity in mice lacking hyperpolarizing GABA A receptor-mediated inhibition in CA1 pyramidal neurons. J Neurophysiol 2008; 99:3075-89. [PMID: 18436638 DOI: 10.1152/jn.00606.2007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
GABA(A) receptor (GABA-AR)-mediated inhibition is critical for proper operation of neuronal networks. Synaptic inhibition either shifts the membrane potential farther away from the action potential firing threshold (hyperpolarizing inhibition) or via increase in the membrane conductance shunts the excitatory currents. However, the relative importance of these different forms of inhibition on the hippocampal function is unclear. To study the functional consequences of the absence of hyperpolarizing inhibition, we have used KCC2-deficient mice (KCC2hy/null) maintaining only 15-20% of the neuron-specific K-Cl-cotransporter. Gramicidin-perforated patch-clamp recordings in hippocampal CA1 pyramidal cells revealed that the reversal potential of the GABA-AR-mediated postsynaptic currents (E(GABA-A)) was approximately 20 mV more positive in KCC2hy/null mice than in wild-type (WT) animals. The basic glutamatergic transmission appeared unaltered in the KCC2hy/null mice, yet they displayed lowered threshold for stimulation-induced synchronous afterdischarges in the CA1 area. Also fatigue of field excitatory postsynaptic potentials/excitatory postsynaptic currents in response to repetitious stimulation was smaller in KCC2hy/null mice, indicating altered synaptic dynamics. Interestingly, this effect was present also under blockade of GABA-ARs and was dependent on the extracellular K+ concentration. Moreover, there were no differences in the levels of either long-term potentiation or long-term depression between the genotypes. The local hippocampal CA1 network can in several aspects maintain its functional viability even in the absence of hyperpolarizing inhibition in pyramidal cells. Our results underscore the central role of shunting type of inhibition in controlling the neuronal excitation/inhibition balance. Moreover, our data demonstrate a novel, unexpected role for the KCC2, namely the modulation of properties of glutamatergic transmission during repetitious afferent activity.
Collapse
Affiliation(s)
- Ruusu Riekki
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | | | | | | | | | | |
Collapse
|
32
|
Schinke T, Gebauer M, Schilling AF, Lamprianou S, Priemel M, Mueldner C, Neunaber C, Streichert T, Ignatius A, Harroch S, Amling M. The protein tyrosine phosphatase Rptpzeta is expressed in differentiated osteoblasts and affects bone formation in mice. Bone 2008; 42:524-34. [PMID: 18178537 DOI: 10.1016/j.bone.2007.11.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2007] [Revised: 09/14/2007] [Accepted: 11/07/2007] [Indexed: 01/07/2023]
Abstract
Tyrosine phosphorylation of intracellular substrates is one mechanism to regulate cellular proliferation and differentiation. Protein tyrosine phosphatases (PTPs) act by dephosphorylation of substrates and thereby counteract the activity of tyrosine kinases. Few PTPs have been suggested to play a role in bone remodeling, one of them being Rptpzeta, since it has been shown to be suppressed by pleiotrophin, a heparin-binding molecule affecting bone formation, when over-expressed in transgenic mice. In a genome-wide expression analysis approach we found that Ptprz1, the gene encoding Rptpzeta, is strongly induced upon terminal differentiation of murine primary calvarial osteoblasts. Using RT-PCR and Western Blotting we further demonstrated that differentiated osteoblasts, in contrast to neuronal cells, specifically express the short transmembrane isoform of Rptpzeta. To uncover a potential role of Rptpzeta in bone remodeling we next analyzed the skeletal phenotype of a Rptpzeta-deficient mouse model using non-decalcified histology and histomorphometry. Compared to wildtype littermates, the Rptpzeta-deficient mice display a decreased trabecular bone volume at the age of 50 weeks, caused by a reduced bone formation rate. Likewise, Rptpzeta-deficient calvarial osteoblasts analyzed ex vivo display decreased expression of osteoblast markers, indicating a cell-autonomous defect. This was confirmed by the finding that Rptpzeta-deficient osteoblasts had a diminished potential to form osteocyte-like cellular extensions on Matrigel-coated surfaces. Taken together, these data provide the first evidence for a physiological role of Rptpzeta in bone remodeling, and thus identify Rptpzeta as the first PTP regulating bone formation in vivo.
Collapse
Affiliation(s)
- T Schinke
- Department of Trauma, Hand, and Reconstructive Surgery, University Medical Center Hamburg Eppendorf, Hamburg 20246, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Bilsland JG, Wheeldon A, Mead A, Znamenskiy P, Almond S, Waters KA, Thakur M, Beaumont V, Bonnert TP, Heavens R, Whiting P, McAllister G, Munoz-Sanjuan I. Behavioral and neurochemical alterations in mice deficient in anaplastic lymphoma kinase suggest therapeutic potential for psychiatric indications. Neuropsychopharmacology 2008; 33:685-700. [PMID: 17487225 DOI: 10.1038/sj.npp.1301446] [Citation(s) in RCA: 132] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The receptor tyrosine kinase product of the anaplastic lymphoma kinase (ALK) gene has been implicated in oncogenesis as a product of several chromosomal translocations, although its endogeneous role in the hematopoietic and neural systems has remained poorly understood. We describe that the generation of animals homozygous for a deletion of the ALK tyrosine kinase domain leads to alterations in adult brain function. Evaluation of adult ALK homozygotes (HOs) revealed an age-dependent increase in basal hippocampal progenitor proliferation and alterations in behavioral tests consistent with a role for this receptor in the adult brain. ALK HO animals displayed an increased struggle time in the tail suspension test and the Porsolt swim test and enhanced performance in a novel object-recognition test. Neurochemical analysis demonstrates an increase in basal dopaminergic signalling selectively within the frontal cortex. Altogether, these results suggest that ALK functions in the adult brain to regulate the function of the frontal cortex and hippocampus and identifies ALK as a new target for psychiatric indications, such as schizophrenia and depression, with an underlying deregulated monoaminergic signalling.
Collapse
Affiliation(s)
- James G Bilsland
- Department of Molecular and Cellular Neuroscience, Merck Sharp and Dohme, The Neuroscience Research Centre, Essex, UK
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Shukla K, Kim J, Blundell J, Powell CM. Learning-induced glutamate receptor phosphorylation resembles that induced by long term potentiation. J Biol Chem 2007; 282:18100-18107. [PMID: 17472959 PMCID: PMC3910102 DOI: 10.1074/jbc.m702906200] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Long term potentiation and long term depression of synaptic responses in the hippocampus are thought to be critical for certain forms of learning and memory, although until recently it has been difficult to demonstrate that long term potentiation or long term depression occurs during hippocampus-dependent learning. Induction of long term potentiation or long term depression in hippocampal slices in vitro modulates phosphorylation of the alpha-amino-3-hydrozy-5-methylisoxazole-4-propionic acid subtype of glutamate receptor subunit GluR1 at distinct phosphorylation sites. In long term potentiation, GluR1 phosphorylation is increased at the Ca2+/calmodulin-dependent protein kinase and protein kinase C site serine 831, whereas in long term depression, phosphorylation of the protein kinase A site serine 845 is decreased. Indeed, phosphorylation of one or both of these sites is required for long term synaptic plasticity and for certain forms of learning and memory. Here we demonstrate that training in a hippocampus-dependent learning task, contextual fear conditioning is associated with increased phosphorylation of GluR1 at serine 831 in the hippocampal formation. This increased phosphorylation is specific to learning, has a similar time course to that in long term potentiation, and like memory and long term potentiation, is dependent on N-methyl-D-aspartate receptor activation during training. Furthermore, the learning-induced increase in serine 831 phosphorylation is present at synapses and is in heteromeric complexes with the glutamate receptor subunit GluR2. These data indicate that a biochemical correlate of long term potentiation occurs at synapses in receptor complexes in a final, downstream, postsynaptic effector of long term potentiation during learning in vivo, further strengthening the link between long term potentiation and memory.
Collapse
Affiliation(s)
- Kajal Shukla
- Departments of Neurology and Psychiatry, The University of Texas Southwestern Medical Center, Dallas, Texas 75390-8813
| | - James Kim
- Departments of Neurology and Psychiatry, The University of Texas Southwestern Medical Center, Dallas, Texas 75390-8813
| | - Jacqueline Blundell
- Departments of Neurology and Psychiatry, The University of Texas Southwestern Medical Center, Dallas, Texas 75390-8813
| | - Craig M Powell
- Departments of Neurology and Psychiatry, The University of Texas Southwestern Medical Center, Dallas, Texas 75390-8813.
| |
Collapse
|
35
|
Mi R, Chen W, Höke A. Pleiotrophin is a neurotrophic factor for spinal motor neurons. Proc Natl Acad Sci U S A 2007; 104:4664-9. [PMID: 17360581 PMCID: PMC1838658 DOI: 10.1073/pnas.0603243104] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Regeneration in the peripheral nervous system is poor after chronic denervation. Denervated Schwann cells act as a "transient target" by secreting growth factors to promote regeneration of axons but lose this ability with chronic denervation. We discovered that the mRNA for pleiotrophin (PTN) was highly up-regulated in acutely denervated distal sciatic nerves, but high levels of PTN mRNA were not maintained in chronically denervated nerves. PTN protected spinal motor neurons against chronic excitotoxic injury and caused increased outgrowth of motor axons out of the spinal cord explants and formation of "miniventral rootlets." In neonatal mice, PTN protected the facial motor neurons against cell death induced by deprivation from target-derived growth factors. Similarly, PTN significantly enhanced regeneration of myelinated axons across a graft in the transected sciatic nerve of adult rats. Our findings suggest a neurotrophic role for PTN that may lead to previously unrecognized treatment options for motor neuron disease and motor axonal regeneration.
Collapse
Affiliation(s)
- Ruifa Mi
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Weiran Chen
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Ahmet Höke
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- *To whom correspondence should be addressed at:
Department of Neurology, Johns Hopkins University, 600 North Wolfe Street, Path 509, Baltimore, MD 21287. E-mail:
| |
Collapse
|
36
|
Hamma-Kourbali Y, Bernard-Pierrot I, Heroult M, Dalle S, Caruelle D, Milhiet PE, Fernig DG, Delbé J, Courty J. Inhibition of the mitogenic, angiogenic and tumorigenic activities of pleiotrophin by a synthetic peptide corresponding to its C-thrombospondin repeat-I domain. J Cell Physiol 2007; 214:250-9. [PMID: 17607711 DOI: 10.1002/jcp.21191] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Pleiotrophin (PTN), is a heparin-dependent growth factor involved in angiogenesis and tumor growth. PTN contains a thrombospondin repeat-I (TSR-I) motif in its two beta-sheet domains that are involved in its binding to heparin and its neurite outgrowth activity. Based on the importance of the binding of PTN to heparin in its dimerization and biological activities, we have designed two synthetic peptides, P(13-39) and P(65-97) corresponding to a part of the N-terminal and C-terminal TSR-I motif of PTN, respectively. P(65-97) inhibited the mitogenic, tumorigenic and angiogenic activities of PTN, as well as the mitogenic and an angiogenic activity of fibroblast growth factor-2 (FGF-2). However, P(65-97) had no effect on the mitogenic activity of epidermal growth factor, which does not bind heparin. P(65-97) but not P(13-39) inhibited the binding of PTN and to a lesser extent of FGF-2 to heparin using an immunoassay and an optical biosensor assay and bound directly to heparin with a K(d) of 120 nM. These findings suggest that P(65-97), containing amino acids 65-97 of the TSR-I motif of the C-terminal domain of PTN, inhibits the activities of PTN and FGF-2 by virtue of its ability to bind heparin very effectively and so compete with the growth factors for their polysaccharide co-receptor.
Collapse
Affiliation(s)
- Yamina Hamma-Kourbali
- Laboratoire de Recherche sur la Croissance Cellulaire, la Réparation et la Régénération Tissulaires (CRRET), CNRS UMR 7149, Université Paris XII, Créteil Cedex, France
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Wright JW, Brown TE, Harding JW. Inhibition of hippocampal matrix metalloproteinase-3 and -9 disrupts spatial memory. Neural Plast 2006; 2007:73813. [PMID: 17502908 PMCID: PMC1838960 DOI: 10.1155/2007/73813] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2006] [Revised: 09/21/2006] [Accepted: 09/26/2006] [Indexed: 11/24/2022] Open
Abstract
Memory consolidation requires synaptic reconfiguration dependent upon extracellular matrix (ECM) molecules interacting with cell adhesion molecules. Matrix metalloproteinase (MMP) activity is responsible for transient alterations in the ECM that may be prerequisite to hippocampal-dependent learning. In support of this hypothesis we have measured increases in MMP-3 and MMP-9 levels within the hippocampus and
prefrontal cortex during Morris water maze training. The present investigation extends these findings by determining that infusion of an MMP inhibitor (FN-439) into the dorsal hippocampus disrupted acquisition of this task. In vitro fluorescence enzyme assays to determine the specificity of FN-439 against the catalytic domains of MMP-3 and MMP-9 indicated mean ± SEM IC50s of 16.2 ± 7.8 and 210.5 ± 37.8 μM, respectively, while in situ zymography using hippocampal sections treated with FN-439 indicated significant reductions in MMP gelatinase activity. These results suggest that compromising the ability of the dorsal hippocampus to reconfigure ECM molecules by inhibiting MMP activity interferes with appropriate spatial
memory acquisition, and support a role for hippocampal MMPs in the phenomena of spatial memory acquisition and storage.
Collapse
Affiliation(s)
- John W. Wright
- Department of Psychology, Washington State University, Pullman, WA 99164-4820, USA
- Department of Veterinary and Comparative Anatomy, Pharmacology and Physiology, Washington State University,
Pullman, WA 99164-6520, USA
- Programs in Neuroscience and Biotechnology, Washington State University, Pullman, WA 99164-6520, USA
- *John W. Wright:
| | - Travis E. Brown
- Department of Veterinary and Comparative Anatomy, Pharmacology and Physiology, Washington State University,
Pullman, WA 99164-6520, USA
- Programs in Neuroscience and Biotechnology, Washington State University, Pullman, WA 99164-6520, USA
| | - Joseph W. Harding
- Department of Psychology, Washington State University, Pullman, WA 99164-4820, USA
- Department of Veterinary and Comparative Anatomy, Pharmacology and Physiology, Washington State University,
Pullman, WA 99164-6520, USA
- Programs in Neuroscience and Biotechnology, Washington State University, Pullman, WA 99164-6520, USA
| |
Collapse
|
38
|
Zou P, Muramatsu H, Sone M, Hayashi H, Nakashima T, Muramatsu T. Mice doubly deficient in the midkine and pleiotrophin genes exhibit deficits in the expression of beta-tectorin gene and in auditory response. J Transl Med 2006; 86:645-53. [PMID: 16619002 DOI: 10.1038/labinvest.3700428] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
alpha-Tectorin and beta-tectorin are major noncollagenous proteins of the tectorial membrane, which plays a crucial role in the reception of sonic signals in the cochlea. Midkine and pleiotrophin are closely related proteins that serve as growth factors and cytokines. In mice doubly deficient in the midkine gene and pleiotrophin gene, expression of beta-tectorin mRNA was nearly abolished in the cochlea on day 1 and 7 after birth. Expression of alpha-tectorin mRNA was unaffected in the double knockout mice, and expression of beta-tectorin mRNA was not altered in mice deficient in only the midkine or pleiotrophin gene. In newborn wild-type mice, both midkine and pleiotrophin were expressed in the greater epithelial ridge of the cochlea, in which beta-tectorin mRNA was strongly expressed. These results indicate that either midkine or pleiotrophin is required for significant expression of beta-tectorin. In agreement with the view that beta-tectorin is essential for normal auditory function, mice doubly deficient in both midkine and pleiotrophin genes exhibited very severe auditory deficits. We observed that mice deficient in either midkine or pleiotrophin gene were also impaired in their auditory response, but the level of the deficit was generally low or moderate. The present finding illustrates the importance of growth factor expression in the cochlea for auditory function.
Collapse
Affiliation(s)
- Peng Zou
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | | | | | | | | | | |
Collapse
|
39
|
Dityatev A, Frischknecht R, Seidenbecher CI. Extracellular matrix and synaptic functions. Results Probl Cell Differ 2006; 43:69-97. [PMID: 17068968 DOI: 10.1007/400_025] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Comprehensive analysis of neuromuscular junction formation and recent data on synaptogenesis and long-term potentiation in the central nervous system revealed a number of extracellular matrix (ECM) molecules regulating different aspects of synaptic differentiation and function. The emerging mechanisms comprise interactions of ECM components with their cell surface receptors coupled to tyrosine kinase activities (agrin, integrin ligands, and reelin) and interactions with ion channels and transmitter receptors (Narp, tenascin-R and tenascin-C). These interactions may shape synaptic transmission and plasticity of excitatory synapses either via regulation of Ca2+ entry and postsynaptic expression of transmitter receptors or via control of GABAergic inhibition. The ECM molecules, derived from both neurons and glial cells and secreted into the extracellular space in an activity-dependent manner, may also shape synaptic plasticity through setting diffusion constraints for neurotransmitters, trophic factors and ions.
Collapse
Affiliation(s)
- Alexander Dityatev
- Institut für Neurophysiologie und Pathophysiologie, Universitätsklinikum Hamburg-Eppendorf, Germany.
| | | | | |
Collapse
|
40
|
Boerboom D, White LD, Dalle S, Courty J, Richards JS. Dominant-stable beta-catenin expression causes cell fate alterations and Wnt signaling antagonist expression in a murine granulosa cell tumor model. Cancer Res 2006; 66:1964-73. [PMID: 16488995 DOI: 10.1158/0008-5472.can-05-3493] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Wnt/beta-catenin signaling is normally involved in embryonic development and tissue homeostasis, and its misregulation leads to several forms of cancer. We have reported that misregulated Wnt/beta-catenin signaling occurs in ovarian granulosa cell tumors (GCT) and have created the Catnb(flox(ex3)/+);Amhr2(cre/+) mouse model, which expresses a dominant-stable mutant of beta-catenin in granulosa cells and develops late-onset GCT. To study the mechanisms leading to GCT development, gene expression analysis was done using microarrays comparing Catnb(flox(ex3)/+);Amhr2(cre/+) ovaries bearing pretumoral lesions with control ovaries. Overexpressed genes identified in Catnb(flox(ex3)/+);Amhr2(cre/+) ovaries included the Wnt/beta-catenin signaling antagonists Wif1, Nkd1, Dkk4, and Axin2, consistent with the induction of negative feedback loops that counteract uncontrolled Wnt/beta-catenin signaling. Expression of the antagonists was localized to cells forming the pretumoral lesions but not to normal granulosa cells. Microarray analyses also revealed the ectopic expression of bone markers, including Ibsp, Cdkn1c, Bmp4, and Tnfrsf11b, as well as neuronal/neurosecretory cell markers, such as Cck, Amph, Pitx1, and Sp5. Increased expression of the gene encoding the cytokine pleiotrophin was also found in Catnb(flox(ex3)/+);Amhr2(cre/+) ovaries and GCT but was not associated with increased serum pleiotrophin levels. In situ hybridization analyses using GCT from Catnb(flox(ex3)/+);Amhr2(cre/+) mice revealed that Wnt/beta-catenin antagonists and neuronal markers localized to a particular cell population, whereas the bone markers localized to a distinct cell type associated with areas of osseous metaplasia. Together, these results suggest that misregulated Wnt/beta-catenin signaling alters the fate of granulosa cells and that the GCT that arise in Catnb(flox(ex3)/+);Amhr2(cre/+) mice result from the clonal expansion of metaplastic cells.
Collapse
Affiliation(s)
- Derek Boerboom
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | |
Collapse
|
41
|
Tamura H, Fukada M, Fujikawa A, Noda M. Protein tyrosine phosphatase receptor type Z is involved in hippocampus-dependent memory formation through dephosphorylation at Y1105 on p190 RhoGAP. Neurosci Lett 2006; 399:33-8. [PMID: 16513268 DOI: 10.1016/j.neulet.2006.01.045] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2005] [Revised: 01/11/2006] [Accepted: 01/13/2006] [Indexed: 10/25/2022]
Abstract
Ptprz is a receptor-type protein tyrosine phosphatase predominantly expressed in the brain as a chondroitin sulfate proteoglycan. Ptprz-deficient mice exhibit an age (maturation)-dependent impairment of spatial learning in the Morris water maze test and enhancement of long-term potentiation (LTP) in the CA1 region in hippocampal slices. The enhanced LTP is canceled out by pharmacological inhibition of Rho-associated kinase (ROCK), suggesting that the lack of Ptprz causes learning impairment due to aberrant activation of ROCK. Here, we report that Ptprz-deficient mice exhibit impairments in hippocampus-dependent contextual fear memory because of abnormal tyrosine phosphorylation of p190 RhoGAP, a GTPase-activating protein (GAP) for Rho GTPase. We found that phosphorylation at Y1105, a major tyrosine phosphorylation site on p190 RhoGAP, is decreased 1h after the conditioning in the hippocampus of wild-type mice, but not of Ptprz-deficient mice. Pleiotrophin, a ligand for Ptprz, increased tyrosine phosphorylation of p190 RhoGAP in B103 neuroblastoma cells. Furthermore, Ptprz selectively dephosphorylated pY1105 of p190 RhoGAP in vitro, and the tyrosine phosphorylation at Y1105 controls p190 RhoGAP activity in vivo. These results suggest that Ptprz plays a critical role in memory formation by modulating Rho GTPase activity through dephosphorylation at Y1105 on p190 RhoGAP.
Collapse
Affiliation(s)
- Hiroshi Tamura
- Division of Molecular Neurobiology, National Institute for Basic Biology, 5-1 Higashiyama, Okazaki 444-8787, Japan
| | | | | | | |
Collapse
|
42
|
Pavlov I, Rauvala H, Taira T. Enhanced hippocampal GABAergic inhibition in mice overexpressing heparin-binding growth-associated molecule. Neuroscience 2006; 139:505-11. [PMID: 16473473 DOI: 10.1016/j.neuroscience.2005.11.070] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2004] [Revised: 11/07/2005] [Accepted: 11/20/2005] [Indexed: 11/15/2022]
Abstract
Heparin-binding growth-associated molecule is a developmentally regulated extracellular matrix protein promoting neurite outgrowth, axonal guidance and synaptogenesis. In the hippocampus, heparin-binding growth-associated molecule is expressed in an activity-dependent manner, and has been shown to suppress long-term potentiation of glutamatergic synapses in the area CA1, but the mechanisms underlying this action are unknown. One of the mechanisms by which extracellular matrix proteins might modulate fast synaptic transmission is by altering GABAergic function. Therefore, we have studied the properties of GABAA receptor-mediated inhibition in hippocampus of mutant mice overexpressing heparin-binding growth-associated molecule (heparin-binding growth-associated molecule transgenics). Under control conditions the wild-type mice have much higher level of long-term potentiation than the transgenics. However, in the absence of the GABAA receptor-mediated-inhibition a similar level of long-term potentiation is seen in both strains. In field potential recordings blockade of GABAA receptors by picrotoxin resulted in more accentuated increase in the CA1 population spike in the transgenics than in the wild-type animals. Whole-cell patch-clamp recordings revealed that when compared with the wild-type animals the transgenic mice had higher frequency of spontaneous inhibitory postsynaptic currents in CA1 pyramidal neurons. However, the frequency of action potential-independent miniature inhibitory postsynaptic currents was similar in both strains. Further, the transgenics had reduced paired-pulse depression of inhibitory postsynaptic currents, which was insensitive to the blockade of GABAB receptors in contrast to wild-type mice. The results demonstrate that the mice overexpressing heparin-binding growth-associated molecule have accentuated hippocampal GABAA receptor-mediated inhibition, which in turn may explain the lowered predisposition of glutamatergic synapses to undergo plastic changes in these animals. Thus, our findings suggest a mechanism by which heparin-binding growth-associated molecule can regulate synaptic plasticity.
Collapse
Affiliation(s)
- I Pavlov
- Neuroscience Center and Department of Biological and Environmental Sciences, University of Helsinki, P.O. Box 65, Viikinkaari 1, FIN-00014 Helsinki, Finland
| | | | | |
Collapse
|
43
|
Meighan SE, Meighan PC, Choudhury P, Davis CJ, Olson ML, Zornes PA, Wright JW, Harding JW. Effects of extracellular matrix-degrading proteases matrix metalloproteinases 3 and 9 on spatial learning and synaptic plasticity. J Neurochem 2006; 96:1227-41. [PMID: 16464240 DOI: 10.1111/j.1471-4159.2005.03565.x] [Citation(s) in RCA: 218] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Rats learning the Morris water maze exhibit hippocampal changes in synaptic morphology and physiology that manifest as altered synaptic efficacy. Learning requires structural changes in the synapse, and multiple cell adhesion molecules appear to participate. The activity of these cell adhesion molecules is, in large part, dependent on their interaction with the extracellular matrix (ECM). Given that matrix metalloproteinases (MMPs) are responsible for transient alterations in the ECM, we predicted that MMP function is critical for hippocampal-dependent learning. In support of this, it was observed that hippocampal MMP-3 and -9 increased transiently during water maze acquisition as assessed by western blotting and mRNA analysis. The ability of the NMDA receptor channel blocker MK801 to attenuate these changes indicated that the transient MMP changes were in large part dependent upon NMDA receptor activation. Furthermore, inhibition of MMP activity with MMP-3 and -9 antisense oligonucleotides and/or MMP inhibitor FN-439 altered long-term potentiation and prevented acquisition in the Morris water maze. The learning-dependent MMP alterations were shown to modify the stability of the actin-binding protein cortactin, which plays an essential role in regulating the dendritic cytoskeleton and synaptic efficiency. Together these results indicate that changes in MMP function are critical to synaptic plasticity and hippocampal-dependent learning.
Collapse
Affiliation(s)
- Starla E Meighan
- Department of Veterinary Comparative Anatomy, Pharmacology and Physiology, Washington State University, Pullman, Washington 99164, USA.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Taravini IRE, Ferrario JE, Delbe J, Ginestet L, Debeir T, Courty J, Murer MG, Gershanik OS, Raisman-Vozari R. Immunodetection of heparin-binding growth associated molecule (pleiotrophin) in striatal interneurons. Brain Res 2005; 1066:196-200. [PMID: 16325783 DOI: 10.1016/j.brainres.2005.10.055] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2005] [Revised: 10/03/2005] [Accepted: 10/11/2005] [Indexed: 11/21/2022]
Abstract
Pleiotrophin (PTN), a developmentally-regulated trophic factor, is over-expressed in the striatum of parkinsonian rats. Because striatal PTN can provide trophic support to dopamine neurons, we identified the cellular types containing PTN in the striatum of adult rats. By means of fluorescent double-immunolabeling, we found PTN to co-localize with a neuronal nuclei marker but not with glial fibrillary acidic protein. The number, distribution, and morphology of the PTN-immunolabeled cells suggested that they were interneurons. Further double-immunolabeling studies ruled out PTN localization to calretinin- and parvalbumin-containing interneurons. Instead, approximately 40% of the PTN-immunolabeled neurons contained nitric oxide synthase or somatostatin and approximately 60% expressed the vesicular acetylcholine transporter, supporting that they were GABAergic nitric oxide synthase/somatostatin-containing and cholinergic interneurons. Further work is necessary to determine if PTN from striatal interneurons can provide trophic support to dopamine neurons.
Collapse
Affiliation(s)
- Irene R E Taravini
- Laboratorio de Parkinson Experimental, Instituto de Investigaciones Farmacológicas, CONICET, Junín 956, 5 Piso, C1113AAD Buenos Aires, Argentina.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Arighi E, Borrello MG, Sariola H. RET tyrosine kinase signaling in development and cancer. Cytokine Growth Factor Rev 2005; 16:441-67. [PMID: 15982921 DOI: 10.1016/j.cytogfr.2005.05.010] [Citation(s) in RCA: 319] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
The variety of diseases caused by mutations in RET receptor tyrosine kinase provides a classic example of phenotypic heterogeneity. Gain-of-function mutations of RET are associated with human cancer. Gene rearrangements juxtaposing the tyrosine kinase domain to heterologous gene partners have been found in sporadic papillary carcinomas of the thyroid (PTC). These rearrangements generate chimeric RET/PTC oncogenes. In the germline, point mutations of RET are responsible for multiple endocrine neoplasia type 2 (MEN 2A and 2B) and familial medullary thyroid carcinoma (FMTC). Both MEN 2 mutations and PTC gene rearrangements potentiate the intrinsic tyrosine kinase activity of RET and, ultimately, activate the RET downstream targets. Loss-of-function mutations of RET cause Hirschsprung's disease (HSCR) or colonic aganglionosis. A deeper understanding of the molecular signaling of normal versus abnormal RET activity in cancer will enable the development of potential new treatments for patients with sporadic and inherited thyroid cancer or MEN 2 syndrome. We now review the role and mechanisms of RET signaling in development and carcinogenesis.
Collapse
Affiliation(s)
- Elena Arighi
- Developmental Biology, Institute of Biomedicine, Biomedicum Helsinki, University of Helsinki, Finland
| | | | | |
Collapse
|
46
|
Herradon G, Ezquerra L, Nguyen T, Silos-Santiago I, Deuel TF. Midkine regulates pleiotrophin organ-specific gene expression: evidence for transcriptional regulation and functional redundancy within the pleiotrophin/midkine developmental gene family. Biochem Biophys Res Commun 2005; 333:714-21. [PMID: 15985215 DOI: 10.1016/j.bbrc.2005.05.160] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2005] [Accepted: 05/23/2005] [Indexed: 11/15/2022]
Abstract
Midkine (MK) and the highly related cytokine pleiotrophin (PTN) constitute the PTN/MK developmental gene family. The Mk and Ptn genes are essential for normal development of the catecholamine and renin-angiotensin pathways and the synthesis of different collagens. It is not known whether the Ptn and Mk genes regulate each other or whether PTN and MK are functionally redundant in development. We have now compared the levels of expression of Ptn and Mk in genetically deficient Mk -/- and Ptn -/- mice and found highly significant increases in Ptn gene expression in spinal cord, dorsal root ganglia, eye, heart, aorta, bladder, and urethra, but not in brain, bone marrow, testis, and lung of Mk -/- mice compared with wild type mice; a remarkable approximately 230-fold increase in Ptn expression levels was found in heart of Mk -/- mice and highly significant but lesser increases were found in six other organs. Differences in levels of Mk gene expression in Ptn -/- mice could not be detected in any of the organs tested. The data demonstrate that MK regulates Ptn gene expression with a high degree of organ specificity, suggesting that Ptn gene expression follows Mk gene expression in development, that the increase in Ptn gene expression is compensatory for the absence of MK in Mk -/- mice, that PTN and MK share a high degree of functional redundancy, and that MK may be very important in the development of heart in mouse.
Collapse
Affiliation(s)
- Gonzalo Herradon
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | | | |
Collapse
|
47
|
Hayashi N, Oohira A, Miyata S. Synaptic localization of receptor-type protein tyrosine phosphatase zeta/beta in the cerebral and hippocampal neurons of adult rats. Brain Res 2005; 1050:163-9. [PMID: 15982644 DOI: 10.1016/j.brainres.2005.05.047] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2004] [Revised: 05/16/2005] [Accepted: 05/18/2005] [Indexed: 11/30/2022]
Abstract
Receptor-type protein tyrosine phosphatase (RPTP) zeta/beta is a nervous tissue-specific chondroitin sulfate proteoglycan. In this study, we investigated the immunohistochemical localization of RPTPzeta/beta in adult rat cerebral cortex and hippocampus at light and electron microscopic levels. Double labeling immunofluorescence microscopy revealed that the immunoreactivity of RPTPzeta/beta was observed at MAP2-positive dendrites and PSD-95-positive spines of pyramidal neurons in the cerebral cortex and hippocampus. Electron microscopic observation demonstrated a strong immunoreactivity of RPTPzeta/beta at the postsynaptic membrane of dendritic spines and shafts, and its moderate immunoreactivity at the dendritic membrane. In cultured cortical neurons, the immunoreactivity of RPTPzeta/beta was observed at some of PSD-95-positive spines. These results demonstrate that RPTPzeta/beta is localized mainly at the postsynaptic membrane of pyramidal neurons in adult cerebral cortex and hippocampus.
Collapse
Affiliation(s)
- Noriko Hayashi
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Kyoto 606-8585, Japan
| | | | | |
Collapse
|
48
|
Niisato K, Fujikawa A, Komai S, Shintani T, Watanabe E, Sakaguchi G, Katsuura G, Manabe T, Noda M. Age-dependent enhancement of hippocampal long-term potentiation and impairment of spatial learning through the Rho-associated kinase pathway in protein tyrosine phosphatase receptor type Z-deficient mice. J Neurosci 2005; 25:1081-8. [PMID: 15689543 PMCID: PMC6725950 DOI: 10.1523/jneurosci.2565.04.2005] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Although protein tyrosine phosphatases (PTPs) are expressed abundantly in the brain, their roles in synaptic plasticity have not been well elucidated. In this study, we have examined the physiological functions of Ptprz, which is a receptor-type PTP expressed predominantly in the brain as a chondroitin sulfate proteoglycan. We have examined phenotypes of mutant mice deficient in Ptprz using electrophysiological, pharmacological, and behavioral approaches. Mutant mice exhibit enhanced long-term potentiation (LTP) in the CA1 region of hippocampal slices and impaired spatial learning abilities in an age-dependent manner: young adult (<10 weeks old) mutant mice show normal LTP and learning abilities in the Morris water maze task, whereas adult (>13 weeks old) mutant mice exhibit enhanced LTP and impairment in the task. The enhanced LTP is specifically canceled out by pharmacological inhibition of Rho-associated kinase (ROCK), a major downstream effector of Rho. These findings suggest that the lack of Ptprz leads to aberrant activation of ROCK and resultantly to enhanced LTP in the slice and learning impairments in the animal.
Collapse
Affiliation(s)
- Kazue Niisato
- Division of Neuronal Network, Department of Basic Medical Sciences, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Raulo E, Tumova S, Pavlov I, Pekkanen M, Hienola A, Klankki E, Kalkkinen N, Taira T, Kilpelaïnen I, Rauvala H. The two thrombospondin type I repeat domains of the heparin-binding growth-associated molecule bind to heparin/heparan sulfate and regulate neurite extension and plasticity in hippocampal neurons. J Biol Chem 2005; 280:41576-83. [PMID: 16155004 DOI: 10.1074/jbc.m506457200] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
HB-GAM (heparin-binding growth-associated molecule, also designated as pleiotrophin) and midkine form a two-member family of extracellular matrix proteins that bind tightly to sulfated carbohydrate structures such as heparan sulfate. These proteins are used by developing neurons as extracellular cues in axonal growth and guidance. HB-GAM was recently reported to enhance differentiation of neural stem cells. Based on the solution structure of HB-GAM, we have recently shown that HB-GAM consists of two beta-sheet domains flanked by flexible lysine-rich N- and C-terminal tails with no apparent structure. These domains are homologous to thrombospondin type I repeats present in numerous extracellular proteins that interact with the cell surface. Our findings showed that the two beta-sheet domains fold independently. We showed that the domains (but not the lysine-rich tails) in HB-GAM are required and sufficient for interaction with hippocampal neurons. The individual domains bind heparan sulfate weakly and fail to produce significant biological effects in neurite outgrowth and long term potentiation assays. The amino acids in the linker region joining the two domains may be replaced with glycines with no effect on protein function. These results suggest a co-operative action of the two beta-sheet domains in the biologically relevant interaction with neuron surface heparan sulfate.
Collapse
Affiliation(s)
- Erkki Raulo
- Neuroscience Center, University of Helsinki, Helsinki FIN-00014, Finland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Matsuyama S, Doe N, Kurihara N, Tanizawa K, Kuroda S, Iso H, Horie M. Spatial learning of mice lacking a neuron-specific epidermal growth factor family protein, NELL2. J Pharmacol Sci 2005; 98:239-43. [PMID: 15988128 DOI: 10.1254/jphs.fp0050211] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
NELL2 is a neuron-specific thrombospondin-1-like extracellular protein containing six epidermal growth factor-like domains. We previously disrupted the NELL2 gene in mice by gene targeting and showed that long-term potentiation is enhanced in vivo in the dentate gyrus of NELL2-deficient mice. To further elucidate the physiological roles of NELL2, we performed a behavioral characterization of NELL2(-/-) and their heterozygous control mice. NELL2-deficient mice exhibited learning impairment in the Morris water maze task. However, we observed no difference in passive avoidance learning between NELL2(-/-) and NELL2(+/-) mice. These observations suggest that NELL2 plays an important role in hippocampus-dependent spatial learning and that emotional learning does not depend critically on NELL2.
Collapse
Affiliation(s)
- Shogo Matsuyama
- Division of Molecular Pharmacology and Pharmacogenomics, Department of Genome Sciences, Kobe University Graduate School of Medicine, Hyogo, Japan
| | | | | | | | | | | | | |
Collapse
|