1
|
Li Q, Morrill NK, Moerman-Herzog AM, Barger SW, Joly-Amado A, Peters M, Soueidan H, Diemler C, Prabhudeva S, Weeber EJ, Nash KR. Central repeat fragment of reelin leads to active reelin intracellular signaling and rescues cognitive deficits in a mouse model of reelin deficiency. Cell Signal 2023:110763. [PMID: 37315752 DOI: 10.1016/j.cellsig.2023.110763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/19/2023] [Accepted: 06/07/2023] [Indexed: 06/16/2023]
Abstract
Reelin and its receptor, ApoER2, play important roles in prenatal brain development and postnatally in synaptic plasticity, learning, and memory. Previous reports suggest that reelin's central fragment binds to ApoER2 and receptor clustering is involved in subsequent intracellular signaling. However, limitations of currently available assays have not established cellular evidence of ApoER2 clustering upon binding of the central reelin fragment. In the present study, we developed a novel, cell-based assay of ApoER2 dimerization using a "split-luciferase" approach. Specifically, cells were co-transfected with one recombinant ApoER2 receptor fused to the N-terminus of luciferase and one ApoER2 receptor fused to the C-terminus of luciferase. Using this assay, we directly observed basal ApoER2 dimerization/clustering in transfected HEK293T cells and, significantly, an increase in ApoER2 clustering in response to that central fragment of reelin. Furthermore, the central fragment of reelin activated intracellular signal transduction of ApoER2, indicated by increased levels of phosphorylation of Dab1, ERK1/2, and Akt in primary cortical neurons. Functionally, we were able to demonstrate that injection of the central fragment of reelin rescued phenotypic deficits observed in the heterozygous reeler mouse. These data are the first to test the hypothesis that the central fragment of reelin contributes to facilitating the reelin intracellular signaling pathway through receptor clustering.
Collapse
Affiliation(s)
- Qingyou Li
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Nicole K Morrill
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Andréa M Moerman-Herzog
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States of America
| | - Steven W Barger
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States of America
| | - Aurelie Joly-Amado
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Melinda Peters
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Hana Soueidan
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Cory Diemler
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Sahana Prabhudeva
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Edwin J Weeber
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Kevin R Nash
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA.
| |
Collapse
|
2
|
Lopez-Font I, Lennol MP, Iborra-Lazaro G, Zetterberg H, Blennow K, Sáez-Valero J. Altered Balance of Reelin Proteolytic Fragments in the Cerebrospinal Fluid of Alzheimer's Disease Patients. Int J Mol Sci 2022; 23:7522. [PMID: 35886870 PMCID: PMC9318932 DOI: 10.3390/ijms23147522] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/28/2022] [Accepted: 07/01/2022] [Indexed: 02/01/2023] Open
Abstract
Reelin binds to the apolipoprotein E receptor apoER2 to activate an intracellular signaling cascade. The proteolytic cleavage of reelin follows receptor binding but can also occur independently of its binding to receptors. This study assesses whether reelin proteolytic fragments are differentially affected in the cerebrospinal fluid (CSF) of Alzheimer's disease (AD) subjects. CSF reelin species were analyzed by Western blotting, employing antibodies against the N- and C-terminal domains. In AD patients, we found a decrease in the 420 kDa full-length reelin compared with controls. In these patients, we also found an increase in the N-terminal 310 kDa fragment resulting from the cleavage at the so-called C-t site, whereas the 180 kDa fragment originated from the N-t site remained unchanged. Regarding the C-terminal proteolytic fragments, the 100 kDa fragment resulting from the cleavage at the C-t site also displayed increased levels, whilst the one resulting from the N-t site, the 250 kDa fragment, decreased. We also detected the presence of an aberrant reelin species with a molecular mass of around 500 kDa present in AD samples (34 of 43 cases), while it was absent in the 14 control cases analyzed. These 500 kDa species were only immunoreactive to N-terminal antibodies. We validated the occurrence of these aberrant reelin species in an Aβ42-treated reelin-overexpressing cell model. When we compared the AD samples from APOE genotype subgroups, we only found minor differences in the levels of reelin fragments associated to the APOE genotype, but interestingly, the levels of fragments of apoER2 were lower in APOE ε4 carriers with regards to APOE ε3/ε3. The altered proportion of reelin/apoER2 fragments and the occurrence of reelin aberrant species suggest a complex regulation of the reelin signaling pathway, which results impaired in AD subjects.
Collapse
Affiliation(s)
- Inmaculada Lopez-Font
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, 03550 Sant Joan d’Alacant, Spain; (M.P.L.); (G.I.-L.)
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 03550 Sant Joan d’Alacant, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - Matthew P. Lennol
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, 03550 Sant Joan d’Alacant, Spain; (M.P.L.); (G.I.-L.)
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 03550 Sant Joan d’Alacant, Spain
| | - Guillermo Iborra-Lazaro
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, 03550 Sant Joan d’Alacant, Spain; (M.P.L.); (G.I.-L.)
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 03550 Sant Joan d’Alacant, Spain
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 413 45 Mölndal, Sweden; (H.Z.); (K.B.)
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, 413 90 Mölndal, Sweden
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London WC1E 6BT, UK
- UK Dementia Research Institute at UCL, London WC1E 6BT, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 413 45 Mölndal, Sweden; (H.Z.); (K.B.)
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, 413 90 Mölndal, Sweden
| | - Javier Sáez-Valero
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, 03550 Sant Joan d’Alacant, Spain; (M.P.L.); (G.I.-L.)
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 03550 Sant Joan d’Alacant, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| |
Collapse
|
3
|
Cuchillo-Ibañez I, Lennol MP, Escamilla S, Mata-Balaguer T, Valverde-Vozmediano L, Lopez-Font I, Ferrer I, Sáez-Valero J. The apolipoprotein receptor LRP3 compromises APP levels. Alzheimers Res Ther 2021; 13:181. [PMID: 34727970 PMCID: PMC8565065 DOI: 10.1186/s13195-021-00921-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 10/19/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND Members of the low-density lipoprotein (LDL) receptor family are involved in endocytosis and in transducing signals, but also in amyloid precursor protein (APP) processing and β-amyloid secretion. ApoER2/LRP8 is a member of this family with key roles in synaptic plasticity in the adult brain. ApoER2 is cleaved after the binding of its ligand, the reelin protein, generating an intracellular domain (ApoER2-ICD) that modulates reelin gene transcription itself. We have analyzed whether ApoER2-ICD is able to regulate the expression of other LDL receptors, and we focused on LRP3, the most unknown member of this family. We analyzed LRP3 expression in middle-aged individuals (MA) and in cases with Alzheimer's disease (AD)-related pathology, and the relation of LRP3 with APP. METHODS The effects of full-length ApoER2 and ApoER2-ICD overexpression on protein levels, in the presence of recombinant reelin or Aβ42 peptide, were evaluated by microarray, qRT-PCRs, and western blots in SH-SY5Y cells. LRP3 expression was analyzed in human frontal cortex extracts from MA subjects (mean age 51.8±4.8 years) and AD-related pathology subjects [Braak neurofibrillary tangle stages I-II, 68.4±8.8 years; III-IV, 80.4 ± 8.8 years; V-VI, 76.5±9.7 years] by qRT-PCRs and western blot; LRP3 interaction with other proteins was assessed by immunoprecipitation. In CHO cells overexpressing LRP3, protein levels of full-length APP and fragments were evaluated by western blots. Chloroquine was employed to block the lysosomal/autophagy function. RESULTS We have identified that ApoER2 overexpression increases LRP3 expression, also after reelin stimulation of ApoER2 signaling. The same occurred following ApoER2-ICD overexpression. In extracts from subjects with AD-related pathology, the levels of LRP3 mRNA and protein were lower than those in MA subjects. Interestingly, LRP3 transfection in CHO-PS70 cells induced a decrease of full-length APP levels and APP-CTF, particularly in the membrane fraction. In cell supernatants, levels of APP fragments from the amyloidogenic (sAPPα) or non-amyloidogenic (sAPPβ) pathways, as well as Aβ peptides, were drastically reduced with respect to mock-transfected cells. The inhibitor of lysosomal/autophagy function, chloroquine, significantly increased full-length APP, APP-CTF, and sAPPα levels. CONCLUSIONS ApoER2/reelin signaling regulates LRP3 expression, whose levels are affected in AD; LRP3 is involved in the regulation of APP levels.
Collapse
Affiliation(s)
- Inmaculada Cuchillo-Ibañez
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández de Elche-CSIC, Sant Joan d'Alacant, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain.
| | - Matthew P Lennol
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández de Elche-CSIC, Sant Joan d'Alacant, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Sergio Escamilla
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández de Elche-CSIC, Sant Joan d'Alacant, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Trinidad Mata-Balaguer
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández de Elche-CSIC, Sant Joan d'Alacant, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Lucía Valverde-Vozmediano
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández de Elche-CSIC, Sant Joan d'Alacant, Spain
| | - Inmaculada Lopez-Font
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández de Elche-CSIC, Sant Joan d'Alacant, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
| | - Isidro Ferrer
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Instituto de Neuropatología, Hospital Universitario de Bellvitge, Universidad de Barcelona, Hospitalet de Llobregat, Barcelona, Spain
| | - Javier Sáez-Valero
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández de Elche-CSIC, Sant Joan d'Alacant, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain.
| |
Collapse
|
4
|
Turk LS, Kuang X, Dal Pozzo V, Patel K, Chen M, Huynh K, Currie MJ, Mitchell D, Dobson RCJ, D'Arcangelo G, Dai W, Comoletti D. The structure-function relationship of a signaling-competent, dimeric Reelin fragment. Structure 2021; 29:1156-1170.e6. [PMID: 34089653 DOI: 10.1016/j.str.2021.05.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/26/2021] [Accepted: 05/14/2021] [Indexed: 01/21/2023]
Abstract
Reelin operates through canonical and non-canonical pathways that mediate several aspects of brain development and function. Reelin's dimeric central fragment (CF), generated through proteolytic cleavage, is required for the lipoprotein-receptor-dependent canonical pathway activation. Here, we analyze the signaling properties of a variety of Reelin fragments and measure the differential binding affinities of monomeric and dimeric CF fragments to lipoprotein receptors to investigate the mode of canonical signal activation. We also present the cryoelectron tomography-solved dimeric structure of Reelin CF and support it using several other biophysical techniques. Our findings suggest that Reelin CF forms a covalent parallel dimer with some degree of flexibility between the two protein chains. As a result of this conformation, Reelin binds to lipoprotein receptors in a manner inaccessible to its monomeric form and is capable of stimulating canonical pathway signaling.
Collapse
Affiliation(s)
- Liam S Turk
- Child Health Institute of New Jersey, New Brunswick, NJ 08901, USA; Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA; School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Xuyuan Kuang
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA; Institute for Quantitative Biomedicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA; Department of Hyperbaric Oxygen, Central South University, Changsha, Hunan Province, China
| | - Valentina Dal Pozzo
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Khush Patel
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Muyuan Chen
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kevin Huynh
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA; Institute for Quantitative Biomedicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Michael J Currie
- Biomolecular Interactions Centre and School of Biological Sciences, University of Canterbury, Christchurch 8041, New Zealand
| | - Daniel Mitchell
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Renwick C J Dobson
- Biomolecular Interactions Centre and School of Biological Sciences, University of Canterbury, Christchurch 8041, New Zealand; Bio21 Molecular Science and Biotechnology Institute, Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Gabriella D'Arcangelo
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Wei Dai
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA; Institute for Quantitative Biomedicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA.
| | - Davide Comoletti
- Child Health Institute of New Jersey, New Brunswick, NJ 08901, USA; Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA; School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand.
| |
Collapse
|
5
|
Ishii K, Kohno T, Hattori M. Differential binding of anti-Reelin monoclonal antibodies reveals the characteristics of Reelin protein under various conditions. Biochem Biophys Res Commun 2019; 514:815-820. [PMID: 31079931 DOI: 10.1016/j.bbrc.2019.05.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 05/01/2019] [Indexed: 01/09/2023]
Abstract
Reelin is a large secreted protein that is essential for the development and function of the central nervous system. Dimerization and/or oligomerization is required for its biological activity, but the underlying mechanism is not fully understood. There are several widely used anti-Reelin antibodies and we noticed that their reactivity to monomeric or dimeric Reelin protein is different. We also found that their reactivity to Reelin in the solution or in fixed brain tissues also differs. Our results provide the information regarding how the N-terminal region of Reelin folds and contributes to the formation of higher order structure. We also provide a caveat that appropriate use of anti-Reelin antibody is necessary for quantitative analyses.
Collapse
Affiliation(s)
- Keisuke Ishii
- Department of Biomedical Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi, 467-8603, Japan
| | - Takao Kohno
- Department of Biomedical Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi, 467-8603, Japan
| | - Mitsuharu Hattori
- Department of Biomedical Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi, 467-8603, Japan.
| |
Collapse
|
6
|
Dlugosz P, Tresky R, Nimpf J. Differential Action of Reelin on Oligomerization of ApoER2 and VLDL Receptor in HEK293 Cells Assessed by Time-Resolved Anisotropy and Fluorescence Lifetime Imaging Microscopy. Front Mol Neurosci 2019; 12:53. [PMID: 30873003 PMCID: PMC6403468 DOI: 10.3389/fnmol.2019.00053] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 02/12/2019] [Indexed: 01/12/2023] Open
Abstract
The canonical Reelin signaling cascade regulates correct neuronal layering during embryonic brain development. Details of this pathway are still not fully understood since the participating components are highly variable and create a complex mixture of interacting molecules. Reelin is proteolytically processed resulting in five different fragments some of which carrying the binding site for two different but highly homologous receptors, apolipoprotein E receptor 2 (ApoER2) and very low density lipoprotein receptor (VLDLR). The receptors are expressed in different variants in different areas of the developing brain. Binding of Reelin and its central fragment to the receptors results in phosphorylation of the intracellular adapter disabled-1 (Dab1) in neurons. Here, we studied the changes of the arrangement of the receptors upon Reelin binding and its central fragment at the molecular level in human embryonic kidney 293 (HEK293) cells by time-resolved anisotropy and fluorescence lifetime imaging microscopy (FLIM). In the off-state of the pathway ApoER2 and VLDLR form homo or hetero-di/oligomers. Upon binding of full length Reelin ApoER2 and VLDLR homo-oligomers are rearranged to higher order receptor clusters which leads to Dab1 phosphorylation. When the central fragment of Reelin binds to the receptors the cluster size of homo-oligomers is not affected and Dab1 is not phosphorylated. Hetero-oligomerization, however, can be induced, but does not lead to Dab1 phosphorylation. Cells expressing only ApoER2 or VLDLR change their shape when stimulated with the central fragment. Cells expressing ApoER2 produce filopodia/lamellipodia and cell size increases, whereas VLDLR-expressing cells decrease in size. These findings demonstrate that the primary event in the canonical Reelin pathway is the rearrangement of preformed receptor homo-oligomers to higher order clusters. In addition the possibility of yet another signaling mechanism which is mediated by the central Reelin fragment independent of Dab1 phosphorylation became apparent.
Collapse
Affiliation(s)
- Paula Dlugosz
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University Vienna, Vienna, Austria
| | - Roland Tresky
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University Vienna, Vienna, Austria
| | - Johannes Nimpf
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University Vienna, Vienna, Austria
| |
Collapse
|
7
|
Mata‐Balaguer T, Cuchillo‐Ibañez I, Calero M, Ferrer I, Sáez‐Valero J. Decreased generation of C‐terminal fragments of ApoER2 and increased reelin expression in Alzheimer's disease. FASEB J 2018; 32:3536-3546. [DOI: 10.1096/fj.201700736rr] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Trinidad Mata‐Balaguer
- Instituto de Neurociencias de AlicanteUniversidad Miguel Hernández‐Consejo Superior de Investigaciones Cientificas (CSIC)Sant Joan d'AlacantAlicanteSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| | - Inmaculada Cuchillo‐Ibañez
- Instituto de Neurociencias de AlicanteUniversidad Miguel Hernández‐Consejo Superior de Investigaciones Cientificas (CSIC)Sant Joan d'AlacantAlicanteSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| | - Miguel Calero
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
- Alzheimer Disease Research UnitCentro Nacional de Investigación en Enfermedades Neurológicas (CIEN) FoundationQueen Sofia Foundation Alzheimer CenterMadridSpain
- Chronic Disease ProgrammeCarlos III Institute of HealthMadridSpain
| | - Isidro Ferrer
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
- Instituto de NeuropatologíaHospital Universitario de BellvitgeUniversidad de BarcelonaHospitalet de LlobregatBarcelonaSpain
| | - Javier Sáez‐Valero
- Instituto de Neurociencias de AlicanteUniversidad Miguel Hernández‐Consejo Superior de Investigaciones Cientificas (CSIC)Sant Joan d'AlacantAlicanteSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| |
Collapse
|
8
|
Yakovlev S, Medved L. Interaction of Fibrin with the Very Low-Density Lipoprotein (VLDL) Receptor: Further Characterization and Localization of the VLDL Receptor-Binding Site in Fibrin βN-Domains. Biochemistry 2017; 56:2518-2528. [PMID: 28437098 DOI: 10.1021/acs.biochem.7b00087] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Our recent study revealed that fibrin and the very low-density lipoprotein receptor (VLDLR) interact with each other through a pair of fibrin βN-domains and CR domains of the receptor and this interaction promotes transendothelial migration of leukocytes and thereby inflammation. The major objectives of this study were to further clarify the molecular mechanism of fibrin-VLDLR interaction and to identify amino acid residues in the βN-domains involved in this interaction. Our binding experiments with the (β15-66)2 fragment, which corresponds to a pair of fibrin βN-domains, and the VLDLR(1-8) fragment, consisting of eight CR domains of VLDLR, revealed that interaction between them strongly depends on ionic strength and chemical modification of all Lys or Arg residues in (β15-66)2 results in abrogation of this interaction. To identify which of these residues are involved in the interaction, we mutated all Lys or Arg residues in each of the three positively charged Lys/Arg clusters of the (β15-66)2 fragment, as well as single Arg17 and Arg30, and tested the affinity of the mutants obtained for VLDLR(1-8) by an enzyme-linked immunosorbent assay and surface plasmon resonance. The experiments revealed that the second and third Lys/Arg clusters make the major contribution to this interaction while the contribution of the first cluster is moderate. The results obtained suggest that interaction between fibrin and the VLDL receptor employs the "double-Lys/Arg" recognition mode previously proposed for the interaction of the LDL receptor family members with their ligands. They also provide valuable information for the development of highly specific peptide-based inhibitors of fibrin-VLDLR interaction.
Collapse
Affiliation(s)
- Sergiy Yakovlev
- Center for Vascular and Inflammatory Diseases and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine , Baltimore, Maryland 21201, United States
| | - Leonid Medved
- Center for Vascular and Inflammatory Diseases and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine , Baltimore, Maryland 21201, United States
| |
Collapse
|
9
|
Guénette S, Strecker P, Kins S. APP Protein Family Signaling at the Synapse: Insights from Intracellular APP-Binding Proteins. Front Mol Neurosci 2017; 10:87. [PMID: 28424586 PMCID: PMC5371672 DOI: 10.3389/fnmol.2017.00087] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 03/13/2017] [Indexed: 12/17/2022] Open
Abstract
Understanding the molecular mechanisms underlying amyloid precursor protein family (APP/APP-like proteins, APLP) function in the nervous system can be achieved by studying the APP/APLP interactome. In this review article, we focused on intracellular APP interacting proteins that bind the YENPTY internalization motif located in the last 15 amino acids of the C-terminal region. These proteins, which include X11/Munc-18-interacting proteins (Mints) and FE65/FE65Ls, represent APP cytosolic binding partners exhibiting different neuronal functions. A comparison of FE65 and APP family member mutant mice revealed a shared function for APP/FE65 protein family members in neurogenesis and neuronal positioning. Accumulating evidence also supports a role for membrane-associated APP/APLP proteins in synapse formation and function. Therefore, it is tempting to speculate that APP/APLP C-terminal interacting proteins transmit APP/APLP-dependent signals at the synapse. Herein, we compare our current knowledge of the synaptic phenotypes of APP/APLP mutant mice with those of mice lacking different APP/APLP interaction partners and discuss the possible downstream effects of APP-dependent FE65/FE65L or X11/Mint signaling on synaptic vesicle release, synaptic morphology and function. Given that the role of X11/Mint proteins at the synapse is well-established, we propose a model highlighting the role of FE65 protein family members for transduction of APP/APLP physiological function at the synapse.
Collapse
Affiliation(s)
| | - Paul Strecker
- Department of Biology, Division of Human Biology, University of KaiserslauternKaiserslautern, Germany
| | - Stefan Kins
- Department of Biology, Division of Human Biology, University of KaiserslauternKaiserslautern, Germany
| |
Collapse
|
10
|
Pohlkamp T, Wasser CR, Herz J. Functional Roles of the Interaction of APP and Lipoprotein Receptors. Front Mol Neurosci 2017; 10:54. [PMID: 28298885 PMCID: PMC5331069 DOI: 10.3389/fnmol.2017.00054] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 02/16/2017] [Indexed: 11/24/2022] Open
Abstract
The biological fates of the key initiator of Alzheimer’s disease (AD), the amyloid precursor protein (APP), and a family of lipoprotein receptors, the low-density lipoprotein (LDL) receptor-related proteins (LRPs) and their molecular roles in the neurodegenerative disease process are inseparably interwoven. Not only does APP bind tightly to the extracellular domains (ECDs) of several members of the LRP group, their intracellular portions are also connected through scaffolds like the one established by FE65 proteins and through interactions with adaptor proteins such as X11/Mint and Dab1. Moreover, the ECDs of APP and LRPs share common ligands, most notably Reelin, a regulator of neuronal migration during embryonic development and modulator of synaptic transmission in the adult brain, and Agrin, another signaling protein which is essential for the formation and maintenance of the neuromuscular junction (NMJ) and which likely also has critical, though at this time less well defined, roles for the regulation of central synapses. Furthermore, the major independent risk factors for AD, Apolipoprotein (Apo) E and ApoJ/Clusterin, are lipoprotein ligands for LRPs. Receptors and ligands mutually influence their intracellular trafficking and thereby the functions and abilities of neurons and the blood-brain-barrier to turn over and remove the pathological product of APP, the amyloid-β peptide. This article will review and summarize the molecular mechanisms that are shared by APP and LRPs and discuss their relative contributions to AD.
Collapse
Affiliation(s)
- Theresa Pohlkamp
- Department of Molecular Genetics, UT Southwestern Medical CenterDallas, TX, USA; Center for Translational Neurodegeneration Research, UT Southwestern Medical CenterDallas, TX, USA
| | - Catherine R Wasser
- Department of Molecular Genetics, UT Southwestern Medical CenterDallas, TX, USA; Center for Translational Neurodegeneration Research, UT Southwestern Medical CenterDallas, TX, USA
| | - Joachim Herz
- Department of Molecular Genetics, UT Southwestern Medical CenterDallas, TX, USA; Center for Translational Neurodegeneration Research, UT Southwestern Medical CenterDallas, TX, USA; Department of Neuroscience, UT Southwestern Medical CenterDallas, TX, USA; Department of Neurology and Neurotherapeutics, UT Southwestern Medical CenterDallas, TX, USA
| |
Collapse
|
11
|
Bock HH, May P. Canonical and Non-canonical Reelin Signaling. Front Cell Neurosci 2016; 10:166. [PMID: 27445693 PMCID: PMC4928174 DOI: 10.3389/fncel.2016.00166] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 06/08/2016] [Indexed: 12/11/2022] Open
Abstract
Reelin is a large secreted glycoprotein that is essential for correct neuronal positioning during neurodevelopment and is important for synaptic plasticity in the mature brain. Moreover, Reelin is expressed in many extraneuronal tissues; yet the roles of peripheral Reelin are largely unknown. In the brain, many of Reelin's functions are mediated by a molecular signaling cascade that involves two lipoprotein receptors, apolipoprotein E receptor-2 (Apoer2) and very low density-lipoprotein receptor (Vldlr), the neuronal phosphoprotein Disabled-1 (Dab1), and members of the Src family of protein tyrosine kinases as crucial elements. This core signaling pathway in turn modulates the activity of adaptor proteins and downstream protein kinase cascades, many of which target the neuronal cytoskeleton. However, additional Reelin-binding receptors have been postulated or described, either as coreceptors that are essential for the activation of the "canonical" Reelin signaling cascade involving Apoer2/Vldlr and Dab1, or as receptors that activate alternative or additional signaling pathways. Here we will give an overview of canonical and alternative Reelin signaling pathways, molecular mechanisms involved, and their potential physiological roles in the context of different biological settings.
Collapse
Affiliation(s)
- Hans H Bock
- Clinic of Gastroenterology and Hepatology, Heinrich-Heine-University Düsseldorf Düsseldorf, Germany
| | - Petra May
- Clinic of Gastroenterology and Hepatology, Heinrich-Heine-University Düsseldorf Düsseldorf, Germany
| |
Collapse
|
12
|
Cuchillo-Ibañez I, Balmaceda V, Mata-Balaguer T, Lopez-Font I, Sáez-Valero J. Reelin in Alzheimer’s Disease, Increased Levels but Impaired Signaling: When More is Less. J Alzheimers Dis 2016; 52:403-16. [DOI: 10.3233/jad-151193] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Inmaculada Cuchillo-Ibañez
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Sant Joan d’Alacant, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Valeria Balmaceda
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Sant Joan d’Alacant, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Trinidad Mata-Balaguer
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Sant Joan d’Alacant, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Inmaculada Lopez-Font
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Sant Joan d’Alacant, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Javier Sáez-Valero
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Sant Joan d’Alacant, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| |
Collapse
|
13
|
Lussier AL, Weeber EJ, Rebeck GW. Reelin Proteolysis Affects Signaling Related to Normal Synapse Function and Neurodegeneration. Front Cell Neurosci 2016; 10:75. [PMID: 27065802 PMCID: PMC4809875 DOI: 10.3389/fncel.2016.00075] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 03/11/2016] [Indexed: 01/13/2023] Open
Abstract
Reelin is a neurodevelopmental protein important in adult synaptic plasticity and learning and memory. Recent evidence points to the importance for Reelin proteolysis in normal signaling and in cognitive function. Support for the dysfunction of Reelin proteolysis in neurodegeneration and cognitive dysfunction comes from postmortem analysis of Alzheimer’s diseases (AD) tissues including cerebral spinal fluid (CSF), showing that levels of Reelin fragments are altered in AD compared to control. Potential key proteases involved in Reelin proteolysis have recently been defined, identifying processes that could be altered in neurodegeneration. Introduction of full-length Reelin and its proteolytic fragments into several mouse models of neurodegeneration and neuropsychiatric disorders quickly promote learning and memory. These findings support a role for Reelin in learning and memory and suggest further understanding of these processes are important to harness the potential of this pathway in treating cognitive symptoms in neuropsychiatric and neurodegenerative diseases.
Collapse
Affiliation(s)
- April L Lussier
- Department of Molecular Pharmacology and Physiology, USF Health Byrd Alzheimer's Disease Institute, University of South Florida Tampa, FL, USA
| | - Edwin J Weeber
- Department of Molecular Pharmacology and Physiology, USF Health Byrd Alzheimer's Disease Institute, University of South Florida Tampa, FL, USA
| | - G William Rebeck
- Department of Neuroscience, Georgetown University Washington, DC, USA
| |
Collapse
|
14
|
Vera A, Recabal A, Saldivia N, Stanic K, Torrejón M, Montecinos H, Caprile T. Interaction between SCO-spondin and low density lipoproteins from embryonic cerebrospinal fluid modulates their roles in early neurogenesis. Front Neuroanat 2015; 9:72. [PMID: 26074785 PMCID: PMC4446542 DOI: 10.3389/fnana.2015.00072] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 05/14/2015] [Indexed: 01/20/2023] Open
Abstract
During early stages of development, encephalic vesicles are composed by a layer of neuroepithelial cells surrounding a central cavity filled with embryonic cerebrospinal fluid (eCSF). This fluid contains several morphogens that regulate proliferation and differentiation of neuroepithelial cells. One of these neurogenic factors is SCO-spondin, a giant protein secreted to the eCSF from early stages of development. Inhibition of this protein in vivo or in vitro drastically decreases the neurodifferentiation process. Other important neurogenic factors of the eCSF are low density lipoproteins (LDL), the depletion of which generates a 60% decrease in mesencephalic explant neurodifferentiation. The presence of several LDL receptor class A (LDLrA) domains (responsible for LDL binding in other proteins) in the SCO-spondin sequence suggests a possible interaction between both molecules. This possibility was analyzed using three different experimental approaches: (1) Bioinformatics analyses of the SCO-spondin region, that contains eight LDLrA domains in tandem, and of comparisons with the LDL receptor consensus sequence; (2) Analysis of the physical interactions of both molecules through immunohistochemical colocalization in embryonic chick brains and through the immunoprecipitation of LDL with anti-SCO-spondin antibodies; and (3) Analysis of functional interactions during the neurodifferentiation process when these molecules were added to a culture medium of mesencephalic explants. The results revealed that LDL and SCO-spondin interact to form a complex that diminishes the neurogenic capacities that both molecules have separately. Our work suggests that the eCSF is an active signaling center with a complex regulation system that allows for correct brain development.
Collapse
Affiliation(s)
- América Vera
- Department of Cell Biology, Faculty of Biological Sciences, University of Concepción Concepción, Chile
| | - Antonia Recabal
- Department of Cell Biology, Faculty of Biological Sciences, University of Concepción Concepción, Chile
| | - Natalia Saldivia
- Department of Cell Biology, Faculty of Biological Sciences, University of Concepción Concepción, Chile
| | - Karen Stanic
- Department of Cell Biology, Faculty of Biological Sciences, University of Concepción Concepción, Chile
| | - Marcela Torrejón
- Faculty of Biological Sciences, Department of Biochemistry and Molecular Biology, University of Concepción Concepción, Chile
| | - Hernán Montecinos
- Department of Cell Biology, Faculty of Biological Sciences, University of Concepción Concepción, Chile
| | - Teresa Caprile
- Department of Cell Biology, Faculty of Biological Sciences, University of Concepción Concepción, Chile
| |
Collapse
|
15
|
Reelin expression in brain endothelial cells: an electron microscopy study. BMC Neurosci 2015; 16:16. [PMID: 25887698 PMCID: PMC4374371 DOI: 10.1186/s12868-015-0156-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 03/11/2015] [Indexed: 12/20/2022] Open
Abstract
Background Reelin expression and function have been extensively studied in the brain, although its expression has been also reported in other tissues including blood. This raises the possibility that reelin might be able to cross the blood-brain barrier, which could be functionally relevant. Up-to-date no studies have been conducted to assess if reelin is present in the blood-brain barrier, which is mainly constituted by tightly packed endothelial cells. In this report we assessed the expression of reelin in brain capillaries using immunocytochemistry and electron microscopy. Results At the light microscope, reelin immunolabeling appeared in specific endothelial cells in brain areas that presented abundant diffuse labeling for this protein (e.g., layer I of the cortex, or the stratum lacunosum moleculare of the hippocampus), while it was mostly absent from capillaries in other brain areas (e.g., deeper cortical layers, or the CA1 layer of the hippocampus). As expected, at the electron microscope reelin labeling was observed in neurons of the cortex, where most of the labeling was associated with the rough endoplasmic reticulum. Importantly, reelin was also observed in some endothelial cells located in small capillaries, which confirmed the findings obtained at the light microscope. In these cells, reelin labeling was located primarily in caveolae (i.e., vesicles of transcytosis), and associated with the plasma membrane of the luminal side of endothelial cells. In addition, some scarce labeling was observed in the nuclear membrane. Conclusions The presence of reelin immunolabeling in brain endothelial cells, and particularly in caveolar vesicles within these cells, suggests that reelin and/or reelin peptides may be able to cross the blood-brain barrier, which could have important physiological, pathological, and therapeutic implications.
Collapse
|
16
|
DiBattista AM, Dumanis SB, Song JM, Bu G, Weeber E, Rebeck GW, Hoe HS. Very low density lipoprotein receptor regulates dendritic spine formation in a RasGRF1/CaMKII dependent manner. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:904-17. [PMID: 25644714 DOI: 10.1016/j.bbamcr.2015.01.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 12/20/2014] [Accepted: 01/22/2015] [Indexed: 11/17/2022]
Abstract
Very Low Density Lipoprotein Receptor (VLDLR) is an apolipoprotein E receptor involved in synaptic plasticity, learning, and memory. However, it is unknown how VLDLR can regulate synaptic and cognitive function. In the present study, we found that VLDLR is present at the synapse both pre- and post-synaptically. Overexpression of VLDLR significantly increases, while knockdown of VLDLR decreases, dendritic spine number in primary hippocampal cultures. Additionally, knockdown of VLDLR significantly decreases synaptophysin puncta number while differentially regulating cell surface and total levels of glutamate receptor subunits. To identify the mechanism by which VLDLR induces these synaptic effects, we investigated whether VLDLR affects dendritic spine formation through the Ras signaling pathway, which is involved in spinogenesis and neurodegeneration. Interestingly, we found that VLDLR interacts with RasGRF1, a Ras effector, and knockdown of RasGRF1 blocks the effect of VLDLR on spinogenesis. Moreover, we found that VLDLR did not rescue the deficits induced by the absence of Ras signaling proteins CaMKIIα or CaMKIIβ. Taken together, our results suggest that VLDLR requires RasGRF1/CaMKII to alter dendritic spine formation.
Collapse
Affiliation(s)
| | - Sonya B Dumanis
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC 20057, USA; Max Delbreuck Center for Molecular Medicine, Berlin, Germany
| | - Jung Min Song
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Edwin Weeber
- Department of Molecular Pharmacology and Physiology, USF Health Byrd Alzheimer's Institute, University of South Florida, Tampa, FL 33613, USA
| | - G William Rebeck
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Hyang-Sook Hoe
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC 20057, USA; Department of Neurology, Georgetown University Medical Center, Washington, DC 20057, USA.
| |
Collapse
|
17
|
Burrell TC, Divekar SD, Weeber EJ, Rebeck GW. Fyn tyrosine kinase increases Apolipoprotein E Receptor 2 levels and phosphorylation. PLoS One 2014; 9:e110845. [PMID: 25340851 PMCID: PMC4207760 DOI: 10.1371/journal.pone.0110845] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 09/25/2014] [Indexed: 11/27/2022] Open
Abstract
Apolipoprotein E Receptor 2 (ApoER2) and the tyrosine kinase Fyn are both members of the Reelin pathway, a signaling pathway essential for the laminar formation of the cortex during development and proper dendritic spine density and long-term potential (LTP) in the adult brain. In the presence of extracellular Reelin, ApoER2 binds the intracellular protein Dab1, an adaptor protein that is phosphorylated by Fyn. However, direct interactions between ApoER2 and Fyn are not well defined. Here, we show that total levels of ApoER2 and surface levels of ApoER2 are increased by active Fyn. Via a separate mechanism, ApoER2 is also phosphorylated by Fyn, an event that peaks in the postnatal cortex at day 5 and can occur at multiple ApoER2 tyrosine residues. Dab1 is also involved in this phosphorylation, promoting the phosphorylation of ApoER2 by Fyn when it is itself phosphorylated. These results elucidate some of the intracellular mechanisms that give rise to a functional Reelin pathway.
Collapse
Affiliation(s)
- Teal C. Burrell
- Department of Neuroscience, Georgetown University Medical Center, Washington, District of Columbia, United States of America
| | - Shailaja D. Divekar
- Department of Neuroscience, Georgetown University Medical Center, Washington, District of Columbia, United States of America
| | - Edwin J. Weeber
- Department of Molecular Pharmacology and Physiology, University of South Florida Health Byrd Alzheimer’s Institute, University of South Florida, Tampa, Florida, United States of America
| | - G. William Rebeck
- Department of Neuroscience, Georgetown University Medical Center, Washington, District of Columbia, United States of America
- * E-mail:
| |
Collapse
|
18
|
Oka H, Kitagawa M, Takata T. F-spondin inhibits differentiation of clastic precursors via lipoprotein receptor-related protein 8 (LRP8). J Periodontol 2014; 86:465-72. [PMID: 25299387 DOI: 10.1902/jop.2014.140419] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND F-spondin, known to be a secreted neuronal glycoprotein, is highly expressed on the tooth root surface. The authors previously reported that F-spondin is one of the specific markers of cementoblasts in periodontal tissue. In chronic periodontitis, significant cemental resorption rarely occurs on the root side, although alveolar bone resorption by osteoclasts is one of the major pathologic changes. Thus, it was hypothesized that secretory F-spondin from cementoblasts might be involved in differentiation of clastic cells on the root surface. The authors studied effects of secretory F-spondin from F-spondin-expressing cells and its pathway on receptor activator of nuclear factor-κB ligand (RANKL)-mediated differentiation of clastic cells. METHODS Osteoclast precursors were used in this study. With a chamber assay, the authors examined effects of secretory molecules from F-spondin-expressing cells of transgenic mice on RANKL-induced clastic cell differentiation. RESULTS Secretory molecules from F-spondin-overexpressing cells significantly inhibited the RANKL-mediated tartrate-resistant acid phosphatase (TRAP)-positive cells from primary progenitor cells with the chamber system. F-spondin suppressed RANKL-mediated nuclear factor of activated T-cells, cytoplasmic 1 (NFATc1); TRAP; cathepsin K; and dendritic cell-specific transmembrane protein (DC-STAMP) expression in the cells. The suppressive effect of F-spondin on RANKL-induced differentiation of clastic cells was partially blocked by knockdown of low-density lipoprotein receptor-related protein 8 (LRP8). CONCLUSIONS These findings indicate that secretory factors from F-spondin-expressing cells, including F-spondin, downregulate differentiation of clastic precursors. Moreover, F-spondin inhibits RANKL-mediated differentiation of clastic cells partially via LRP8. It is suggested that secretory F-spondin may act protectively from cemental resorption partially via LRP8 in periodontal tissue.
Collapse
Affiliation(s)
- Hiroko Oka
- Department of International Collaboration Development for Dentistry, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | | | | |
Collapse
|