1
|
Huang B, Li X. Mechanisms of GPM6A in Malignant Tumors. Cancer Rep (Hoboken) 2025; 8:e70137. [PMID: 39957375 PMCID: PMC11831008 DOI: 10.1002/cnr2.70137] [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: 06/28/2024] [Revised: 09/20/2024] [Accepted: 01/25/2025] [Indexed: 02/18/2025] Open
Abstract
BACKGROUND Glycoprotein M6A (GPM6A) encodes a transmembrane protein, expressing in large quantities on the cell surface of central nervous system (CNS) neurons. GPM6A acts importantly in neurodevelopment by modulating neuronal differentiation, migration, axon growth, synaptogenesis, and spine formation, but its role in malignancy remains controversial and requires further research. This article reviewed the mechanisms of GPM6A in colorectal cancer, liver cancer, lung cancer, glioblastoma, and other malignant tumors, and made a "one-stop" summary of the relevant mechanisms. RECENT FINDINGS Researches have indicated that GPM6A is related to malignant tumors. It affects epithelial-mesenchymal transition and induces the formation of filopodia, participating in the adhesion, migration, and metastasis of cancer cells. Its role in malignant tumors remains controversial, however. On the one hand, GPM6A may have carcinogenic properties and is related to poor prognosis of malignant tumors. It is highly expressed in lymphoblastic leukemia and is a potential oncogene. It also shows carcinogenic properties in colorectal cancer, glioblastoma, gonadotroph adenomas and so on. On the other hand, the expression of GPM6A decreases in lung adenocarcinoma, liver cancer, thyroid cancer, and so forth as the tumor progresses, and it can inhibit the progression of malignant tumors by inhibiting some signaling pathways, suggesting that it may be a tumor suppressor gene. CONCLUSION Carcinogenic or tumor suppressive? Although the biological function of GPM6A in the development of malignant tumors is still unclear, according to the current research progress, it is still expected to become an effective molecular marker for predicting tumor occurrence, metastasis and prognosis, as well as a new target for diagnosis and treatment.
Collapse
Affiliation(s)
- Bei Huang
- Operation Management and Evaluation Department, West China Second University HospitalSichuan UniversityChengduSichuanPeople's Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University)Ministry of EducationChengduSichuanPeople's Republic of China
| | - Xihong Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University)Ministry of EducationChengduSichuanPeople's Republic of China
- Emergency Department, West China Second University HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| |
Collapse
|
2
|
Li S, Zhang P, Chen W, Ye L, Brannan KW, Le NT, Abe JI, Cooke JP, Wang G. A relay velocity model infers cell-dependent RNA velocity. Nat Biotechnol 2024; 42:99-108. [PMID: 37012448 PMCID: PMC10545816 DOI: 10.1038/s41587-023-01728-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 02/28/2023] [Indexed: 04/05/2023]
Abstract
RNA velocity provides an approach for inferring cellular state transitions from single-cell RNA sequencing (scRNA-seq) data. Conventional RNA velocity models infer universal kinetics from all cells in an scRNA-seq experiment, resulting in unpredictable performance in experiments with multi-stage and/or multi-lineage transition of cell states where the assumption of the same kinetic rates for all cells no longer holds. Here we present cellDancer, a scalable deep neural network that locally infers velocity for each cell from its neighbors and then relays a series of local velocities to provide single-cell resolution inference of velocity kinetics. In the simulation benchmark, cellDancer shows robust performance in multiple kinetic regimes, high dropout ratio datasets and sparse datasets. We show that cellDancer overcomes the limitations of existing RNA velocity models in modeling erythroid maturation and hippocampus development. Moreover, cellDancer provides cell-specific predictions of transcription, splicing and degradation rates, which we identify as potential indicators of cell fate in the mouse pancreas.
Collapse
Affiliation(s)
- Shengyu Li
- Center for Bioinformatics and Computational Biology, Houston Methodist Research Institute, Houston, TX, USA
- Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, USA
- Center for RNA Therapeutics, Houston Methodist Research Institute, Houston, TX, USA
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Pengzhi Zhang
- Center for Bioinformatics and Computational Biology, Houston Methodist Research Institute, Houston, TX, USA
- Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, USA
- Center for RNA Therapeutics, Houston Methodist Research Institute, Houston, TX, USA
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Weiqing Chen
- Center for Bioinformatics and Computational Biology, Houston Methodist Research Institute, Houston, TX, USA
- Department of Physiology, Biophysics & Systems Biology, Weill Cornell Graduate School of Medical Science, Weill Cornell Medicine, Cornell University, Ithaca, NY, USA
| | - Lingqun Ye
- Center for Bioinformatics and Computational Biology, Houston Methodist Research Institute, Houston, TX, USA
- Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, USA
- Center for RNA Therapeutics, Houston Methodist Research Institute, Houston, TX, USA
| | - Kristopher W Brannan
- Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, USA
- Center for RNA Therapeutics, Houston Methodist Research Institute, Houston, TX, USA
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Nhat-Tu Le
- Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, USA
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Jun-Ichi Abe
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John P Cooke
- Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, USA
| | - Guangyu Wang
- Center for Bioinformatics and Computational Biology, Houston Methodist Research Institute, Houston, TX, USA.
- Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, USA.
- Center for RNA Therapeutics, Houston Methodist Research Institute, Houston, TX, USA.
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, Cornell University, New York, NY, USA.
| |
Collapse
|
3
|
Aparicio GI, León A, Gutiérrez Fuster R, Ravenscraft B, Monje PV, Scorticati C. Endogenous Glycoprotein GPM6a Is Involved in Neurite Outgrowth in Rat Dorsal Root Ganglion Neurons. Biomolecules 2023; 13:biom13040594. [PMID: 37189342 DOI: 10.3390/biom13040594] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/09/2023] [Accepted: 03/17/2023] [Indexed: 03/29/2023] Open
Abstract
The peripheral nervous system (PNS) has a unique ability for self-repair. Dorsal root ganglion (DRG) neurons regulate the expression of different molecules, such as neurotrophins and their receptors, to promote axon regeneration after injury. However, the molecular players driving axonal regrowth need to be better defined. The membrane glycoprotein GPM6a has been described to contribute to neuronal development and structural plasticity in central-nervous-system neurons. Recent evidence indicates that GPM6a interacts with molecules from the PNS, although its role in DRG neurons remains unknown. Here, we characterized the expression of GPM6a in embryonic and adult DRGs by combining analysis of public RNA-seq datasets with immunochemical approaches utilizing cultures of rat DRG explants and dissociated neuronal cells. M6a was detected on the cell surfaces of DRG neurons throughout development. Moreover, GPM6a was required for DRG neurite elongation in vitro. In summary, we provide evidence on GPM6a being present in DRG neurons for the first time. Data from our functional experiments support the idea that GPM6a could contribute to axon regeneration in the PNS.
Collapse
|
4
|
Olivieri J, Salzman J. Analysis of RNA processing directly from spatial transcriptomics data reveals previously unknown regulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.13.532412. [PMID: 36993757 PMCID: PMC10054993 DOI: 10.1101/2023.03.13.532412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Technical advances have led to an explosion in the amount of biological data available in recent years, especially in the field of RNA sequencing. Specifically, spatial transcriptomics (ST) datasets, which allow each RNA molecule to be mapped to the 2D location it originated from within a tissue, have become readily available. Due to computational challenges, ST data has rarely been used to study RNA processing such as splicing or differential UTR usage. We apply the ReadZS and the SpliZ, methods developed to analyze RNA process in scRNA-seq data, to analyze spatial localization of RNA processing directly from ST data for the first time. Using Moran's I metric for spatial autocorrelation, we identify genes with spatially regulated RNA processing in the mouse brain and kidney, re-discovering known spatial regulation in Myl6 and identifying previously-unknown spatial regulation in genes such as Rps24, Gng13, Slc8a1, Gpm6a, Gpx3, ActB, Rps8, and S100A9. The rich set of discoveries made here from commonly used reference datasets provides a small taste of what can be learned by applying this technique more broadly to the large quantity of Visium data currently being created.
Collapse
Affiliation(s)
- Julia Olivieri
- Department of Computer Science, University of the Pacific
| | - Julia Salzman
- Department of Biological Data Science, Stanford University
- Department of Biochemistry, Stanford University
| |
Collapse
|
5
|
Molecular Mechanisms Involved in the Regulation of Neurodevelopment by miR-124. Mol Neurobiol 2023; 60:3569-3583. [PMID: 36840845 DOI: 10.1007/s12035-023-03271-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 02/04/2023] [Indexed: 02/26/2023]
Abstract
miR-124 is a miRNA predominantly expressed in the nervous system and accounts for more than a quarter of the total miRNAs in the brain. It regulates neurogenesis, neuronal differentiation, neuronal maturation, and synapse formation and is the most important miRNA in the brain. Furthermore, emerging evidence has suggested miR-124 may be associated with the pathogenesis of various neurodevelopmental and neuropsychiatric disorders. Here, we provide an overview of the role of miR-124 in neurodevelopment and the underling mechanisms, and finally, we prospect the significance of miR-124 research to the field of neuroscience.
Collapse
|
6
|
The retinal pigmentation pathway in human albinism: Not so black and white. Prog Retin Eye Res 2022; 91:101091. [PMID: 35729001 DOI: 10.1016/j.preteyeres.2022.101091] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/24/2022] [Accepted: 05/26/2022] [Indexed: 12/16/2022]
Abstract
Albinism is a pigment disorder affecting eye, skin and/or hair. Patients usually have decreased melanin in affected tissues and suffer from severe visual abnormalities, including foveal hypoplasia and chiasmal misrouting. Combining our data with those of the literature, we propose a single functional genetic retinal signalling pathway that includes all 22 currently known human albinism disease genes. We hypothesise that defects affecting the genesis or function of different intra-cellular organelles, including melanosomes, cause syndromic forms of albinism (Hermansky-Pudlak (HPS) and Chediak-Higashi syndrome (CHS)). We put forward that specific melanosome impairments cause different forms of oculocutaneous albinism (OCA1-8). Further, we incorporate GPR143 that has been implicated in ocular albinism (OA1), characterised by a phenotype limited to the eye. Finally, we include the SLC38A8-associated disorder FHONDA that causes an even more restricted "albinism-related" ocular phenotype with foveal hypoplasia and chiasmal misrouting but without pigmentation defects. We propose the following retinal pigmentation pathway, with increasingly specific genetic and cellular defects causing an increasingly specific ocular phenotype: (HPS1-11/CHS: syndromic forms of albinism)-(OCA1-8: OCA)-(GPR143: OA1)-(SLC38A8: FHONDA). Beyond disease genes involvement, we also evaluate a range of (candidate) regulatory and signalling mechanisms affecting the activity of the pathway in retinal development, retinal pigmentation and albinism. We further suggest that the proposed pigmentation pathway is also involved in other retinal disorders, such as age-related macular degeneration. The hypotheses put forward in this report provide a framework for further systematic studies in albinism and melanin pigmentation disorders.
Collapse
|
7
|
The Glycoprotein M6a Is Associated with Invasiveness and Radioresistance of Glioblastoma Stem Cells. Cells 2022; 11:cells11142128. [PMID: 35883571 PMCID: PMC9321762 DOI: 10.3390/cells11142128] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 11/20/2022] Open
Abstract
Systematic recurrence of glioblastoma (GB) despite surgery and chemo-radiotherapy is due to GB stem cells (GBSC), which are particularly invasive and radioresistant. Therefore, there is a need to identify new factors that might be targeted to decrease GBSC invasive capabilities as well as radioresistance. Patient-derived GBSC were used in this study to demonstrate a higher expression of the glycoprotein M6a (GPM6A) in invasive GBSC compared to non-invasive cells. In 3D invasion assays performed on primary neurospheres of GBSC, we showed that blocking GPM6A expression by siRNA significantly reduced cell invasion. We also demonstrated a high correlation of GPM6A with the oncogenic protein tyrosine phosphatase, PTPRZ1, which regulates GPM6A expression and cell invasion. The results of our study also show that GPM6A and PTPRZ1 are crucial for GBSC sphere formation. Finally, we demonstrated that targeting GPM6A or PTPRZ1 in GBSC increases the radiosensitivity of GBSC. Our results suggest that blocking GPM6A or PTPRZ1 could represent an interesting approach in the treatment of glioblastoma since it would simultaneously target proliferation, invasion, and radioresistance.
Collapse
|
8
|
Piniella D, Martínez-Blanco E, Bartolomé-Martín D, Sanz-Martos AB, Zafra F. Identification by proximity labeling of novel lipidic and proteinaceous potential partners of the dopamine transporter. Cell Mol Life Sci 2021; 78:7733-7756. [PMID: 34709416 PMCID: PMC8629785 DOI: 10.1007/s00018-021-03998-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 10/07/2021] [Accepted: 10/17/2021] [Indexed: 12/05/2022]
Abstract
Dopamine (DA) transporters (DATs) are regulated by trafficking and modulatory processes that probably rely on stable and transient interactions with neighboring proteins and lipids. Using proximity-dependent biotin identification (BioID), we found novel potential partners for DAT, including several membrane proteins, such as the transmembrane chaperone 4F2hc, the proteolipid M6a and a potential membrane receptor for progesterone (PGRMC2). We also detected two cytoplasmic proteins: a component of the Cullin1-dependent ubiquitination machinery termed F-box/LRR-repeat protein 2 (FBXL2), and the enzyme inositol 5-phosphatase 2 (SHIP2). Immunoprecipitation (IP) and immunofluorescence studies confirmed either a physical association or a close spatial proximity between these proteins and DAT. M6a, SHIP2 and the Cullin1 system were shown to increase DAT activity in coexpression experiments, suggesting a functional role for their association. Deeper analysis revealed that M6a, which is enriched in neuronal protrusions (filopodia or dendritic spines), colocalized with DAT in these structures. In addition, the product of SHIP2 enzymatic activity (phosphatidylinositol 3,4-bisphosphate [PI(3,4)P2]) was tightly associated with DAT, as shown by co-IP and by colocalization of mCherry-DAT with a specific biosensor for this phospholipid. PI(3,4)P2 strongly stimulated transport activity in electrophysiological recordings, and conversely, inhibition of SHIP2 reduced DA uptake in several experimental systems including striatal synaptosomes and the dopaminergic cell line SH-SY5Y. In summary, here we report several potential new partners for DAT and a novel regulatory lipid, which may represent new pharmacological targets for DAT, a pivotal protein in dopaminergic function of the brain.
Collapse
Affiliation(s)
- Dolores Piniella
- Centro de Biología Molecular Severo Ochoa and Departamento de Biología Molecular, Facultad de Ciencias, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, C / Nicolás Cabrera 1, 28049, Madrid, Spain
- IdiPAZ, Instituto de Salud Carlos III, Madrid, Spain
| | - Elena Martínez-Blanco
- Centro de Biología Molecular Severo Ochoa and Departamento de Biología Molecular, Facultad de Ciencias, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, C / Nicolás Cabrera 1, 28049, Madrid, Spain
- IdiPAZ, Instituto de Salud Carlos III, Madrid, Spain
| | - David Bartolomé-Martín
- Centro de Biología Molecular Severo Ochoa and Departamento de Biología Molecular, Facultad de Ciencias, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, C / Nicolás Cabrera 1, 28049, Madrid, Spain
- IdiPAZ, Instituto de Salud Carlos III, Madrid, Spain
- Departamento de Bioquímica, Microbiología, Biología Celular y Genética, Universidad de La Laguna, Tenerife, Spain
| | - Ana B Sanz-Martos
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, CEU Universities, 28925, Madrid, Spain
| | - Francisco Zafra
- Centro de Biología Molecular Severo Ochoa and Departamento de Biología Molecular, Facultad de Ciencias, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, C / Nicolás Cabrera 1, 28049, Madrid, Spain.
- IdiPAZ, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
9
|
Fernández EM, Cutraro YB, Adams J, Monteleone MC, Hughes KJ, Frasch AC, Vidal-Gadea AG, Brocco MA. Neuronal membrane glycoprotein (nmgp-1) gene deficiency affects chemosensation-related behaviors, dauer exit and egg-laying in Caenorhabditis elegans. J Neurochem 2021; 160:234-255. [PMID: 34816431 DOI: 10.1111/jnc.15543] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 11/10/2021] [Accepted: 11/14/2021] [Indexed: 11/30/2022]
Abstract
The nervous system monitors the environment to maintain homeostasis, which can be affected by stressful conditions. Using mammalian models of chronic stress, we previously observed altered brain levels of GPM6A, a protein involved in neuronal morphology. However, GPM6A's role in systemic stress responses remains unresolved. The nematode Caenorhabditis elegans expresses a GPM6A ortholog, the neuronal membrane glycoprotein 1 (NMGP-1). Because of the shared features between nematode and mammalian nervous systems and the vast genetic tools available in C. elegans, we used the worm to elucidate the role of GPM6A in the stress response. We first identified nmgp-1 expression in different amphid and phasmid neurons. To understand the nmgp-1 role, we characterized the behavior of nmgp-1(RNAi) animals and two nmgp-1 mutant alleles. Compared to control animals, mutant and RNAi-treated worms exhibited increased recovery time from the stress-resistant dauer stage, altered SDS chemosensation and reduced egg-laying rate resulting in egg retention (bag-of-worms phenotype). Silencing of nmgp-1 expression induced morphological abnormalities in the ASJ sensory neurons, partly responsible for dauer exit. These results indicate that nmgp-1 is required for neuronal morphology and for behaviors associated with chemosensation. Finally, we propose nmgp-1 mutants as a tool to screen drugs for human nervous system pathologies.
Collapse
Affiliation(s)
- Eliana M Fernández
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) San Martín, Buenos Aires, Argentina
| | - Yamila B Cutraro
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) San Martín, Buenos Aires, Argentina
| | - Jessica Adams
- School of Biological Sciences, Illinois State University, Normal, Illinois, USA
| | - Melisa C Monteleone
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) San Martín, Buenos Aires, Argentina
| | - Kiley J Hughes
- School of Biological Sciences, Illinois State University, Normal, Illinois, USA
| | - Alberto C Frasch
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) San Martín, Buenos Aires, Argentina
| | | | - Marcela A Brocco
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) San Martín, Buenos Aires, Argentina
| |
Collapse
|
10
|
Monteleone MC, Billi SC, Viale L, Catoira NP, Frasch AC, Brocco MA. Search of brain-enriched proteins in salivary extracellular vesicles for their use as mental disease biomarkers: A pilot study of the neuronal glycoprotein M6a. JOURNAL OF AFFECTIVE DISORDERS REPORTS 2020. [DOI: 10.1016/j.jadr.2020.100003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
11
|
Aparicio GI, Formoso K, León A, Frasch AC, Scorticati C. Identification of Potential Interacting Proteins With the Extracellular Loops of the Neuronal Glycoprotein M6a by TMT/MS. Front Synaptic Neurosci 2020; 12:28. [PMID: 32848694 PMCID: PMC7396582 DOI: 10.3389/fnsyn.2020.00028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 06/15/2020] [Indexed: 12/21/2022] Open
Abstract
Nowadays, great efforts are made to gain insight into the molecular mechanisms that underlie structural neuronal plasticity. Moreover, the identification of signaling pathways involved in the development of psychiatric disorders aids the screening of possible therapeutic targets. Genetic variations or alterations in GPM6A expression are linked to neurological disorders such as schizophrenia, depression, and Alzheimer's disease. GPM6A encodes the neuronal surface glycoprotein M6a that promotes filopodia/spine, dendrite, and synapse formation by unknown mechanisms. A substantial body of evidence suggests that the extracellular loops of M6a command its function. However, the proteins that associate with them and that modulate neuronal plasticity have not been determined yet. To address this question, we generated a chimera protein that only contains the extracellular loops of M6a and performed a co-immunoprecipitation with rat hippocampus samples followed by TMT/MS. Here, we report 72 proteins, which are good candidates to interact with M6a's extracellular loops and modify its function. Gene ontology (GO) analysis showed that 63% of the potential M6a's interactor proteins belong to the category "synapse," at both sides of the synaptic cleft, "neuron projections" (51%) and "presynapse" (49%). In this sense, we showed that endogenous M6a interacts with piccolo, synaptic vesicle protein 2B, and synapsin 1 in mature cultured hippocampal neurons. Interestingly, about 28% of the proteins left were related to the "myelin sheath" annotation, suggesting that M6a could interact with proteins at the surface of oligodendrocytes. Indeed, we demonstrated the (cis and trans) interaction between M6a and proteolipid protein (PLP) in neuroblastoma N2a cells. Finally, the 72 proteins were subjected to disease-associated genes and variants screening by DisGeNET. Apart from the diseases that have already been associated with M6a, most of the proteins are also involved in "autistic disorder," "epilepsy," and "seizures" increasing the spectrum of disorders in which M6a could play a role. Data are available via ProteomeXchange with identifier PXD017347.
Collapse
Affiliation(s)
- Gabriela I Aparicio
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (IIBio-UNSAM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Argentina
| | - Karina Formoso
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (IIBio-UNSAM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Argentina.,Instituto de Investigaciones Biomédicas (BIOMED), Facultad de Ciencias Médicas, Pontificia Universidad Católica Argentina (UCA), CONICET, San Martín, Argentina
| | - Antonella León
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (IIBio-UNSAM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Argentina
| | - Alberto C Frasch
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (IIBio-UNSAM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Argentina.,Vicerrectorado, Edificio de Gobierno, Universidad Nacional de San Martín (UNSAM), San Martín, Argentina
| | - Camila Scorticati
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (IIBio-UNSAM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Argentina
| |
Collapse
|
12
|
IGARASHI M. Molecular basis of the functions of the mammalian neuronal growth cone revealed using new methods. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2019; 95:358-377. [PMID: 31406059 PMCID: PMC6766448 DOI: 10.2183/pjab.95.026] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 04/26/2019] [Indexed: 05/25/2023]
Abstract
The neuronal growth cone is a highly motile, specialized structure for extending neuronal processes. This structure is essential for nerve growth, axon pathfinding, and accurate synaptogenesis. Growth cones are important not only during development but also for plasticity-dependent synaptogenesis and neuronal circuit rearrangement following neural injury in the mature brain. However, the molecular details of mammalian growth cone function are poorly understood. This review examines molecular findings on the function of the growth cone as a result of the introduction of novel methods such superresolution microscopy and (phospho)proteomics. These results increase the scope of our understating of the molecular mechanisms of growth cone behavior in the mammalian brain.
Collapse
Affiliation(s)
- Michihiro IGARASHI
- Department of Neurochemistry and Molecular Cell Biology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| |
Collapse
|
13
|
Rosas NM, Alvarez Juliá A, Alzuri SE, Frasch AC, Fuchsova B. Alanine Scanning Mutagenesis of the C-Terminal Cytosolic End of Gpm6a Identifies Key Residues Essential for the Formation of Filopodia. Front Mol Neurosci 2018; 11:314. [PMID: 30233315 PMCID: PMC6131581 DOI: 10.3389/fnmol.2018.00314] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 08/15/2018] [Indexed: 12/14/2022] Open
Abstract
Neuronal membrane glycoprotein M6a (Gpm6a) is a protein with four transmembrane regions and the N- and the C-ends facing the cytosol. It functions in processes of neuronal development, outgrowth of neurites, and formation of filopodia, spines, and synapsis. Molecular mechanisms by which Gpm6a acts in these processes are not fully comprehended. Structural similarities of Gpm6a with tetraspanins led us to hypothesize that, similarly to tetraspanins, the cytoplasmic tails function as connections with cytoskeletal and/or signaling proteins. Here, we demonstrate that the C- but not the N-terminal cytosolic end of Gpm6a is required for the formation of filopodia by Gpm6a in cultured neurons from rat hippocampus and in neuroblastoma cells N2a. Further immunofluorescence microcopy and flow cytometry analysis show that deletion of neither the N- nor the C-terminal intracellular domains interferes with the recognition of Gpm6a by the function-blocking antibody directed against the extracellular part of Gpm6a. Expression levels of both truncation mutants were not affected but we observed decrease in the amount of both truncated proteins on cell surface suggesting that the incapacity of the Gpm6a lacking C-terminus to induce filopodium formation is not due to the lower amount of Gpm6a on cell surface. Following colocalization assays shows that deletion of the C- but not the N-terminus diminishes the association of Gpm6a with clathrin implying involvement of clathrin-mediated trafficking events. Next, using comprehensive alanine scanning mutagenesis of the C-terminus we identify K250, K255, and E258 as the key residues for the formation of filopodia by Gpm6a. Substitution of these charged residues with alanine also diminishes the amount of Gpm6a on cell surface and in case of K255 and E258 leads to the lower amount of total expressed protein. Subsequent bioinformatic analysis of Gpm6a amino acid sequence reveals that highly conserved and functional residues cluster preferentially within the C- and not within the N-terminus and that K250, K255, and E258 are predicted as part of sorting signals of transmembrane proteins. Altogether, our results provide evidence that filopodium outgrowth induced by Gpm6a requires functionally critical residues within the C-terminal cytoplasmic tail.
Collapse
Affiliation(s)
- Nicolás M Rosas
- Instituto de Investigaciones Biotecnológicas IIB-INTECH, CONICET-UNSAM, San Martin, Argentina
| | - Anabel Alvarez Juliá
- Instituto de Investigaciones Biotecnológicas IIB-INTECH, CONICET-UNSAM, San Martin, Argentina
| | - Sofia E Alzuri
- Instituto de Investigaciones Biotecnológicas IIB-INTECH, CONICET-UNSAM, San Martin, Argentina
| | - Alberto C Frasch
- Instituto de Investigaciones Biotecnológicas IIB-INTECH, CONICET-UNSAM, San Martin, Argentina
| | - Beata Fuchsova
- Instituto de Investigaciones Biotecnológicas IIB-INTECH, CONICET-UNSAM, San Martin, Argentina
| |
Collapse
|
14
|
Abstract
Genome-wide association studies (GWAS) have identified more than 100 loci that show robust association with schizophrenia risk. However, due to the complexity of linkage disequilibrium and gene regulatory, it is challenging to pinpoint the causal genes at the risk loci and translate the genetic findings from GWAS into disease mechanism and clinical treatment. Here we systematically predicted the plausible candidate causal genes for schizophrenia at genome-wide level. We utilized different approaches and strategies to predict causal genes for schizophrenia, including Sherlock, SMR, DAPPLE, Prix Fixe, NetWAS, and DEPICT. By integrating the results from different prediction approaches, we identified six top candidates that represent promising causal genes for schizophrenia, including CNTN4, GATAD2A, GPM6A, MMP16, PSMA4, and TCF4. Besides, we also identified 35 additional high-confidence causal genes for schizophrenia. The identified causal genes showed distinct spatio-temporal expression patterns in developing and adult human brain. Cell-type-specific expression analysis indicated that the expression level of the predicted causal genes was significantly higher in neurons compared with oligodendrocytes and microglia (P < 0.05). We found that synaptic transmission-related genes were significantly enriched among the identified causal genes (P < 0.05), providing further support for the dysregulation of synaptic transmission in schizophrenia. Finally, we showed that the top six causal genes are dysregulated in schizophrenia cases compared with controls and knockdown of these genes impaired the proliferation of neuronal cells. Our study depicts the landscape of plausible schizophrenia causal genes for the first time. Further genetic and functional validation of these genes will provide mechanistic insights into schizophrenia pathogenesis and may facilitate to provide potential targets for future therapeutics and diagnostics.
Collapse
Affiliation(s)
- Changguo Ma
- 0000000119573309grid.9227.eKey Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223 China
| | - Chunjie Gu
- 0000000119573309grid.9227.eKey Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223 China
| | - Yongxia Huo
- 0000000119573309grid.9227.eKey Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223 China
| | - Xiaoyan Li
- 0000000119573309grid.9227.eKey Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223 China
| | - Xiong-Jian Luo
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China. .,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China.
| |
Collapse
|
15
|
Ito Y, Honda A, Igarashi M. Glycoprotein M6a as a signaling transducer in neuronal lipid rafts. Neurosci Res 2018; 128:19-24. [DOI: 10.1016/j.neures.2017.11.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 11/09/2017] [Accepted: 11/10/2017] [Indexed: 02/07/2023]
|
16
|
Extracellular Signals Induce Glycoprotein M6a Clustering of Lipid Rafts and Associated Signaling Molecules. J Neurosci 2017; 37:4046-4064. [PMID: 28275160 DOI: 10.1523/jneurosci.3319-16.2017] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 01/25/2017] [Accepted: 02/18/2017] [Indexed: 01/08/2023] Open
Abstract
Lipid raft domains, where sphingolipids and cholesterol are enriched, concentrate signaling molecules. To examine how signaling protein complexes are clustered in rafts, we focused on the functions of glycoprotein M6a (GPM6a), which is expressed at a high concentration in developing mouse neurons. Using imaging of lipid rafts, we found that GPM6a congregated in rafts in a GPM6a palmitoylation-dependent manner, thereby contributing to lipid raft clustering. In addition, we found that signaling proteins downstream of GPM6a, such as Rufy3, Rap2, and Tiam2/STEF, accumulated in lipid rafts in a GPM6a-dependent manner and were essential for laminin-dependent polarity during neurite formation in neuronal development. In utero RNAi targeting of GPM6a resulted in abnormally polarized neurons with multiple neurites. These results demonstrate that GPM6a induces the clustering of lipid rafts, which supports the raft aggregation of its associated downstream molecules for acceleration of neuronal polarity determination. Therefore, GPM6a acts as a signal transducer that responds to extracellular signals.SIGNIFICANCE STATEMENT Lipid raft domains, where sphingolipids and cholesterol are enriched, concentrate signaling molecules. We focused on glycoprotein M6a (GPM6a), which is expressed at a high concentration in developing neurons. Using imaging of lipid rafts, we found that GPM6a congregated in rafts in a palmitoylation-dependent manner, thereby contributing to lipid raft clustering. In addition, we found that signaling proteins downstream of GPM6a accumulated in lipid rafts in a GPM6a-dependent manner and were essential for laminin-dependent polarity during neurite formation. In utero RNAi targeting of GPM6a resulted in abnormally polarized neurons with multiple neurites. These results demonstrate that GPM6a induces the clustering of lipid rafts, which supports the raft aggregation of its associated downstream molecules for acceleration of polarity determination. Therefore, GPM6a acts as a signal transducer that responds to extracellular signals.
Collapse
|
17
|
Alvarez Juliá A, Frasch AC, Fuchsova B. Neuronal filopodium formation induced by the membrane glycoprotein M6a (Gpm6a) is facilitated by coronin-1a, Rac1, and p21-activated kinase 1 (Pak1). J Neurochem 2016; 137:46-61. [PMID: 26809475 DOI: 10.1111/jnc.13552] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 01/18/2016] [Accepted: 01/19/2016] [Indexed: 01/01/2023]
Abstract
Stress-responsive neuronal membrane glycoprotein M6a (Gpm6a) functions in neurite extension, filopodium and spine formation and synaptogenesis. The mechanisms of Gpm6a action in these processes are incompletely understood. Previously, we identified the actin regulator coronin-1a (Coro1a) as a putative Gpm6a interacting partner. Here, we used co-immunoprecipitation assays with the anti-Coro1a antibody to show that Coro1a associates with Gpm6a in rat hippocampal neurons. By immunofluorescence microscopy, we demonstrated that in hippocampal neurons Coro1a localizes in F-actin-enriched regions and some of Coro1a spots co-localize with Gpm6a labeling. Notably, the over-expression of a dominant-negative form of Coro1a as well as its down-regulation by siRNA interfered with Gpm6a-induced filopodium formation. Coro1a is known to regulate the plasma membrane translocation and activation of small GTPase Rac1. We show that Coro1a co-immunoprecipitates with Rac1 together with Gpm6a. Pharmacological inhibition of Rac1 resulted in a significant decrease in filopodium formation by Gpm6a. The same was observed upon the co-expression of Gpm6a with the inactive GDP-bound form of Rac1. In this case, the elevated membrane recruitment of GDP-bound Rac1 was detected as well. Moreover, the kinase activity of the p21-activated kinase 1 (Pak1), a main downstream effector of Rac1 that acts downstream of Coro1a, was required for Gpm6a-induced filopodium formation. Taken together, our results provide evidence that a signaling pathway including Coro1a, Rac1, and Pak1 facilitates Gpm6a-induced filopodium formation. Formation of filopodia by membrane glycoprotein M6a (Gpm6a) requires actin regulator coronin-1a (Coro1a), known to regulate plasma membrane localization and activation of Rac1 and its downstream effector Pak1. Coro1a associates with Gpm6a. Blockage of Coro1a, Rac1, or Pak1 interferes with Gpm6a-induced filopodium formation. Moreover, Gpm6a facilitates Rac1 membrane recruitment. Altogether, a mechanistic insight into the process of Gpm6a-induced neuronal filopodium formation is provided.
Collapse
Affiliation(s)
- Anabel Alvarez Juliá
- Instituto de Investigaciones Biotecnológicas IIB-INTECH, CONICET-UNSAM, San Martin, Argentina
| | - Alberto C Frasch
- Instituto de Investigaciones Biotecnológicas IIB-INTECH, CONICET-UNSAM, San Martin, Argentina
| | - Beata Fuchsova
- Instituto de Investigaciones Biotecnológicas IIB-INTECH, CONICET-UNSAM, San Martin, Argentina
| |
Collapse
|
18
|
Gregor A, Kramer JM, van der Voet M, Schanze I, Uebe S, Donders R, Reis A, Schenck A, Zweier C. Altered GPM6A/M6 dosage impairs cognition and causes phenotypes responsive to cholesterol in human and Drosophila. Hum Mutat 2015; 35:1495-505. [PMID: 25224183 DOI: 10.1002/humu.22697] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 09/05/2014] [Indexed: 12/28/2022]
Abstract
Glycoprotein M6A (GPM6A) is a neuronal transmembrane protein of the PLP/DM20 (proteolipid protein) family that associates with cholesterol-rich lipid rafts and promotes filopodia formation. We identified a de novo duplication of the GPM6A gene in a patient with learning disability and behavioral anomalies. Expression analysis in blood lymphocytes showed increased GPM6A levels. An increase of patient-derived lymphoblastoid cells carrying membrane protrusions supports a functional effect of this duplication. To study the consequences of GPM6A dosage alterations in an intact nervous system, we employed Drosophila melanogaster as a model organism. We found that knockdown of Drosophila M6, the sole member of the PLP family in flies, in the wing, and whole organism causes malformation and lethality, respectively. These phenotypes as well as the protrusions of patient-derived lymphoblastoid cells with increased GPM6A levels can be alleviated by cholesterol supplementation. Notably, overexpression as well as loss of M6 in neurons specifically compromises long-term memory in the courtship conditioning paradigm. Our findings thus indicate a critical role of correct GPM6A/M6 levels for cognitive function and support a role of the GPM6A duplication for the patient's phenotype. Together with other recent findings, this study highlights compromised cholesterol homeostasis as a recurrent feature in cognitive phenotypes.
Collapse
Affiliation(s)
- Anne Gregor
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Fuchsova B, Alvarez Juliá A, Rizavi HS, Frasch AC, Pandey GN. Altered expression of neuroplasticity-related genes in the brain of depressed suicides. Neuroscience 2015; 299:1-17. [PMID: 25934039 DOI: 10.1016/j.neuroscience.2015.04.057] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 04/02/2015] [Accepted: 04/22/2015] [Indexed: 02/04/2023]
Abstract
BACKGROUND Expression of the neuronal membrane glycoprotein M6a (GPM6A), the proteolipid protein (PLP/DM20) family member, is downregulated in the hippocampus of chronically stressed animals. Its neuroplastic function involves a role in neurite formation, filopodium outgrowth and synaptogenesis through an unknown mechanism. Disruptions in neuroplasticity mechanisms have been shown to play a significant part in the etiology of depression. Thus, the current investigation examined whether GPM6A expression is also altered in human depressed brain. METHODS Expression levels and coexpression patterns of GPM6A, GPM6B, and PLP1 (two other members of PLP/DM20 family) as well as of the neuroplasticity-related genes identified to associate with GPM6A were determined using quantitative polymerase chain reaction (qPCR) in postmortem samples from the hippocampus (n = 18) and the prefrontal cortex (PFC) (n = 25) of depressed suicide victims and compared with control subjects (hippocampus n = 18; PFC n = 25). Neuroplasticity-related proteins that form complexes with GPM6A were identified by coimmunoprecipitation technique followed by mass spectrometry. RESULTS Results indicated transcriptional downregulation of GPM6A and GPM6B in the hippocampus of depressed suicides. The expression level of calcium/calmodulin-dependent protein kinase II alpha (CAMK2A) and coronin1A (CORO1A) was also significantly decreased. Subsequent analysis of coexpression patterns demonstrated coordinated gene expression in the hippocampus and in the PFC indicating that the function of these genes might be coregulated in the human brain. However, in the brain of depressed suicides this coordinated response was disrupted. CONCLUSIONS Disruption of coordinated gene expression as well as abnormalities in GPM6A and GPM6B expression and expression of the components of GPM6A complexes were detected in the brain of depressed suicides.
Collapse
Affiliation(s)
- B Fuchsova
- Instituto de Investigaciones Biotecnológicas, CONICET-UNSAM, 1650 San Martin, Argentina.
| | - A Alvarez Juliá
- Instituto de Investigaciones Biotecnológicas, CONICET-UNSAM, 1650 San Martin, Argentina
| | - H S Rizavi
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - A C Frasch
- Instituto de Investigaciones Biotecnológicas, CONICET-UNSAM, 1650 San Martin, Argentina
| | - G N Pandey
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
20
|
Mita S, de Monasterio-Schrader P, Fünfschilling U, Kawasaki T, Mizuno H, Iwasato T, Nave KA, Werner HB, Hirata T. Transcallosal Projections Require Glycoprotein M6-Dependent Neurite Growth and Guidance. Cereb Cortex 2014; 25:4111-25. [PMID: 24917275 DOI: 10.1093/cercor/bhu129] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The function of mature neurons critically relies on the developmental outgrowth and projection of their cellular processes. It has long been postulated that the neuronal glycoproteins M6a and M6b are involved in axon growth because these four-transmembrane domain-proteins of the proteolipid protein family are highly enriched on growth cones, but in vivo evidence has been lacking. Here, we report that the function of M6 proteins is required for normal axonal extension and guidance in vivo. In mice lacking both M6a and M6b, a severe hypoplasia of axon tracts was manifested. Most strikingly, the corpus callosum was reduced in thickness despite normal densities of cortical projection neurons. In single neuron tracing, many axons appeared shorter and disorganized in the double-mutant cortex, and some of them were even misdirected laterally toward the subcortex. Probst bundles were not observed. Upon culturing, double-mutant cortical and cerebellar neurons displayed impaired neurite outgrowth, indicating a cell-intrinsic function of M6 proteins. A rescue experiment showed that the intracellular loop of M6a is essential for the support of neurite extension. We propose that M6 proteins are required for proper extension and guidance of callosal axons that follow one of the most complex trajectories in the mammalian nervous system.
Collapse
Affiliation(s)
- Sakura Mita
- Division of Brain Function, National Institute of Genetics, Graduate University for Advanced Studies (Sokendai), Mishima 411-8540, Japan
| | | | - Ursula Fünfschilling
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, D-37075 Goettingen, Germany
| | - Takahiko Kawasaki
- Division of Brain Function, National Institute of Genetics, Graduate University for Advanced Studies (Sokendai), Mishima 411-8540, Japan
| | - Hidenobu Mizuno
- Division of Neurogenetics, National Institute of Genetics, Graduate University for Advanced Studies (Sokendai), Mishima 411-8540, Japan
| | - Takuji Iwasato
- Division of Neurogenetics, National Institute of Genetics, Graduate University for Advanced Studies (Sokendai), Mishima 411-8540, Japan
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, D-37075 Goettingen, Germany
| | - Hauke B Werner
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, D-37075 Goettingen, Germany
| | - Tatsumi Hirata
- Division of Brain Function, National Institute of Genetics, Graduate University for Advanced Studies (Sokendai), Mishima 411-8540, Japan
| |
Collapse
|
21
|
Choi KM, Kim JY, Kim Y. Distribution of the Immunoreactivity for Glycoprotein M6B in the Neurogenic Niche and Reactive Glia in the Injury Penumbra Following Traumatic Brain Injury in Mice. Exp Neurobiol 2013; 22:277-82. [PMID: 24465143 PMCID: PMC3897689 DOI: 10.5607/en.2013.22.4.277] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 11/07/2013] [Accepted: 11/07/2013] [Indexed: 11/19/2022] Open
Abstract
The location and morphology of astrocytes are known to contribute to their diversity, and this diversity is often associated with their selective functions. However, molecular markers for astrocyte subtypes are largely unknown. In this study, we found that the immunoreactivity for glycoprotein GPM6B (M6B-IR) is preferentially expressed in the astrocytes associated with ventricles or neurogenic regions of the adult mouse brain. In particular, M6B-IR in the neurogenic niche was confined to glial fibrillary acidic protein- or nestin-expressing neural stem cells. Furthermore, in the injury penumbra, reactive astrocytes expressing nestin also exhibited strong M6B-IR. These results reveal that GPM6B is a potential molecular marker for a subset of astrocytes, as well as for the injury-dependent activation of astrocytes.
Collapse
Affiliation(s)
- Kyung Mee Choi
- Department of Anatomy, Korea University College of Medicine, Seoul 136-705, Korea
| | - Joo Yeon Kim
- Department of Anatomy, Korea University College of Medicine, Seoul 136-705, Korea
| | - Younghwa Kim
- Department of Emergency Medical Technology, College of Nursing and Public Health, Kyungil University, Gyeongsan 721-701, Korea
| |
Collapse
|
22
|
Monteleone MC, Adrover E, Pallarés ME, Antonelli MC, Frasch AC, Brocco MA. Prenatal stress changes the glycoprotein GPM6A gene expression and induces epigenetic changes in rat offspring brain. Epigenetics 2013; 9:152-60. [PMID: 23959066 DOI: 10.4161/epi.25925] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Prenatal stress (PS) exerts strong impact on fetal brain development and on adult offspring brain functions. Previous work demonstrated that chronic stress alters the mRNA expression of GPM6A, a neuronal glycoprotein involved in filopodium extension. In this work, we analyzed the effect of PS on gpm6a expression and the epigenetic mechanisms involved. Pregnant Wistar rats received restraint stress during the last week of gestation. Male offspring were sacrificed on postnatal days 28 and 60. Hippocampus and prefrontal cortex samples were analyzed for gene expression (qPCR for mRNAs and microRNAs), methylation status (bisulfite conversion) and protein levels. Hippocampal neurons in culture were used to analyze microRNA overexpression effects. Prenatal stress induced changes in gpm6a levels in both tissues and at both ages analyzed, indicating a persistent effect. Two CpG islands in the gpm6a gene were identified. Variations in the methylation pattern at three specific CpGs were found in hippocampus, but not in PFC samples from PS offspring. microRNAs predicted to target gpm6a were identified in silico. qPCR measurements showed that PS modified the expression of several microRNAs in both tissues, being microRNA-133b the most significantly altered. Further studies overexpressing this microRNA in neuronal cultures showed a reduction in gmp6a mRNA and protein level. Moreover filopodium density was also reduced, suggesting that GPM6A function was affected. Gestational stress affected gpm6a gene expression in offspring likely through changes in methylation status and in posttranscriptional regulation by microRNAs. Thus, our findings propose gpm6a as a novel target for epigenetic regulation during prenatal stress.
Collapse
Affiliation(s)
- Melisa C Monteleone
- Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico de Chascomus (IIB-INTECH); Universidad Nacional de San Martín (UNSAM); Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) San Martín; Buenos Aires, Argentina
| | - Ezequiela Adrover
- IQUIFIB; Facultad de Farmacia y Bioquímica; Universidad de Buenos Aires; Buenos Aires, Argentina
| | - María Eugenia Pallarés
- IQUIFIB; Facultad de Farmacia y Bioquímica; Universidad de Buenos Aires; Buenos Aires, Argentina
| | - Marta C Antonelli
- IQUIFIB; Facultad de Farmacia y Bioquímica; Universidad de Buenos Aires; Buenos Aires, Argentina
| | - Alberto C Frasch
- Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico de Chascomus (IIB-INTECH); Universidad Nacional de San Martín (UNSAM); Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) San Martín; Buenos Aires, Argentina
| | - Marcela A Brocco
- Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico de Chascomus (IIB-INTECH); Universidad Nacional de San Martín (UNSAM); Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) San Martín; Buenos Aires, Argentina
| |
Collapse
|
23
|
Pandiri AR, Sills RC, Ziglioli V, Ton TVT, Hong HHL, Lahousse SA, Gerrish KE, Auerbach SS, Shockley KR, Bushel PR, Peddada SD, Hoenerhoff MJ. Differential transcriptomic analysis of spontaneous lung tumors in B6C3F1 mice: comparison to human non-small cell lung cancer. Toxicol Pathol 2012; 40:1141-59. [PMID: 22688403 DOI: 10.1177/0192623312447543] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Lung cancer is the leading cause of cancer-related death in people and is mainly due to environmental factors such as smoking and radon. The National Toxicology Program (NTP) tests various chemicals and mixtures for their carcinogenic hazard potential. In the NTP chronic bioassay using B6C3F1 mice, the incidence of lung tumors in treated and control animals is second only to the liver tumors. In order to study the molecular mechanisms of chemically induced lung tumors, an understanding of the genetic changes that occur in spontaneous lung (SL) tumors from untreated control animals is needed. The authors have evaluated the differential transcriptomic changes within SL tumors compared to normal lungs from untreated age-matched animals. Within SL tumors, several canonical pathways associated with cancer (eukaryotic initiation factor 2 signaling, RhoA signaling, PTEN signaling, and mammalian target of rapamycin signaling), metabolism (Inositol phosphate metabolism, mitochondrial dysfunction, and purine and pyramidine metabolism), and immune responses (FcγR-mediated phagocytosis, clathrin-mediated endocytosis, interleukin 8 signaling, and CXCR4 signaling) were altered. Meta-analysis of murine SL tumors and human non-small cell lung cancer transcriptomic data sets revealed a high concordance. These data provide important information on the differential transcriptomic changes in murine SL tumors that will be critical to our understanding of chemically induced lung tumors and will aid in hazard analysis in the NTP 2-year carcinogenicity bioassays.
Collapse
Affiliation(s)
- Arun R Pandiri
- Cellular and Molecular Pathology Branch, National Toxicology Program-NTP, National Institute of Environmental Health Sciences-NIEHS, Research Triangle Park, North Carolina, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Actin-independent behavior and membrane deformation exhibited by the four-transmembrane protein M6a. PLoS One 2011; 6:e26702. [PMID: 22162747 PMCID: PMC3230579 DOI: 10.1371/journal.pone.0026702] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Accepted: 09/30/2011] [Indexed: 12/27/2022] Open
Abstract
M6a is a four-transmembrane protein that is abundantly expressed in the nervous system. Previous studies have shown that over-expression of this protein induces various cellular protrusions, such as neurites, filopodia, and dendritic spines. In this detailed characterization of M6a-induced structures, we found their varied and peculiar characteristics. Notably, the M6a-induced protrusions were mostly devoid of actin filaments or microtubules and exhibited free random vibrating motion. Moreover, when an antibody bound to M6a, the membrane-wrapped protrusions were suddenly disrupted, leading to perturbation of the surrounding membrane dynamics involving phosphoinositide signaling. During single-molecule analysis, M6a exhibited cytoskeleton-independent movement and became selectively entrapped along the cell perimeter in an actin-independent manner. These observations highlight the unusual characteristics of M6a, which may have a significant yet unappreciated role in biological systems.
Collapse
|
25
|
Dorrell C, Grompe MT, Pan FC, Zhong Y, Canaday PS, Shultz LD, Greiner DL, Wright CV, Streeter PR, Grompe M. Isolation of mouse pancreatic alpha, beta, duct and acinar populations with cell surface markers. Mol Cell Endocrinol 2011; 339:144-50. [PMID: 21539888 PMCID: PMC3112273 DOI: 10.1016/j.mce.2011.04.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2011] [Revised: 03/17/2011] [Accepted: 04/13/2011] [Indexed: 12/25/2022]
Abstract
Tools permitting the isolation of live pancreatic cell subsets for culture and/or molecular analysis are limited. To address this, we developed a collection of monoclonal antibodies with selective surface labeling of endocrine and exocrine pancreatic cell types. Cell type labeling specificity and cell surface reactivity were validated on mouse pancreatic sections and by gene expression analysis of cells isolated using FACS. Five antibodies which marked populations of particular interest were used to isolate and study viable populations of purified pancreatic ducts, acinar cells, and subsets of acinar cells from whole pancreatic tissue or of alpha or beta cells from isolated mouse islets. Gene expression analysis showed the presence of known endocrine markers in alpha and beta cell populations and revealed that TTR and DPPIV are primarily expressed in alpha cells whereas DGKB and GPM6A have a beta cell specific expression profile.
Collapse
Affiliation(s)
- Craig Dorrell
- Oregon Health and Science University and Oregon Stem Cell Center, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Pao PC, Huang NK, Liu YW, Yeh SH, Lin ST, Hsieh CP, Huang AM, Huang HS, Tseng JT, Chang WC, Lee YC. A novel RING finger protein, Znf179, modulates cell cycle exit and neuronal differentiation of P19 embryonal carcinoma cells. Cell Death Differ 2011; 18:1791-804. [PMID: 21566658 PMCID: PMC3190115 DOI: 10.1038/cdd.2011.52] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Znf179 is a member of the RING finger protein family. During embryogenesis, Znf179 is expressed in a restricted manner in the brain, suggesting a potential role in nervous system development. In this report, we show that the expression of Znf179 is upregulated during P19 cell neuronal differentiation. Inhibition of Znf179 expression by RNA interference significantly attenuated neuronal differentiation of P19 cells and a primary culture of cerebellar granule cells. Using a microarray approach and subsequent functional annotation analysis, we identified differentially expressed genes in Znf179-knockdown cells and found that several genes are involved in development, cellular growth, and cell cycle control. Flow cytometric analyses revealed that the population of G0/G1 cells decreased in Znf179-knockdown cells. In agreement with the flow cytometric data, the number of BrdU-incorporated cells significantly increased in Znf179-knockdown cells. Moreover, in Znf179-knockdown cells, p35, a neuronal-specific Cdk5 activator that is known to activate Cdk5 and may affect the cell cycle, and p27, a cell cycle inhibitor, also decreased. Collectively, these results show that induction of the Znf179 gene may be associated with p35 expression and p27 protein accumulation, which lead to cell cycle arrest in the G0/G1 phase, and is critical for neuronal differentiation of P19 cells.
Collapse
Affiliation(s)
- P-C Pao
- Institute of Bioinformatics and Biosignal Transduction, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Scorticati C, Formoso K, Frasch AC. Neuronal glycoprotein M6a induces filopodia formation via association with cholesterol-rich lipid rafts. J Neurochem 2011; 119:521-31. [PMID: 21426347 DOI: 10.1111/j.1471-4159.2011.07252.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A neuronal integral membrane glycoprotein M6a has been suggested to be involved in a number of biological processes, including neuronal remodeling and differentiation, trafficking of mu-opioid receptors, and Ca(2+) transportation. Moreover, pathological situations such as chronic stress in animals and depression in humans have been associated with alterations in M6a sequence and expression. The mechanism of action of M6a is essentially unknown. In this work, we analyze the relevance of M6a distribution in plasma membrane, namely its lipid microdomain targeting, for its biological function in filopodia formation. We demonstrate that M6a is localized in membrane microdomains compatible with lipid rafts in cultured rat hippocampal neurons. Removal of cholesterol from neuronal membranes with methyl-β-cyclodextrin decreases M6a-induced filopodia formation, an effect that is reversed by the addition of cholesterol. Inhibition of Src kinases and MAPK prevents filopodia formation in M6a-over-expressing neurons. Src-deficient SYF cells over-expressing M6a fail to promote filopodia formation. Taken together, our findings reveal that the association of M6a with lipid rafts is important for its role in filopodia formation and Src and MAPK kinases participate in M6a signal propagation.
Collapse
Affiliation(s)
- Camila Scorticati
- Instituto de Investigaciones Biotecnológicas (IIB-INTECH), Universidad Nacional de San Martín, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Argentina.
| | | | | |
Collapse
|
28
|
Kitamura Y. [Dopaminergic neuroprotection and reconstruction of neural network tiara]. YAKUGAKU ZASSHI 2010; 130:1263-72. [PMID: 20930477 DOI: 10.1248/yakushi.130.1263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Parkinson's disease (PD) is an age-related neurodegenerative disorder in whose brain massive loss of dopaminergic neurons and formation of Lewy bodies occur in the substantia nigra (SN). L-Dihydroxyphenylamine (L-DOPA) substitution is still considered the gold standard of antiparkinsonian drug therapy. However, there has been little information available on neuroprotective and regenerative therapies. Recently, we have found that pramipexole and talipexole (D(2)/D(3)-dopaminergic agonists) inhibit dopaminergic neurotoxin-induced production of reactive oxygen species and apoptotic cell death. In addition, treatment with these drugs induces enhancement of anti-apoptotic Bcl-2 expression and inhibition of α-synuclein aggregation. Interestingly, recent study suggests that pramipexole treatment delays the progression of early PD symptom. On the other hand, we investigated the transplantation strategy for PD by assessing whether double-transplants of mouse embryonic stem (ES) cell-derived neurons in the striatum (ST) and SN, or subthalamic nucleus (STN), induce functional recovery in rat hemi-parkinsonian model. The study indicates that both the involvement of ST as a place of transplantation and the number of ES cell-derived neurons are essential factors for efficacy on PD animal model. Interestingly, an invertebrate planarian can regenerate complete organs, including a well-organized central nervous system (brain), within about 7 days. The regeneration process of the planarian dopaminergic neural network (tiara) may be divided into five steps: 1) anterior blastema formation, 2) brain rudiment formation, 3) brain pattern formation, 4) the formation of dopaminergic tiara, and 5) functional recovery of dopaminergic motor regulation, with several kinds of genes and molecular cascades acting at each step.
Collapse
Affiliation(s)
- Yoshihisa Kitamura
- Department of Neurobiology, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto, Japan.
| |
Collapse
|
29
|
|
30
|
Brocco MA, Fernández ME, Frasch ACC. Filopodial protrusions induced by glycoprotein M6a exhibit high motility and aids synapse formation. Eur J Neurosci 2010; 31:195-202. [PMID: 20074218 DOI: 10.1111/j.1460-9568.2009.07064.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
M6a is a neuronal membrane glycoprotein whose expression diminishes during chronic stress. M6a overexpression in rat primary hippocampal neurons induces the formation of filopodial protrusions that could be spine precursors. As the filopodium and spine motility has been associated with synaptogenesis, we analysed the motility of M6a-induced protrusions by time-lapse imaging. Our data demonstrate that the motile protrusions formed by the neurons overexpressing M6a were more abundant and moved faster than those formed in control cells. When different putative M6a phosphorylation sites were mutated, the neurons transfected with a mutant lacking intracellular phosphorylation sites bore filopodia, but these protrusions did not move as fast as those formed by cells overexpressing wild-type M6a. This suggests a role for M6a phosphorylation state in filopodium motility. Furthermore, we show that M6a-induced protrusions could be stabilized upon contact with presynaptic region. The motility of filopodia contacting or not neurites overexpressing synaptophysin was analysed. We show that the protrusions that apparently contacted synaptophysin-labeled cells exhibited less motility. The behavior of filopodia from M6a-overexpressing cells and control cells was alike. Thus, M6a-induced protrusions may be spine precursors that move to reach presynaptic membrane. We suggest that M6a is a key molecule for spine formation during development.
Collapse
Affiliation(s)
- Marcela A Brocco
- Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico de Chascomús-Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional de General San Martín, Buenos Aires, Argentina.
| | | | | |
Collapse
|
31
|
Fuchsova B, Fernández ME, Alfonso J, Frasch AC. Cysteine residues in the large extracellular loop (EC2) are essential for the function of the stress-regulated glycoprotein M6a. J Biol Chem 2009; 284:32075-88. [PMID: 19737934 DOI: 10.1074/jbc.m109.012377] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Gpm6a was identified as a stress-responsive gene in the hippocampal formation. This gene is down-regulated in the hippocampus of both socially and physically stressed animals, and this effect can be reversed by antidepressant treatment. Previously we showed that the stress-regulated protein M6a is a key modulator for neurite outgrowth and filopodium/spine formation. In the present work, mutational analysis was used to characterize the action of M6a at the molecular level. We show that four cysteines 162, 174, 192, and 202 within EC2 are functionally crucial sites. The presence of cysteines 162 and 202 is essential for the efficient cell surface expression of the M6a protein. In contrast, cysteines 174 and 192, which form a disulfide bridge as shown by biochemical analysis, are not required for the efficient surface expression of M6a. Their mutation to alanine does not interfere with the localization of M6a to filopodial protrusions in primary hippocampal neurons. The neurons expressing C174A and/or C192A mutants display decreased filopodia number. In non-permeabilized cells, these mutant proteins are not recognized by a function-blocking monoclonal antibody directed to M6a. Moreover, neurons in contact with axons expressing C174A/C192A mutant display significantly lower density of presynaptic clusters over their dendrites. Taken together, this study demonstrates that cysteines in the EC2 domain are critical for the role of M6a in filopodium outgrowth and synaptogenesis.
Collapse
Affiliation(s)
- Beata Fuchsova
- Instituto de Investigaciones Biotecnológicas-INTECH, Universidad Nacional de San Martín, Consejo Nacional de Investigaciones Científicas y Técnicas, 1650 San Martin, Argentina.
| | | | | | | |
Collapse
|
32
|
Phylogeny of proteolipid proteins: divergence, constraints, and the evolution of novel functions in myelination and neuroprotection. ACTA ACUST UNITED AC 2009; 4:111-27. [PMID: 19497142 DOI: 10.1017/s1740925x0900009x] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The protein composition of myelin in the central nervous system (CNS) has changed at the evolutionary transition from fish to tetrapods, when a lipid-associated transmembrane-tetraspan (proteolipid protein, PLP) replaced an adhesion protein of the immunoglobulin superfamily (P0) as the most abundant constituent. Here, we review major steps of proteolipid evolution. Three paralog proteolipids (PLP/DM20/DMalpha, M6B/DMgamma and the neuronal glycoprotein M6A/DMbeta) exist in vertebrates from cartilaginous fish to mammals, and one (M6/CG7540) can be traced in invertebrate bilaterians including the planktonic copepod Calanus finmarchicus that possess a functional myelin equivalent. In fish, DMalpha and DMgamma are coexpressed in oligodendrocytes but are not major myelin components. PLP emerged at the root of tetrapods by the acquisition of an enlarged cytoplasmic loop in the evolutionary older DMalpha/DM20. Transgenic experiments in mice suggest that this loop enhances the incorporation of PLP into myelin. The evolutionary recruitment of PLP as the major myelin protein provided oligodendrocytes with the competence to support long-term axonal integrity. We suggest that the molecular shift from P0 to PLP also correlates with the concentration of adhesive forces at the radial component, and that the new balance between membrane adhesion and dynamics was favorable for CNS myelination.
Collapse
|
33
|
Michibata H, Okuno T, Konishi N, Kyono K, Wakimoto K, Aoki K, Kondo Y, Takata K, Kitamura Y, Taniguchi T. Human GPM6A Is Associated With Differentiation and Neuronal Migration of Neurons Derived from Human Embryonic Stem Cells. Stem Cells Dev 2009; 18:629-39. [DOI: 10.1089/scd.2008.0215] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Hideo Michibata
- Advanced Medical Research Laboratory, Mitsubishi Tanabe Pharma Corporation, Osaka, Japan
- Department of Neurobiology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Tsuyoshi Okuno
- Advanced Medical Research Laboratory, Mitsubishi Tanabe Pharma Corporation, Osaka, Japan
| | - Nae Konishi
- Advanced Medical Research Laboratory, Mitsubishi Tanabe Pharma Corporation, Osaka, Japan
| | - Kiyoshi Kyono
- Advanced Medical Research Laboratory, Mitsubishi Tanabe Pharma Corporation, Osaka, Japan
| | - Koji Wakimoto
- Advanced Medical Research Laboratory, Mitsubishi Tanabe Pharma Corporation, Osaka, Japan
| | - Kan Aoki
- Advanced Medical Research Laboratory, Mitsubishi Tanabe Pharma Corporation, Osaka, Japan
| | - Yasushi Kondo
- Advanced Medical Research Laboratory, Mitsubishi Tanabe Pharma Corporation, Osaka, Japan
| | - Kazuyuki Takata
- Department of Neurobiology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Yoshihisa Kitamura
- Department of Neurobiology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Takashi Taniguchi
- Department of Neurobiology, Kyoto Pharmaceutical University, Kyoto, Japan
| |
Collapse
|
34
|
Fernández ME, Alfonso J, Brocco MA, Frasch AC. Conserved cellular function and stress-mediated regulation among members of the proteolipid protein family. J Neurosci Res 2009; 88:1298-308. [DOI: 10.1002/jnr.22298] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|