1
|
Meur S, Karati D. Fyn Kinase in Alzheimer's Disease: Unraveling Molecular Mechanisms and Therapeutic Implications. Mol Neurobiol 2025; 62:643-660. [PMID: 38890236 DOI: 10.1007/s12035-024-04286-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/05/2024] [Indexed: 06/20/2024]
Abstract
Alzheimer's disease, characterized by the accumulation of abnormal protein aggregates and neuronal damage in the brain, leads to a gradual decline in cognitive function and memory. As a complex neurodegenerative disorder, it involves disruptions in various biochemical pathways and neurotransmitter systems, contributing to the progressive loss of neurons and synaptic connections. The complexity of Alzheimer's signaling pathways complicates treatment, presenting a formidable challenge in the quest for effective therapeutic interventions. A member of the Src family of kinases (SFKs), Fyn, is a type of non-receptor tyrosine kinase that has been linked to multiple essential CNS processes, such as myelination and synaptic transmission. Fyn is an appealing target for AD treatments because it is uniquely linked to the two major pathologies in AD by its interaction with tau, in addition to being activated by amyloid-beta (Aβ) through PrPC. Fyn mediates neurotoxicity and synaptic impairments caused by Aβ and is involved in regulating the process of Aβ synthesis.Additionally, the tau protein's tyrosine phosphorylation is induced by Fyn. Fyn is also a challenging target because of its widespread body expression and strong homology with other kinases of the Src family, which could cause unintentional off-target effects. This review emphasizes signaling pathways mediated by Fyn that govern neuronal development and plasticity while also summarizing the most noteworthy recent research relevant to Fyn kinase's function in the brain. Additionally, the therapeutic inhibition of Fyn kinase has been discussed, with a focus on the Fyn kinase inhibitors that are in clinical trials, which presents a fascinating opportunity for targeting Fyn kinase in the creation of possible therapeutic approaches for the management of Alzheimer's disease.
Collapse
Affiliation(s)
- Shreyasi Meur
- Department of Pharmaceutical Technology, School of Pharmacy, Techno India University, Kolkata, 700091, West Bengal, India
| | - Dipanjan Karati
- Department of Pharmaceutical Technology, School of Pharmacy, Techno India University, Kolkata, 700091, West Bengal, India.
| |
Collapse
|
2
|
Cheon YP, Ryou C, Svedružić ŽM. Roles of prion proteins in mammalian development. Anim Cells Syst (Seoul) 2024; 28:551-566. [PMID: 39664939 PMCID: PMC11633422 DOI: 10.1080/19768354.2024.2436860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/02/2024] [Accepted: 11/25/2024] [Indexed: 12/13/2024] Open
Abstract
Prion protein (PrP) is highly conserved and is expressed in most tissues in a developmental stage-specific manner. Glycosylated cellular prion protein (PrPC) is found in most cells and subcellular areas as a physiological regulating molecule. On the other hand, the amyloid form of PrPC, scrapie PrP (PrPSC), causes transmissible pathogenesis in the central nervous system and induces degeneration of the nervous system. Although many amyloids are reversible and critical in determining the fate, differentiation, and physiological functions of cells, thus far, PrPSC originating from PrPC is not. Although many studies have focused on disorders involving PrPC and the deletion mammalian models for PrPC have no severe phenotype, it has been suggested that PrPC has a role in normal development. It is conserved and expressed from gametes to adult somatic cells. In addition, severe developmental phenotypes appear in PrP null zebrafish embryos and in various mammalian cell model systems. In addition, it has been well established that PrPC is strongly involved in the stemness and differentiation of embryonic stem cells and progenitors. Thus far, many studies on PrPC have focused mostly on disease-associated conditions with physiological roles as a complex platform but not on development. The known roles of PrPC depend on the interacting molecules through its flexible tail and domains. PrPC interacts with membrane, and various intracellular and extracellular molecules. In addition, PrPC and amyloid can stimulate signaling pathways differentially. In this review, we summarize the function of prion protein and discuss its role in development.
Collapse
Affiliation(s)
- Yong-Pil Cheon
- Division of Developmental Biology and Physiology, Department of Biotechnology, Institute for Basic Sciences, Sungshin University, Seoul, Korea
| | - Chongsuk Ryou
- Department of Pharmacy, College of Pharmacy, Hanyang University, ekcho Ansan, Korea
| | | |
Collapse
|
3
|
Bizingre C, Bianchi C, Baudry A, Alleaume-Butaux A, Schneider B, Pietri M. Post-translational modifications in prion diseases. Front Mol Neurosci 2024; 17:1405415. [PMID: 39011540 PMCID: PMC11247024 DOI: 10.3389/fnmol.2024.1405415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 06/14/2024] [Indexed: 07/17/2024] Open
Abstract
More than 650 reversible and irreversible post-translational modifications (PTMs) of proteins have been listed so far. Canonical PTMs of proteins consist of the covalent addition of functional or chemical groups on target backbone amino-acids or the cleavage of the protein itself, giving rise to modified proteins with specific properties in terms of stability, solubility, cell distribution, activity, or interactions with other biomolecules. PTMs of protein contribute to cell homeostatic processes, enabling basal cell functions, allowing the cell to respond and adapt to variations of its environment, and globally maintaining the constancy of the milieu interieur (the body's inner environment) to sustain human health. Abnormal protein PTMs are, however, associated with several disease states, such as cancers, metabolic disorders, or neurodegenerative diseases. Abnormal PTMs alter the functional properties of the protein or even cause a loss of protein function. One example of dramatic PTMs concerns the cellular prion protein (PrPC), a GPI-anchored signaling molecule at the plasma membrane, whose irreversible post-translational conformational conversion (PTCC) into pathogenic prions (PrPSc) provokes neurodegeneration. PrPC PTCC into PrPSc is an additional type of PTM that affects the tridimensional structure and physiological function of PrPC and generates a protein conformer with neurotoxic properties. PrPC PTCC into PrPSc in neurons is the first step of a deleterious sequence of events at the root of a group of neurodegenerative disorders affecting both humans (Creutzfeldt-Jakob diseases for the most representative diseases) and animals (scrapie in sheep, bovine spongiform encephalopathy in cow, and chronic wasting disease in elk and deer). There are currently no therapies to block PrPC PTCC into PrPSc and stop neurodegeneration in prion diseases. Here, we review known PrPC PTMs that influence PrPC conversion into PrPSc. We summarized how PrPC PTCC into PrPSc impacts the PrPC interactome at the plasma membrane and the downstream intracellular controlled protein effectors, whose abnormal activation or trafficking caused by altered PTMs promotes neurodegeneration. We discussed these effectors as candidate drug targets for prion diseases and possibly other neurodegenerative diseases.
Collapse
Affiliation(s)
- Chloé Bizingre
- INSERM UMR-S 1124, Paris, France
- Université Paris Cité, UMR-S 1124, Paris, France
| | - Clara Bianchi
- INSERM UMR-S 1124, Paris, France
- Université Paris Cité, UMR-S 1124, Paris, France
| | - Anne Baudry
- INSERM UMR-S 1124, Paris, France
- Université Paris Cité, UMR-S 1124, Paris, France
| | | | - Benoit Schneider
- INSERM UMR-S 1124, Paris, France
- Université Paris Cité, UMR-S 1124, Paris, France
- Ecole polytechnique, Institut Polytechnique de Paris, CNRS UMR7654, Palaiseau, France
| | - Mathéa Pietri
- INSERM UMR-S 1124, Paris, France
- Université Paris Cité, UMR-S 1124, Paris, France
| |
Collapse
|
4
|
Svedružić ŽM, Ryou C, Choi D, Lee SH, Cheon YP. Physiology of Cellular Prion Proteins in Reproduction. Dev Reprod 2024; 28:29-36. [PMID: 39055100 PMCID: PMC11268893 DOI: 10.12717/dr.2024.28.2.29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 03/25/2024] [Accepted: 03/30/2024] [Indexed: 07/27/2024]
Abstract
Cellular prion protein (PrPC) encoded at Prnp gene is well-known to form a misfolded isoform, termed scrapie PrP (PrPSC) that cause transmissible degenerative diseases in central nervous system. The physiological role of PrPC has been proposed by many studies, showing that PrPC interacts with various intracellular, membrane, and extracellular molecules including mitochondrial inner membrane as a scaffold. PrPC is expressed in most cell types including reproductive organs. Numerous studies using PrPC knockout rodent models found no obvious phenotypic changes, in particular the clear phenotypes in development and reproduction have not demonstrated in these knockout models. However, various roles of PrPC have been evaluated at the cellular levels. In this review, we summarized the known roles of PrPC in various cell types and tissues with a special emphasis on those involved in reproduction.
Collapse
Affiliation(s)
| | - Chongsuk Ryou
- Department of Pharmacy, College of
Pharmacy, Hanyang University ERICA, Ansan 15588,
Korea
| | - Donchan Choi
- Department Life Science, College of
Health Science and Welfare, Yong-In University,
Yongin 17092, Korea
| | - Sung-Ho Lee
- Department of Biotechnology, Sangmyung
University, Seoul 03016, Korea
| | - Yong-Pil Cheon
- Division of Developmental Biology and
Physiology, Department of Biotechnology, Institute for Basic Sciences,
Sungshin University, Seoul 02844,
Korea
| |
Collapse
|
5
|
Yagita K, Noguchi H, Koyama S, Hamasaki H, Komori T, Aishima S, Kosaka T, Ueda M, Komohara Y, Watanabe A, Sasagasako N, Ninomiya T, Oda Y, Honda H. Chronological Changes in the Expression Pattern of Hippocampal Prion Proteins During Disease Progression in Sporadic Creutzfeldt-Jakob Disease MM1 Subtype. J Neuropathol Exp Neurol 2022; 81:900-909. [PMID: 36063412 DOI: 10.1093/jnen/nlac078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The differential effects of sporadic Creutzfeldt-Jakob disease (sCJD) on the hippocampus and other neocortical areas are poorly understood. We aimed to reveal the histological patterns of cellular prion protein (PrPC) and abnormal prion protein (PrPSc) in hippocampi of sCJD patients and normal controls (NCs). Our study examined 18 postmortem sCJD patients (MM1, 14 cases; MM1 + 2c, 3 cases; MM1 + 2t, 1 case) and 12 NCs. Immunohistochemistry was conducted using 4 primary antibodies, of which 3 targeted the N-terminus of the prion protein (PrP), and 1 (EP1802Y) targeted the C-terminal domain. PrPC expression was abundant in the hippocampus of NCs, and the distribution of PrPC at CA3/4 was reminiscent of synaptic complexes. In sCJD cases with a disease history of <2 years, antibodies against the N-terminus could not detect synapse-like PrP expression at CA4; however, EP1802Y could characterize the synapse-like expression. PrPSc accumulation and spongiform changes became evident after 2 years of illness, when PrPSc deposits were more noticeably detected by N-terminal-specific antibodies. Our findings highlighted the chronology of histopathological alterations in the CA4 region in sCJD patients.
Collapse
Affiliation(s)
- Kaoru Yagita
- Department of Neuropathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hideko Noguchi
- Department of Neuropathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Sachiko Koyama
- Department of Neuropathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hideomi Hamasaki
- Department of Neuropathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takashi Komori
- Department of Laboratory Medicine and Pathology, Tokyo Metropolitan Neurological Hospital, Tokyo, Japan
| | - Shinichi Aishima
- Department of Pathology and Microbiology, Faculty of Medicine, University of Saga, Saga, Japan
| | - Takayuki Kosaka
- Department of Neurology, National Hospital Organization Kumamoto Medical Center, Kumamoto, Japan
| | - Mitsuharu Ueda
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoshihiro Komohara
- Department of Cell Pathology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Akihiro Watanabe
- Department of Neurology, Neuro-Muscular Center, National Omuta Hospital, Omuta, Japan
| | - Naokazu Sasagasako
- Department of Neurology, Neuro-Muscular Center, National Omuta Hospital, Omuta, Japan
| | - Toshiharu Ninomiya
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroyuki Honda
- Department of Neuropathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
6
|
Shafiq M, Da Vela S, Amin L, Younas N, Harris DA, Zerr I, Altmeppen HC, Svergun D, Glatzel M. The prion protein and its ligands: Insights into structure-function relationships. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119240. [PMID: 35192891 DOI: 10.1016/j.bbamcr.2022.119240] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 01/23/2022] [Accepted: 01/28/2022] [Indexed: 06/14/2023]
Abstract
The prion protein is a multifunctional protein that exists in at least two different folding states. It is subject to diverse proteolytic processing steps that lead to prion protein fragments some of which are membrane-bound whereas others are soluble. A multitude of ligands bind to the prion protein and besides proteinaceous binding partners, interaction with metal ions and nucleic acids occurs. Although of great importance, information on structural and functional consequences of prion protein binding to its partners is limited. Here, we will reflect on the structure-function relationship of the prion protein and its binding partners considering the different folding states and prion protein fragments.
Collapse
Affiliation(s)
- Mohsin Shafiq
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20251 Hamburg, Germany
| | - Stefano Da Vela
- European Molecular Biology Laboratory (EMBL), Hamburg c/o German Electron Synchrotron (DESY), Notkestraße 85, 22607 Hamburg, Germany
| | - Ladan Amin
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
| | - Neelam Younas
- Department of Neurology, University Medical Center Goettingen, Robert-Koch-str. 40, 37075 Goettingen, Germany
| | - David A Harris
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
| | - Inga Zerr
- Department of Neurology, University Medical Center Goettingen, Robert-Koch-str. 40, 37075 Goettingen, Germany
| | - Hermann C Altmeppen
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20251 Hamburg, Germany
| | - Dmitri Svergun
- European Molecular Biology Laboratory (EMBL), Hamburg c/o German Electron Synchrotron (DESY), Notkestraße 85, 22607 Hamburg, Germany
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20251 Hamburg, Germany.
| |
Collapse
|
7
|
Kawarabayashi T, Nakamura T, Sato K, Seino Y, Ichii S, Nakahata N, Takatama M, Westaway D, George-Hyslop PS, Shoji M. Lipid Rafts Act as a Common Platform for Amyloid-β Oligomer-Induced Alzheimer’s Disease Pathology. J Alzheimers Dis 2022; 87:1189-1203. [DOI: 10.3233/jad-215662] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Amyloid-β (Aβ) oligomers induce the overproduction of phosphorylated tau and neurodegeneration. These cascades gradually cause cognitive impairment in Alzheimer’s disease (AD). While each pathological event in AD has been studied in detail separately, the spatial and temporal relationships between pathological events in AD remain unclear. Objective: We demonstrated that lipid rafts function as a common platform for the pathological cascades of AD. Methods: Cellular and synaptosomal lipid rafts were prepared from the brains of Aβ amyloid model mice (Tg2576 mice) and double transgenic mice (Tg2576 x TgTauP301L mice) and longitudinally analyzed. Results: Aβ dimers, the cellular prion protein (PrPc), and Aβ dimer/PrPc complexes were detected in the lipid rafts. The levels of Fyn, the phosphorylated NR2B subunit of the N-methyl-D-aspartate receptor, glycogen synthase kinase 3β, total tau, phosphorylated tau, and tau oligomers increased with Aβ dimer accumulation in both the cellular and synaptosomal lipid rafts. Increases in the levels of these molecules were first seen at 6 months of age and corresponded with the early stages of Aβ accumulation in the amyloid model mice. Conclusion: Lipid rafts act as a common platform for the progression of AD pathology. The findings of this study suggest a novel therapeutic approach to AD, involving the modification of lipid raft components and the inhibition of their roles in the sequential pathological events of AD.
Collapse
Affiliation(s)
- Takeshi Kawarabayashi
- Department of Neurology, Geriatrics Research Institute and Hospital, Maebashi, Gunma, Japan
- Department of Social Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
- Department of Neurology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Takumi Nakamura
- Department of Social Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
- Department of Neurology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Kaoru Sato
- Department of Orthopaedic Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Yusuke Seino
- Department of Neurology, Hirosaki National Hospital, Hirosaki, Aomori, Japan
| | - Sadanobu Ichii
- Department of Social Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Naoko Nakahata
- Department of Social Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
- Department of Speech and Hearing, Hirosaki University of Health and Welfare/JuniorCollege, Hirosaki, Aomori, Japan
| | - Masamitsu Takatama
- Department of Neurology, Geriatrics Research Institute and Hospital, Maebashi, Gunma, Japan
| | - David Westaway
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB, Canada
| | - Peter St. George-Hyslop
- Tanz Centre for Research in Neurodegenerative Diseases and Departments of Medicine, Medical Biophysics, and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Mikio Shoji
- Department of Neurology, Geriatrics Research Institute and Hospital, Maebashi, Gunma, Japan
- Department of Social Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
- Department of Neurology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| |
Collapse
|
8
|
Matamoros-Angles A, Hervera A, Soriano J, Martí E, Carulla P, Llorens F, Nuvolone M, Aguzzi A, Ferrer I, Gruart A, Delgado-García JM, Del Río JA. Analysis of co-isogenic prion protein deficient mice reveals behavioral deficits, learning impairment, and enhanced hippocampal excitability. BMC Biol 2022; 20:17. [PMID: 35027047 PMCID: PMC8759182 DOI: 10.1186/s12915-021-01203-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 12/02/2021] [Indexed: 12/22/2022] Open
Abstract
Background Cellular prion protein (PrPC) is a cell surface GPI-anchored protein, usually known for its role in the pathogenesis of human and animal prionopathies. However, increasing knowledge about the participation of PrPC in prion pathogenesis contrasts with puzzling data regarding its natural physiological role. PrPC is expressed in a number of tissues, including at high levels in the nervous system, especially in neurons and glial cells, and while previous studies have established a neuroprotective role, conflicting evidence for a synaptic function has revealed both reduced and enhanced long-term potentiation, and variable observations on memory, learning, and behavior. Such evidence has been confounded by the absence of an appropriate knock-out mouse model to dissect the biological relevance of PrPC, with some functions recently shown to be misattributed to PrPC due to the presence of genetic artifacts in mouse models. Here we elucidate the role of PrPC in the hippocampal circuitry and its related functions, such as learning and memory, using a recently available strictly co-isogenic Prnp0/0 mouse model (PrnpZH3/ZH3). Results We performed behavioral and operant conditioning tests to evaluate memory and learning capabilities, with results showing decreased motility, impaired operant conditioning learning, and anxiety-related behavior in PrnpZH3/ZH3 animals. We also carried in vivo electrophysiological recordings on CA3-CA1 synapses in living behaving mice and monitored spontaneous neuronal firing and network formation in primary neuronal cultures of PrnpZH3/ZH3 vs wildtype mice. PrPC absence enhanced susceptibility to high-intensity stimulations and kainate-induced seizures. However, long-term potentiation (LTP) was not enhanced in the PrnpZH3/ZH3 hippocampus. In addition, we observed a delay in neuronal maturation and network formation in PrnpZH3/ZH3 cultures. Conclusion Our results demonstrate that PrPC promotes neuronal network formation and connectivity. PrPC mediates synaptic function and protects the synapse from excitotoxic insults. Its deletion may underlie an epileptogenic-susceptible brain that fails to perform highly cognitive-demanding tasks such as associative learning and anxiety-like behaviors. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-021-01203-0.
Collapse
Affiliation(s)
- A Matamoros-Angles
- Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Barcelona, Spain.,Department of Cell Biology, Physiology, and Immunology, University of Barcelona, Barcelona, Spain.,CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Barcelona, Spain.,Institute of Neuroscience, University of Barcelona, Barcelona, Spain.,Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - A Hervera
- Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Barcelona, Spain.,Department of Cell Biology, Physiology, and Immunology, University of Barcelona, Barcelona, Spain.,CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Barcelona, Spain.,Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - J Soriano
- Departament de Física de la Materia Condensada, University of Barcelona, Barcelona, Spain.,Institute of Complex Systems (UBICS), University of Barcelona, Barcelona, Spain
| | - E Martí
- Department of Biomedicine, University of Barcelona, Barcelona, Spain.,Bioinformatics and Genomics, Center for Genomic Regulation, Barcelona, Spain
| | - P Carulla
- Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Barcelona, Spain.,Department of Cell Biology, Physiology, and Immunology, University of Barcelona, Barcelona, Spain.,CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Barcelona, Spain
| | - F Llorens
- CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Barcelona, Spain.,Department of Neurology, University Medical School, Göttingen, Germany.,Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Catalonia, Spain
| | - M Nuvolone
- Institute of Neuropathology, University Hospital of Zürich, Zürich, Switzerland.,Amyloidosis Center, Foundation IRCCS Policlinico San Matteo, Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - A Aguzzi
- Institute of Neuropathology, University Hospital of Zürich, Zürich, Switzerland
| | - I Ferrer
- CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Barcelona, Spain.,Institute of Neuroscience, University of Barcelona, Barcelona, Spain.,Senior Consultant, Bellvitge University Hospital, IDIBELL (Bellvitge Biomedical Research Centre), L'Hospitalet de Llobregat, Spain.,Department of Pathology and Experimental Therapeutics, University of Barcelona, Barcelona, Spain
| | - A Gruart
- Division of Neurosciences, Pablo de Olavide University, 41013, Seville, Spain
| | - J M Delgado-García
- Division of Neurosciences, Pablo de Olavide University, 41013, Seville, Spain.
| | - J A Del Río
- Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Barcelona, Spain. .,Department of Cell Biology, Physiology, and Immunology, University of Barcelona, Barcelona, Spain. .,CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Barcelona, Spain. .,Institute of Neuroscience, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
9
|
Panes JD, Saavedra P, Pineda B, Escobar K, Cuevas ME, Moraga-Cid G, Fuentealba J, Rivas CI, Rezaei H, Muñoz-Montesino C. PrP C as a Transducer of Physiological and Pathological Signals. Front Mol Neurosci 2021; 14:762918. [PMID: 34880726 PMCID: PMC8648500 DOI: 10.3389/fnmol.2021.762918] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 10/18/2021] [Indexed: 11/13/2022] Open
Abstract
After the discovery of prion phenomenon, the physiological role of the cellular prion protein (PrP C ) remained elusive. In the past decades, molecular and cellular analysis has shed some light regarding interactions and functions of PrP C in health and disease. PrP C , which is located mainly at the plasma membrane of neuronal cells attached by a glycosylphosphatidylinositol (GPI) anchor, can act as a receptor or transducer from external signaling. Although the precise role of PrP C remains elusive, a variety of functions have been proposed for this protein, namely, neuronal excitability and viability. Although many issues must be solved to clearly define the role of PrP C , its connection to the central nervous system (CNS) and to several misfolding-associated diseases makes PrP C an interesting pharmacological target. In a physiological context, several reports have proposed that PrP C modulates synaptic transmission, interacting with various proteins, namely, ion pumps, channels, and metabotropic receptors. PrP C has also been implicated in the pathophysiological cell signaling induced by β-amyloid peptide that leads to synaptic dysfunction in the context of Alzheimer's disease (AD), as a mediator of Aβ-induced cell toxicity. Additionally, it has been implicated in other proteinopathies as well. In this review, we aimed to analyze the role of PrP C as a transducer of physiological and pathological signaling.
Collapse
Affiliation(s)
- Jessica D Panes
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Paulina Saavedra
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile.,Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Benjamin Pineda
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Kathleen Escobar
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile.,Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Magdalena E Cuevas
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Gustavo Moraga-Cid
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Jorge Fuentealba
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Coralia I Rivas
- Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Human Rezaei
- Virologie et Immunologie Moléculaires (VIM), Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Jouy-en-Josas, France.,Université de Versailles Saint-Quentin-en-Yvelines (UVSQ), Versailles, France.,Université Paris-Saclay, Jouy-en-Josas, France
| | - Carola Muñoz-Montesino
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| |
Collapse
|
10
|
Arnould H, Baudouin V, Baudry A, Ribeiro LW, Ardila-Osorio H, Pietri M, Caradeuc C, Soultawi C, Williams D, Alvarez M, Crozet C, Djouadi F, Laforge M, Bertho G, Kellermann O, Launay JM, Schmitt-Ulms G, Schneider B. Loss of prion protein control of glucose metabolism promotes neurodegeneration in model of prion diseases. PLoS Pathog 2021; 17:e1009991. [PMID: 34610054 PMCID: PMC8519435 DOI: 10.1371/journal.ppat.1009991] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 10/15/2021] [Accepted: 09/29/2021] [Indexed: 11/18/2022] Open
Abstract
Corruption of cellular prion protein (PrPC) function(s) at the plasma membrane of neurons is at the root of prion diseases, such as Creutzfeldt-Jakob disease and its variant in humans, and Bovine Spongiform Encephalopathies, better known as mad cow disease, in cattle. The roles exerted by PrPC, however, remain poorly elucidated. With the perspective to grasp the molecular pathways of neurodegeneration occurring in prion diseases, and to identify therapeutic targets, achieving a better understanding of PrPC roles is a priority. Based on global approaches that compare the proteome and metabolome of the PrPC expressing 1C11 neuronal stem cell line to those of PrPnull-1C11 cells stably repressed for PrPC expression, we here unravel that PrPC contributes to the regulation of the energetic metabolism by orienting cells towards mitochondrial oxidative degradation of glucose. Through its coupling to cAMP/protein kinase A signaling, PrPC tones down the expression of the pyruvate dehydrogenase kinase 4 (PDK4). Such an event favors the transfer of pyruvate into mitochondria and its conversion into acetyl-CoA by the pyruvate dehydrogenase complex and, thereby, limits fatty acids β-oxidation and subsequent onset of oxidative stress conditions. The corruption of PrPC metabolic role by pathogenic prions PrPSc causes in the mouse hippocampus an imbalance between glucose oxidative degradation and fatty acids β-oxidation in a PDK4-dependent manner. The inhibition of PDK4 extends the survival of prion-infected mice, supporting that PrPSc-induced deregulation of PDK4 activity and subsequent metabolic derangements contribute to prion diseases. Our study posits PDK4 as a potential therapeutic target to fight against prion diseases.
Collapse
Affiliation(s)
- Hélène Arnould
- INSERM, UMR-S 1124, Paris, France
- Université de Paris, UMR-S 1124, Paris, France
| | - Vincent Baudouin
- INSERM, UMR-S 1124, Paris, France
- Université de Paris, UMR-S 1124, Paris, France
| | - Anne Baudry
- INSERM, UMR-S 1124, Paris, France
- Université de Paris, UMR-S 1124, Paris, France
| | - Luiz W. Ribeiro
- INSERM, UMR-S 1124, Paris, France
- Université de Paris, UMR-S 1124, Paris, France
| | | | - Mathéa Pietri
- INSERM, UMR-S 1124, Paris, France
- Université de Paris, UMR-S 1124, Paris, France
| | - Cédric Caradeuc
- CNRS, UMR 8601, Paris, France
- Université de Paris, UMR 8601, Paris, France
| | - Cynthia Soultawi
- INSERM, UMR-S 1124, Paris, France
- Université de Paris, UMR-S 1124, Paris, France
| | - Declan Williams
- University of Toronto, Tanz Centre for Research in Neurodegenerative Diseases, Canada
| | - Marjorie Alvarez
- INSERM, UMR-S 1124, Paris, France
- Université de Paris, UMR-S 1124, Paris, France
| | - Carole Crozet
- IRMB, Université de Montpellier, INSERM, CHU de Montpellier, Montpellier, France
| | - Fatima Djouadi
- INSERM, UMR-S 1138, Paris, France
- Université de Paris, UMR-S 1138, Paris, France
| | - Mireille Laforge
- INSERM, UMR-S 1124, Paris, France
- Université de Paris, UMR-S 1124, Paris, France
| | - Gildas Bertho
- CNRS, UMR 8601, Paris, France
- Université de Paris, UMR 8601, Paris, France
| | - Odile Kellermann
- INSERM, UMR-S 1124, Paris, France
- Université de Paris, UMR-S 1124, Paris, France
| | - Jean-Marie Launay
- Assistance Publique des Hôpitaux de Paris, INSERM UMR942, Hôpital Lariboisière, Paris, France
- Pharma Research Department, Hoffmann La Roche Ltd, Basel, Switzerland
| | - Gerold Schmitt-Ulms
- University of Toronto, Tanz Centre for Research in Neurodegenerative Diseases, Canada
| | - Benoit Schneider
- INSERM, UMR-S 1124, Paris, France
- Université de Paris, UMR-S 1124, Paris, France
- * E-mail:
| |
Collapse
|
11
|
Guglietti B, Sivasankar S, Mustafa S, Corrigan F, Collins-Praino LE. Fyn Kinase Activity and Its Role in Neurodegenerative Disease Pathology: a Potential Universal Target? Mol Neurobiol 2021; 58:5986-6005. [PMID: 34432266 DOI: 10.1007/s12035-021-02518-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 08/03/2021] [Indexed: 12/14/2022]
Abstract
Fyn is a non-receptor tyrosine kinase belonging to the Src family of kinases (SFKs) which has been implicated in several integral functions throughout the central nervous system (CNS), including myelination and synaptic transmission. More recently, Fyn dysfunction has been associated with pathological processes observed in neurodegenerative diseases, such as multiple sclerosis (MS), Alzheimer's disease (AD) and Parkinson's disease (PD). Neurodegenerative diseases are amongst the leading cause of death and disability worldwide and, due to the ageing population, prevalence is predicted to rise in the coming years. Symptoms across neurodegenerative diseases are both debilitating and degenerative in nature and, concerningly, there are currently no disease-modifying therapies to prevent their progression. As such, it is important to identify potential new therapeutic targets. This review will outline the role of Fyn in normal/homeostatic processes, as well as degenerative/pathological mechanisms associated with neurodegenerative diseases, such as demyelination, pathological protein aggregation, neuroinflammation and cognitive dysfunction.
Collapse
Affiliation(s)
- Bianca Guglietti
- Department of Medical Sciences, University of Adelaide, SG31, Helen Mayo South, Adelaide, SA, 5005, Australia
| | - Srisankavi Sivasankar
- Department of Medical Sciences, University of Adelaide, SG31, Helen Mayo South, Adelaide, SA, 5005, Australia
| | - Sanam Mustafa
- Department of Medical Sciences, University of Adelaide, SG31, Helen Mayo South, Adelaide, SA, 5005, Australia.,ARC Centre of Excellence for Nanoscale BioPhotonics, University of Adelaide, Adelaide, Australia
| | - Frances Corrigan
- Department of Medical Sciences, University of Adelaide, SG31, Helen Mayo South, Adelaide, SA, 5005, Australia
| | - Lyndsey E Collins-Praino
- Department of Medical Sciences, University of Adelaide, SG31, Helen Mayo South, Adelaide, SA, 5005, Australia. .,ARC Centre of Excellence for Nanoscale BioPhotonics, University of Adelaide, Adelaide, Australia.
| |
Collapse
|
12
|
Tau Exon 10 Inclusion by PrP C through Downregulating GSK3β Activity. Int J Mol Sci 2021; 22:ijms22105370. [PMID: 34065232 PMCID: PMC8161268 DOI: 10.3390/ijms22105370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/17/2021] [Accepted: 05/18/2021] [Indexed: 11/17/2022] Open
Abstract
Tau protein is largely responsible for tauopathies, including Alzheimer’s disease (AD), where it accumulates in the brain as insoluble aggregates. Tau mRNA is regulated by alternative splicing, and inclusion or exclusion of exon 10 gives rise to the 3R and 4R isoforms respectively, whose balance is physiologically regulated. In this sense, one of the several factors that regulate alternative splicing of tau is GSK3β, whose activity is inhibited by the cellular prion protein (PrPC), which has different physiological functions in neuroprotection and neuronal differentiation. Moreover, a relationship between PrPC and tau expression levels has been reported during AD evolution. For this reason, in this study we aimed to analyze the role of PrPC and the implication of GSK3β in the regulation of tau exon 10 alternative splicing. We used AD human samples and mouse models of PrPC ablation and tau overexpression. In addition, we used primary neuronal cultures to develop functional studies. Our results revealed a paralleled association between PrPC expression and tau 4R isoforms in all models analyzed. In this sense, reduction or ablation of PrPC levels induces an increase in tau 3R/4R balance. More relevantly, our data points to GSK3β activity downstream from PrPC in this phenomenon. Our results indicate that PrPC plays a role in tau exon 10 inclusion through the inhibitory capacity of GSK3β.
Collapse
|
13
|
A New Take on Prion Protein Dynamics in Cellular Trafficking. Int J Mol Sci 2020; 21:ijms21207763. [PMID: 33092231 PMCID: PMC7589859 DOI: 10.3390/ijms21207763] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 10/14/2020] [Accepted: 10/14/2020] [Indexed: 12/12/2022] Open
Abstract
The mobility of cellular prion protein (PrPC) in specific cell membrane domains and among distinct cell compartments dictates its molecular interactions and directs its cell function. PrPC works in concert with several partners to organize signaling platforms implicated in various cellular processes. The scaffold property of PrPC is able to gather a molecular repertoire to create heterogeneous membrane domains that favor endocytic events. Dynamic trafficking of PrPC through multiple pathways, in a well-orchestrated mechanism of intra and extracellular vesicular transport, defines its functional plasticity, and also assists the conversion and spreading of its infectious isoform associated with neurodegenerative diseases. In this review, we highlight how PrPC traffics across intra- and extracellular compartments and the consequences of this dynamic transport in governing cell functions and contributing to prion disease pathogenesis.
Collapse
|
14
|
Cellular Prion Protein (PrPc): Putative Interacting Partners and Consequences of the Interaction. Int J Mol Sci 2020; 21:ijms21197058. [PMID: 32992764 PMCID: PMC7583789 DOI: 10.3390/ijms21197058] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/20/2020] [Accepted: 09/23/2020] [Indexed: 02/08/2023] Open
Abstract
Cellular prion protein (PrPc) is a small glycosylphosphatidylinositol (GPI) anchored protein most abundantly found in the outer leaflet of the plasma membrane (PM) in the central nervous system (CNS). PrPc misfolding causes neurodegenerative prion diseases in the CNS. PrPc interacts with a wide range of protein partners because of the intrinsically disordered nature of the protein’s N-terminus. Numerous studies have attempted to decipher the physiological role of the prion protein by searching for proteins which interact with PrPc. Biochemical characteristics and biological functions both appear to be affected by interacting protein partners. The key challenge in identifying a potential interacting partner is to demonstrate that binding to a specific ligand is necessary for cellular physiological function or malfunction. In this review, we have summarized the intracellular and extracellular interacting partners of PrPc and potential consequences of their binding. We also briefly describe prion disease-related mutations at the end of this review.
Collapse
|
15
|
Clustering of human prion protein and α-synuclein oligomers requires the prion protein N-terminus. Commun Biol 2020; 3:365. [PMID: 32647130 PMCID: PMC7347944 DOI: 10.1038/s42003-020-1085-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 06/18/2020] [Indexed: 12/31/2022] Open
Abstract
The interaction of prion protein (PrP) and α-synuclein (αSyn) oligomers causes synaptic impairment that might trigger Parkinson’s disease and other synucleinopathies. Here, we report that αSyn oligomers (αSynO) cluster with human PrP (huPrP) into micron-sized condensates. Multivalency of αSyn within oligomers is required for condensation, since clustering with huPrP is not observed for monomeric αSyn. The stoichiometry of the heteroassemblies is well defined with an αSyn:huPrP molar ratio of about 1:1. The αSynO−huPrP interaction is of high affinity, signified by slow dissociation. The huPrP region responsible for condensation of αSynO, residues 95−111 in the intrinsically disordered N-terminus, corresponds to the region required for αSynO-mediated cognitive impairment. HuPrP, moreover, achieves co-clustering of αSynO and Alzheimer’s disease-associated amyloid-β oligomers, providing a case of a cross-interaction of two amyloidogenic proteins through an interlinking intrinsically disordered protein region. The results suggest that αSynO-mediated condensation of huPrP is involved in the pathogenesis of synucleinopathies. Nadine S. Rösener et al. show that α-synuclein oligomers can cluster with human prion protein (huPrP) into condensates. The intrinsically disordered N-terminus of huPrP is necessary for this high-affinity interaction, suggesting a link between cluster formation and toxicity.
Collapse
|
16
|
Wang S, Zhang Z, Almenar-Queralt A, Leem J, DerMardirossian C, Roth DM, Patel PM, Patel HH, Head BP. Caveolin-1 Phosphorylation Is Essential for Axonal Growth of Human Neurons Derived From iPSCs. Front Cell Neurosci 2019; 13:324. [PMID: 31379509 PMCID: PMC6650578 DOI: 10.3389/fncel.2019.00324] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 07/01/2019] [Indexed: 01/02/2023] Open
Abstract
Proper axonal growth and guidance is essential for neuron differentiation and development. Abnormal neuronal development due to genetic or epigenetic influences can contribute to neurological and mental disorders such as Down syndrome, Rett syndrome, and autism. Identification of the molecular targets that promote proper neuronal growth and differentiation may restore structural and functional neuroplasticity, thus improving functional performance in neurodevelopmental disorders. Using differentiated human neuronal progenitor cells (NPCs) derived from induced pluripotent stem cells (iPSCs), the present study demonstrates that during early stage differentiation of human NPCs, neuron-targeted overexpression constitutively active Rac1 (Rac1CA) and constitutively active Cdc42 (Cdc42CA) enhance expression of P-Cav-1, T-Cav-1, and P-cofilin and increases axonal growth. Similarly, neuron-targeted over-expression of Cav-1 (termed SynCav1) increases axonal development by increasing both axon length and volume. Moreover, inhibition of Cav-1(Y14A) phosphorylation blunts Rac1/Cdc42-mediated both axonal growth and differentiation of human NPCs and SynCav1(Y14A)-treated NPCs exhibited blunted axonal growth. These results suggest that: (1) SynCav1-mediated dendritic and axonal growth in human NPCs is dependent upon P-Cav-1, (2) P-Cav-1 is necessary for proper axonal growth during early stages of neuronal differentiation, and (3) Rac1/Cdc42CA-mediated neuronal growth is in part dependent upon P-Cav-1. In conclusion, Cav-1 phosphorylation is essential for human neuronal axonal growth during early stages of neuronal differentiation.
Collapse
Affiliation(s)
- Shanshan Wang
- Veterans Affairs San Diego Healthcare System, San Diego, CA, United States.,Department of Anesthesiology, UC San Diego, La Jolla, CA, United States
| | - Zheng Zhang
- Veterans Affairs San Diego Healthcare System, San Diego, CA, United States.,Department of Anesthesiology, UC San Diego, La Jolla, CA, United States
| | - Angels Almenar-Queralt
- Department of Cellular and Molecular Medicine, Sanford Consortium for Regenerative Medicine, La Jolla, CA, United States
| | - Joseph Leem
- Veterans Affairs San Diego Healthcare System, San Diego, CA, United States.,Department of Anesthesiology, UC San Diego, La Jolla, CA, United States
| | - Celine DerMardirossian
- Department of Immunology and Microbial Sciences, The Scripps Research Institute, La Jolla, CA, United States.,Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, CA, United States
| | - David M Roth
- Veterans Affairs San Diego Healthcare System, San Diego, CA, United States.,Department of Anesthesiology, UC San Diego, La Jolla, CA, United States
| | - Piyush M Patel
- Veterans Affairs San Diego Healthcare System, San Diego, CA, United States.,Department of Anesthesiology, UC San Diego, La Jolla, CA, United States
| | - Hemal H Patel
- Veterans Affairs San Diego Healthcare System, San Diego, CA, United States.,Department of Anesthesiology, UC San Diego, La Jolla, CA, United States
| | - Brian P Head
- Veterans Affairs San Diego Healthcare System, San Diego, CA, United States.,Department of Anesthesiology, UC San Diego, La Jolla, CA, United States
| |
Collapse
|
17
|
Kong C, Xie H, Gao Z, Shao M, Li H, Shi R, Cai L, Gao S, Sun T, Li C. Binding between Prion Protein and Aβ Oligomers Contributes to the Pathogenesis of Alzheimer's Disease. Virol Sin 2019; 34:475-488. [PMID: 31093882 DOI: 10.1007/s12250-019-00124-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Accepted: 03/26/2019] [Indexed: 12/26/2022] Open
Abstract
A plethora of evidence suggests that protein misfolding and aggregation are underlying mechanisms of various neurodegenerative diseases, such as prion diseases and Alzheimer's disease (AD). Like prion diseases, AD has been considered as an infectious disease in the past decades as it shows strain specificity and transmission potential. Although it remains elusive how protein aggregation leads to AD, it is becoming clear that cellular prion protein (PrPC) plays an important role in AD pathogenesis. Here, we briefly reviewed AD pathogenesis and focused on recent progresses how PrPC contributed to AD development. In addition, we proposed a potential mechanism to explain why infectious agents, such as viruses, conduce AD pathogenesis. Microbe infections cause Aβ deposition and upregulation of PrPC, which lead to high affinity binding between Aβ oligomers and PrPC. The interaction between PrPC and Aβ oligomers in turn activates the Fyn signaling cascade, resulting in neuron death in the central nervous system (CNS). Thus, silencing PrPC expression may turn out be an effective treatment for PrPC dependent AD.
Collapse
Affiliation(s)
- Chang Kong
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China.,State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.,Affiliated Cancer Hospital, Institute of Guangzhou Medical University, Guangzhou, 510095, China
| | - Hao Xie
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China
| | - Zhenxing Gao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Ming Shao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Huan Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Run Shi
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Lili Cai
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Shanshan Gao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Taolei Sun
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China
| | - Chaoyang Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China. .,Affiliated Cancer Hospital, Institute of Guangzhou Medical University, Guangzhou, 510095, China.
| |
Collapse
|
18
|
Zhong W, Huang Q, Zeng L, Hu Z, Tang X. Caveolin-1 and MLRs: A potential target for neuronal growth and neuroplasticity after ischemic stroke. Int J Med Sci 2019; 16:1492-1503. [PMID: 31673241 PMCID: PMC6818210 DOI: 10.7150/ijms.35158] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 09/03/2019] [Indexed: 12/22/2022] Open
Abstract
Ischemic stroke is a leading cause of morbidity and mortality worldwide. Thrombolytic therapy, the only established treatment to reduce the neurological deficits caused by ischemic stroke, is limited by time window and potential complications. Therefore, it is necessary to develop new therapeutic strategies to improve neuronal growth and neurological function following ischemic stroke. Membrane lipid rafts (MLRs) are crucial structures for neuron survival and growth signaling pathways. Caveolin-1 (Cav-1), the main scaffold protein present in MLRs, targets many neural growth proteins and promotes growth of neurons and dendrites. Targeting Cav-1 may be a promising therapeutic strategy to enhance neuroplasticity after cerebral ischemia. This review addresses the role of Cav-1 and MLRs in neuronal growth after ischemic stroke, with an emphasis on the mechanisms by which Cav-1/MLRs modulate neuroplasticity via related receptors, signaling pathways, and gene expression. We further discuss how Cav-1/MLRs may be exploited as a potential therapeutic target to restore neuroplasticity after ischemic stroke. Finally, several representative pharmacological agents known to enhance neuroplasticity are discussed in this review.
Collapse
Affiliation(s)
- Wei Zhong
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Qianyi Huang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Liuwang Zeng
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Zhiping Hu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Xiangqi Tang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| |
Collapse
|
19
|
Rösener NS, Gremer L, Reinartz E, König A, Brener O, Heise H, Hoyer W, Neudecker P, Willbold D. A d-enantiomeric peptide interferes with heteroassociation of amyloid-β oligomers and prion protein. J Biol Chem 2018; 293:15748-15764. [PMID: 30131337 PMCID: PMC6187637 DOI: 10.1074/jbc.ra118.003116] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 08/17/2018] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that affects millions of people worldwide. One AD hallmark is the aggregation of β-amyloid (Aβ) into soluble oligomers and insoluble fibrils. Several studies have reported that oligomers rather than fibrils are the most toxic species in AD progression. Aβ oligomers bind with high affinity to membrane-associated prion protein (PrP), leading to toxic signaling across the cell membrane, which makes the Aβ-PrP interaction an attractive therapeutic target. Here, probing this interaction in more detail, we found that both full-length, soluble human (hu) PrP(23-230) and huPrP(23-144), lacking the globular C-terminal domain, bind to Aβ oligomers to form large complexes above the megadalton size range. Following purification by sucrose density-gradient ultracentrifugation, the Aβ and huPrP contents in these heteroassemblies were quantified by reversed-phase HPLC. The Aβ:PrP molar ratio in these assemblies exhibited some limited variation depending on the molar ratio of the initial mixture. Specifically, a molar ratio of about four Aβ to one huPrP in the presence of an excess of huPrP(23-230) or huPrP(23-144) suggested that four Aβ units are required to form one huPrP-binding site. Of note, an Aβ-binding all-d-enantiomeric peptide, RD2D3, competed with huPrP for Aβ oligomers and interfered with Aβ-PrP heteroassembly in a concentration-dependent manner. Our results highlight the importance of multivalent epitopes on Aβ oligomers for Aβ-PrP interactions and have yielded an all-d-peptide-based, therapeutically promising agent that competes with PrP for these interactions.
Collapse
Affiliation(s)
- Nadine S Rösener
- From the Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany and
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Lothar Gremer
- From the Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany and
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Elke Reinartz
- From the Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany and
| | - Anna König
- From the Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany and
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Oleksandr Brener
- From the Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany and
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Henrike Heise
- From the Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany and
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Wolfgang Hoyer
- From the Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany and
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Philipp Neudecker
- From the Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany and
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Dieter Willbold
- From the Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany and
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425 Jülich, Germany
| |
Collapse
|
20
|
Leighton PLA, Kanyo R, Neil GJ, Pollock NM, Allison WT. Prion gene paralogs are dispensable for early zebrafish development and have nonadditive roles in seizure susceptibility. J Biol Chem 2018; 293:12576-12592. [PMID: 29903907 DOI: 10.1074/jbc.ra117.001171] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 06/07/2018] [Indexed: 11/06/2022] Open
Abstract
Normally folded prion protein (PrPC) and its functions in healthy brains remain underappreciated compared with the intense study of its misfolded forms ("prions," PrPSc) during the pathobiology of prion diseases. This impedes the development of therapeutic strategies in Alzheimer's and prion diseases. Disrupting the zebrafish homologs of PrPC has provided novel insights; however, mutagenesis of the zebrafish paralog prp2 did not recapitulate previous dramatic developmental phenotypes, suggesting redundancy with the prp1 paralog. Here, we generated zebrafish prp1 loss-of-function mutant alleles and dual prp1-/-;prp2-/- mutants. Zebrafish prp1-/- and dual prp1-/-;prp2-/- mutants resemble mammalian Prnp knockouts insofar as they lack overt phenotypes, which surprisingly contrasts with reports of severe developmental phenotypes when either prp1 or prp2 is knocked down acutely. Previous studies suggest that PrPC participates in neural cell development/adhesion, including in zebrafish where loss of prp2 affects adhesion and deposition patterns of lateral line neuromasts. In contrast with the expectation that prp1's functions would be redundant to prp2, they appear to have opposing functions in lateral line neurodevelopment. Similarly, loss of prp1 blunted the seizure susceptibility phenotypes observed in prp2 mutants, contrasting the expected exacerbation of phenotypes if these prion gene paralogs were serving redundant roles. In summary, prion mutant fish lack the overt phenotypes previously predicted, and instead they have subtle phenotypes similar to mammals. No evidence was found for functional redundancy in the zebrafish prion gene paralogs, and the phenotypes observed when each gene is disrupted individually are consistent with ancient functions of prion proteins in neurodevelopment and modulation of neural activity.
Collapse
Affiliation(s)
- Patricia L A Leighton
- From the Department of Biological Sciences and the Centre for Prion and Protein Folding Diseases, University of Alberta, Edmonton, Alberta T6G 2E9, Canada
| | - Richard Kanyo
- From the Department of Biological Sciences and the Centre for Prion and Protein Folding Diseases, University of Alberta, Edmonton, Alberta T6G 2E9, Canada
| | - Gavin J Neil
- From the Department of Biological Sciences and the Centre for Prion and Protein Folding Diseases, University of Alberta, Edmonton, Alberta T6G 2E9, Canada
| | - Niall M Pollock
- From the Department of Biological Sciences and the Centre for Prion and Protein Folding Diseases, University of Alberta, Edmonton, Alberta T6G 2E9, Canada
| | - W Ted Allison
- From the Department of Biological Sciences and the Centre for Prion and Protein Folding Diseases, University of Alberta, Edmonton, Alberta T6G 2E9, Canada
| |
Collapse
|
21
|
Leighton PLA, Allison WT. Protein Misfolding in Prion and Prion-Like Diseases: Reconsidering a Required Role for Protein Loss-of-Function. J Alzheimers Dis 2018; 54:3-29. [PMID: 27392869 DOI: 10.3233/jad-160361] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Prion disease research has contributed much toward understanding other neurodegenerative diseases, including recent demonstrations that Alzheimer's disease (AD) and other neurodegenerative diseases are prion-like. Prion-like diseases involve the spread of degeneration between individuals and/or among cells or tissues via template directed misfolding, wherein misfolded protein conformers propagate disease by causing normal proteins to misfold. Here we use the premise that AD, amyotrophic lateral sclerosis, Huntington's disease, and other similar diseases are prion-like and ask: Can we apply knowledge gained from studies of these prion-like diseases to resolve debates about classical prion diseases? We focus on controversies about what role(s) protein loss-of-function might have in prion diseases because this has therapeutic implications, including for AD. We examine which loss-of-function events are recognizable in prion-like diseases by considering the normal functions of the proteins before their misfolding and aggregation. We then delineate scenarios wherein gain-of-function and/or loss-of-function would be necessary or sufficient for neurodegeneration. We consider roles of PrPC loss-of-function in prion diseases and in AD, and conclude that the conventional wisdom that prion diseases are 'toxic gain-of-function diseases' has limitations. While prion diseases certainly have required gain-of-function components, we propose that disease phenotypes are predominantly caused by deficits in the normal physiology of PrPC and its interaction partners as PrPC converts to PrPSc. In this model, gain-of-function serves mainly to spread disease, and loss-of-function directly mediates neuron dysfunction. We propose experiments and predictions to assess our conclusion. Further study on the normal physiological roles of these key proteins is warranted.
Collapse
Affiliation(s)
- Patricia L A Leighton
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, AB, Canada.,Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| | - W Ted Allison
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, AB, Canada.,Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada.,Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
22
|
Allison WT, DuVal MG, Nguyen-Phuoc K, Leighton PLA. Reduced Abundance and Subverted Functions of Proteins in Prion-Like Diseases: Gained Functions Fascinate but Lost Functions Affect Aetiology. Int J Mol Sci 2017; 18:E2223. [PMID: 29064456 PMCID: PMC5666902 DOI: 10.3390/ijms18102223] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 10/18/2017] [Accepted: 10/20/2017] [Indexed: 12/12/2022] Open
Abstract
Prions have served as pathfinders that reveal many aspects of proteostasis in neurons. The recent realization that several prominent neurodegenerative diseases spread via a prion-like mechanism illuminates new possibilities for diagnostics and therapeutics. Thus, key proteins in Alzheimer Disease and Amyotrophic lateral sclerosis (ALS), including amyloid-β precursor protein, Tau and superoxide dismutase 1 (SOD1), spread to adjacent cells in their misfolded aggregated forms and exhibit template-directed misfolding to induce further misfolding, disruptions to proteostasis and toxicity. Here we invert this comparison to ask what these prion-like diseases can teach us about the broad prion disease class, especially regarding the loss of these key proteins' function(s) as they misfold and aggregate. We also consider whether functional amyloids might reveal a role for subverted protein function in neurodegenerative disease. Our synthesis identifies SOD1 as an exemplar of protein functions being lost during prion-like protein misfolding, because SOD1 is inherently unstable and loses function in its misfolded disease-associated form. This has under-appreciated parallels amongst the canonical prion diseases, wherein the normally folded prion protein, PrPC, is reduced in abundance in fatal familial insomnia patients and during the preclinical phase in animal models, apparently via proteostatic mechanisms. Thus while template-directed misfolding and infectious properties represent gain-of-function that fascinates proteostasis researchers and defines (is required for) the prion(-like) diseases, loss and subversion of the functions attributed to hallmark proteins in neurodegenerative disease needs to be integrated into design towards effective therapeutics. We propose experiments to uniquely test these ideas.
Collapse
Affiliation(s)
- W Ted Allison
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, AB T6G 2M8, Canada.
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada.
- Department of Medical Genetics, University of Alberta, Edmonton, AB T6G 2M8, Canada.
| | - Michèle G DuVal
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada.
| | - Kim Nguyen-Phuoc
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, AB T6G 2M8, Canada.
- Department of Medical Genetics, University of Alberta, Edmonton, AB T6G 2M8, Canada.
| | - Patricia L A Leighton
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, AB T6G 2M8, Canada.
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada.
| |
Collapse
|
23
|
Ezpeleta J, Boudet-Devaud F, Pietri M, Baudry A, Baudouin V, Alleaume-Butaux A, Dagoneau N, Kellermann O, Launay JM, Schneider B. Protective role of cellular prion protein against TNFα-mediated inflammation through TACE α-secretase. Sci Rep 2017; 7:7671. [PMID: 28794434 PMCID: PMC5550509 DOI: 10.1038/s41598-017-08110-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 07/11/2017] [Indexed: 12/21/2022] Open
Abstract
Although cellular prion protein PrPC is well known for its implication in Transmissible Spongiform Encephalopathies, its functions remain elusive. Combining in vitro and in vivo approaches, we here show that PrPC displays the intrinsic capacity to protect neuronal cells from a pro-inflammatory TNFα noxious insult. Mechanistically, PrPC coupling to the NADPH oxidase-TACE α-secretase signaling pathway promotes TACE-mediated cleavage of transmembrane TNFα receptors (TNFRs) and the release of soluble TNFR, which limits the sensitivity of recipient cells to TNFα. We further show that PrPC expression is necessary for TACE α-secretase to stay at the plasma membrane in an active state for TNFR shedding. Such PrPC control of TACE localization depends on PrPC modulation of β1 integrin signaling and downstream activation of ROCK-I and PDK1 kinases. Loss of PrPC provokes TACE internalization, which in turn cancels TACE-mediated cleavage of TNFR and renders PrPC-depleted neuronal cells as well as PrPC knockout mice highly vulnerable to pro-inflammatory TNFα insult. Our work provides the prime evidence that in an inflammatory context PrPC adjusts the response of neuronal cells targeted by TNFα through TACE α-secretase. Our data also support the view that abnormal TACE trafficking and activity in prion diseases originate from a-loss-of-PrPC cytoprotective function.
Collapse
Affiliation(s)
- Juliette Ezpeleta
- INSERM, UMR-S 1124, F-75006, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, UMR-S 1124, F-75006, Paris, France
| | - François Boudet-Devaud
- INSERM, UMR-S 1124, F-75006, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, UMR-S 1124, F-75006, Paris, France
| | - Mathéa Pietri
- INSERM, UMR-S 1124, F-75006, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, UMR-S 1124, F-75006, Paris, France
| | - Anne Baudry
- INSERM, UMR-S 1124, F-75006, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, UMR-S 1124, F-75006, Paris, France
| | - Vincent Baudouin
- INSERM, UMR-S 1124, F-75006, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, UMR-S 1124, F-75006, Paris, France
| | - Aurélie Alleaume-Butaux
- INSERM, UMR-S 1124, F-75006, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, UMR-S 1124, F-75006, Paris, France
| | - Nathalie Dagoneau
- INSERM, UMR-S 1124, F-75006, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, UMR-S 1124, F-75006, Paris, France
| | - Odile Kellermann
- INSERM, UMR-S 1124, F-75006, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, UMR-S 1124, F-75006, Paris, France
| | - Jean-Marie Launay
- AP-HP, INSERM UMR-S 942, Hôpital Lariboisière, F-75010, Paris, France.,Pharma Research Department, Hoffmann-La-Roche Ltd, CH4070, Basel, Switzerland
| | - Benoit Schneider
- INSERM, UMR-S 1124, F-75006, Paris, France. .,Université Paris Descartes, Sorbonne Paris Cité, UMR-S 1124, F-75006, Paris, France.
| |
Collapse
|
24
|
Hirsch TZ, Martin-Lannerée S, Mouillet-Richard S. Functions of the Prion Protein. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 150:1-34. [PMID: 28838656 DOI: 10.1016/bs.pmbts.2017.06.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Although initially disregarded compared to prion pathogenesis, the functions exerted by the cellular prion protein PrPC have gained much interest over the past two decades. Research aiming at unraveling PrPC functions started to intensify when it became appreciated that it would give clues as to how it is subverted in the context of prion infection and, more recently, in the context of Alzheimer's disease. It must now be admitted that PrPC is implicated in an incredible variety of biological processes, including neuronal homeostasis, stem cell fate, protection against stress, or cell adhesion. It appears that these diverse roles can all be fulfilled through the involvement of PrPC in cell signaling events. Our aim here is to provide an overview of our current understanding of PrPC functions from the animal to the molecular scale and to highlight some of the remaining gaps that should be addressed in future research.
Collapse
Affiliation(s)
- Théo Z Hirsch
- INSERM UMR 1124, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, UMR 1124, Paris, France
| | - Séverine Martin-Lannerée
- INSERM UMR 1124, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, UMR 1124, Paris, France
| | - Sophie Mouillet-Richard
- INSERM UMR 1124, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, UMR 1124, Paris, France.
| |
Collapse
|
25
|
Abstract
The misfolding of the cellular prion protein (PrPC) causes fatal neurodegenerative diseases. Yet PrPC is highly conserved in mammals, suggesting that it exerts beneficial functions preventing its evolutionary elimination. Ablation of PrPC in mice results in well-defined structural and functional alterations in the peripheral nervous system. Many additional phenotypes were ascribed to the lack of PrPC, but some of these were found to arise from genetic artifacts of the underlying mouse models. Here, we revisit the proposed physiological roles of PrPC in the central and peripheral nervous systems and highlight the need for their critical reassessment using new, rigorously controlled animal models.
Collapse
Affiliation(s)
- Marie-Angela Wulf
- Institute of Neuropathology, University of Zurich, Rämistrasse 100, CH-8091, Zürich, Switzerland
| | - Assunta Senatore
- Institute of Neuropathology, University of Zurich, Rämistrasse 100, CH-8091, Zürich, Switzerland
| | - Adriano Aguzzi
- Institute of Neuropathology, University of Zurich, Rämistrasse 100, CH-8091, Zürich, Switzerland.
| |
Collapse
|
26
|
Linden R. The Biological Function of the Prion Protein: A Cell Surface Scaffold of Signaling Modules. Front Mol Neurosci 2017; 10:77. [PMID: 28373833 PMCID: PMC5357658 DOI: 10.3389/fnmol.2017.00077] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Accepted: 03/06/2017] [Indexed: 12/18/2022] Open
Abstract
The prion glycoprotein (PrPC) is mostly located at the cell surface, tethered to the plasma membrane through a glycosyl-phosphatydil inositol (GPI) anchor. Misfolding of PrPC is associated with the transmissible spongiform encephalopathies (TSEs), whereas its normal conformer serves as a receptor for oligomers of the β-amyloid peptide, which play a major role in the pathogenesis of Alzheimer’s Disease (AD). PrPC is highly expressed in both the nervous and immune systems, as well as in other organs, but its functions are controversial. Extensive experimental work disclosed multiple physiological roles of PrPC at the molecular, cellular and systemic levels, affecting the homeostasis of copper, neuroprotection, stem cell renewal and memory mechanisms, among others. Often each such process has been heralded as the bona fide function of PrPC, despite restricted attention paid to a selected phenotypic trait, associated with either modulation of gene expression or to the engagement of PrPC with a single ligand. In contrast, the GPI-anchored prion protein was shown to bind several extracellular and transmembrane ligands, which are required to endow that protein with the ability to play various roles in transmembrane signal transduction. In addition, differing sets of those ligands are available in cell type- and context-dependent scenarios. To account for such properties, we proposed that PrPC serves as a dynamic platform for the assembly of signaling modules at the cell surface, with widespread consequences for both physiology and behavior. The current review advances the hypothesis that the biological function of the prion protein is that of a cell surface scaffold protein, based on the striking similarities of its functional properties with those of scaffold proteins involved in the organization of intracellular signal transduction pathways. Those properties are: the ability to recruit spatially restricted sets of binding molecules involved in specific signaling; mediation of the crosstalk of signaling pathways; reciprocal allosteric regulation with binding partners; compartmentalized responses; dependence of signaling properties upon posttranslational modification; and stoichiometric requirements and/or oligomerization-dependent impact on signaling. The scaffold concept may contribute to novel approaches to the development of effective treatments to hitherto incurable neurodegenerative diseases, through informed modulation of prion protein-ligand interactions.
Collapse
Affiliation(s)
- Rafael Linden
- Laboratory of Neurogenesis, Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| |
Collapse
|
27
|
Wu ZQ, Li D, Huang Y, Chen XP, Huang W, Liu CF, Zhao HQ, Xu RX, Cheng M, Schachner M, Ma QH. Caspr Controls the Temporal Specification of Neural Progenitor Cells through Notch Signaling in the Developing Mouse Cerebral Cortex. Cereb Cortex 2017; 27:1369-1385. [PMID: 26740489 DOI: 10.1093/cercor/bhv318] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The generation of layer-specific neurons and astrocytes by radial glial cells during development of the cerebral cortex follows a precise temporal sequence, which is regulated by intrinsic and extrinsic factors. The molecular mechanisms controlling the timely generation of layer-specific neurons and astrocytes remain not fully understood. In this study, we show that the adhesion molecule contactin-associated protein (Caspr), which is involved in the maintenance of the polarized domains of myelinated axons, is essential for the timing of generation of neurons and astrocytes in the developing mouse cerebral cortex. Caspr is expressed by radial glial cells, which are neural progenitor cells that generate both neurons and astrocytes. Absence of Caspr in neural progenitor cells delays the production cortical neurons and induces precocious formation of cortical astrocytes, without affecting the numbers of progenitor cells. At the molecular level, Caspr cooperates with the intracellular domain of Notch to repress transcription of the Notch effector Hes1. Suppression of Notch signaling via a Hes1 shRNA rescues the abnormal neurogenesis and astrogenesis in Caspr-deficient mice. These findings establish Caspr as a novel key regulator that controls the temporal specification of cell fate in radial glial cells of the developing cerebral cortex through Notch signaling.
Collapse
Affiliation(s)
- Zhi-Qiang Wu
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Di Li
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Ya Huang
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Xi-Ping Chen
- Department of Forensic Medicine, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Wenhui Huang
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Homburg D-66421, Germany
| | - Chun-Feng Liu
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - He-Qing Zhao
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Ru-Xiang Xu
- Affiliated Bayi Brain Hospital, Beijing Military Hospital, Southern Medical University, Beijing 100070, China
| | - Mei Cheng
- Binzhou Medical University, Yantai, Shandong Province 264000, China
| | - Melitta Schachner
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong Province 515041, China
| | - Quan-Hong Ma
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu Province 215123, China
| |
Collapse
|
28
|
Peggion C, Bertoli A, Sorgato MC. Almost a century of prion protein(s): From pathology to physiology, and back to pathology. Biochem Biophys Res Commun 2016; 483:1148-1155. [PMID: 27581199 DOI: 10.1016/j.bbrc.2016.07.118] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 07/27/2016] [Indexed: 12/30/2022]
Abstract
Prions are one of the few pathogens whose name is renowned at all population levels, after the dramatic years pervaded by the fear of eating prion-infected food. If now this, somehow irrational, scare of bovine meat inexorably transmitting devastating brain disorders is largely subdued, several prion-related issues are still unsolved, precluding the design of therapeutic approaches that could slow, if not halt, prion diseases. One unsolved issue is, for example, the role of the prion protein (PrPC), whole conformational misfolding originates the prion but whose physiologic reason d'etre in neurons, and in cells at large, remains enigmatic. Preceded by a historical outline, the present review will discuss the functional pleiotropicity ascribed to PrPC, and whether this aspect could fall, at least in part, into a more concise framework. It will also be devoted to radically different perspectives for PrPC, which have been recently brought to the attention of the scientific world with unexpected force. Finally, it will discuss the possible reasons allowing an evolutionary conserved and benign protein, as PrPC is, to turn into a high affinity receptor for pathologic misfolded oligomers, and to transmit their toxic message into neurons.
Collapse
Affiliation(s)
- Caterina Peggion
- Department of Biomedical Sciences, University of Padova, Via Bassi 58/B, 35131 Padova, Italy.
| | - Alessandro Bertoli
- Department of Biomedical Sciences, University of Padova, Via Bassi 58/B, 35131 Padova, Italy
| | - M Catia Sorgato
- Department of Biomedical Sciences, University of Padova, Via Bassi 58/B, 35131 Padova, Italy; C.N.R. Institute of Neuroscience, University of Padova, Via Bassi 58/B, 35131 Padova, Italy.
| |
Collapse
|
29
|
Strup-Perrot C, Vozenin MC, Monceau V, Pouzoulet F, Petit B, Holler V, Perrot S, Desquibert L, Fouquet S, Souquere S, Pierron G, Rousset M, Thenet S, Cardot P, Benderitter M, Deutsch E, Aigueperse J. PrP(c) deficiency and dasatinib protect mouse intestines against radiation injury by inhibiting of c-Src. Radiother Oncol 2016; 120:175-83. [PMID: 27406443 DOI: 10.1016/j.radonc.2016.06.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 04/13/2016] [Accepted: 06/14/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND & AIM Despite extensive study of the contribution of cell death and apoptosis to radiation-induced acute intestinal injury, our knowledge of the signaling mechanisms involved in epithelial barrier dysfunction remains inadequate. Because PrP(c) plays a key role in intestinal homeostasis by renewing epithelia, we sought to study its role in epithelial barrier function after irradiation. DESIGN Histology, morphometry and plasma FD-4 levels were used to examine ileal architecture, wound healing, and intestinal leakage in PrP(c)-deficient (KO) and wild-type (WT) mice after total-body irradiation. Impairment of the PrP(c) Src pathway after irradiation was explored by immunofluorescence and confocal microscopy, with Caco-2/Tc7 cells. Lastly, dasatinib treatment was used to switch off the Src pathway in vitro and in vivo. RESULTS The decrease in radiation-induced lethality, improved intestinal wound healing, and reduced intestinal leakage promoted by PrP(c) deficiency demonstrate its involvement in acute intestinal damage. Irradiation of Cacao2/Tc7 cells induced PrP(c) to target the nuclei associated with Src activation. Finally, the protective effect triggered by dasatinib confirmed Src involvement in radiation-induced acute intestinal toxicity. CONCLUSION Our data are the first to show a role for the PrP(c)-Src pathway in acute intestinal response to radiation injury and offer a novel therapeutic opportunity.
Collapse
Affiliation(s)
- Carine Strup-Perrot
- Institut de Radioprotection et de Sûreté Nucléaire, PRP-HOM, SRBE, Laboratoire de Recherche sur la Régénération des tissus sains Irradiés, Fontenay-aux-Roses, France
| | - Marie-Catherine Vozenin
- Inserm U1030, Radiotherapie experimentale, Institut Gustave Roussy, Villejuif, France; Laboratoire de Radio-Oncologie, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Virginie Monceau
- Institut de Radioprotection et de Sûreté Nucléaire, PRP-HOM, SRBE, Laboratoire de Recherche sur la Régénération des tissus sains Irradiés, Fontenay-aux-Roses, France; Inserm U1030, Radiotherapie experimentale, Institut Gustave Roussy, Villejuif, France
| | - Frederic Pouzoulet
- Institut Curie, Translational Research Department, Hopital St Louis, Paris, France
| | - Benoit Petit
- Laboratoire de Radio-Oncologie, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland; Service Commun d'Expérimentation Animale, Institut Gustave Roussy, Villejuif, France
| | - Valérie Holler
- Institut de Radioprotection et de Sûreté Nucléaire, PRP-HOM, SRBE, Laboratoire de Recherche sur la Régénération des tissus sains Irradiés, Fontenay-aux-Roses, France
| | - Sébastien Perrot
- Université Paris-Est, Ecole Nationale Vétérinaire d'Alfort, Institut de Recherche Clinique Animale, Maisons-Alfort Cedex, France
| | - Loïc Desquibert
- Université Paris-Est, Ecole Nationale Vétérinaire d'Alfort, Institut de Recherche Clinique Animale, Maisons-Alfort Cedex, France
| | - Stéphane Fouquet
- Stéphane FOUQUET, Centre de Recherche Institut de la Vision, UMR_S968 Inserm/UPMC/CHNO des Quinze-Vingts, Paris, France
| | | | - Gérard Pierron
- CNRS, UMR-8122, Institut Gustave Roussy, Villejuif, France
| | - Monique Rousset
- Centre de Recherche des Cordeliers, Université Pierre et Marie Curie-Paris 6, UMR S 872, France; INSERM, U 872, Paris, France; Université Paris Descartes-Paris 5, UMR S 872, France
| | - Sophie Thenet
- Centre de Recherche des Cordeliers, Université Pierre et Marie Curie-Paris 6, UMR S 872, France; INSERM, U 872, Paris, France; Université Paris Descartes-Paris 5, UMR S 872, France; Ecole Pratique des Hautes Etudes, Laboratoire de Pharmacologie Cellulaire et Moléculaire, Paris, France
| | - Philippe Cardot
- Centre de Recherche des Cordeliers, Université Pierre et Marie Curie-Paris 6, UMR S 872, France; INSERM, U 872, Paris, France; Université Paris Descartes-Paris 5, UMR S 872, France
| | - Marc Benderitter
- Institut de Radioprotection et de Sûreté Nucléaire, PRP-HOM, SRBE, Laboratoire de Recherche sur la Régénération des tissus sains Irradiés, Fontenay-aux-Roses, France
| | - Eric Deutsch
- Inserm U1030, Radiotherapie experimentale, Institut Gustave Roussy, Villejuif, France
| | - Jocelyne Aigueperse
- Institut de Radioprotection et de Sûreté Nucléaire, PRP-HOM, Fontenay-aux-Roses, France
| |
Collapse
|
30
|
De Mario A, Castellani A, Peggion C, Massimino ML, Lim D, Hill AF, Sorgato MC, Bertoli A. The prion protein constitutively controls neuronal store-operated Ca(2+) entry through Fyn kinase. Front Cell Neurosci 2015; 9:416. [PMID: 26578881 PMCID: PMC4623396 DOI: 10.3389/fncel.2015.00416] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 10/02/2015] [Indexed: 11/23/2022] Open
Abstract
The prion protein (PrPC) is a cell surface glycoprotein mainly expressed in neurons, whose misfolded isoforms generate the prion responsible for incurable neurodegenerative disorders. Whereas PrPC involvement in prion propagation is well established, PrPC physiological function is still enigmatic despite suggestions that it could act in cell signal transduction by modulating phosphorylation cascades and Ca2+ homeostasis. Because PrPC binds neurotoxic protein aggregates with high-affinity, it has also been proposed that PrPC acts as receptor for amyloid-β (Aβ) oligomers associated with Alzheimer’s disease (AD), and that PrPC-Aβ binding mediates AD-related synaptic dysfunctions following activation of the tyrosine kinase Fyn. Here, use of gene-encoded Ca2+ probes targeting different cell domains in primary cerebellar granule neurons (CGN) expressing, or not, PrPC, allowed us to investigate whether PrPC regulates store-operated Ca2+ entry (SOCE) and the implication of Fyn in this control. Our findings show that PrPC attenuates SOCE, and Ca2+ accumulation in the cytosol and mitochondria, by constitutively restraining Fyn activation and tyrosine phosphorylation of STIM1, a key molecular component of SOCE. This data establishes the existence of a PrPC-Fyn-SOCE triad in neurons. We also demonstrate that treating cerebellar granule and cortical neurons with soluble Aβ(1–42) oligomers abrogates the control of PrPC over Fyn and SOCE, suggesting a PrPC-dependent mechanizm for Aβ-induced neuronal Ca2+ dyshomeostasis.
Collapse
Affiliation(s)
- Agnese De Mario
- Department of Biomedical Science, University of Padova Padova, Italy
| | - Angela Castellani
- Department of Biomedical Science, University of Padova Padova, Italy
| | - Caterina Peggion
- Department of Biomedical Science, University of Padova Padova, Italy
| | | | - Dmitry Lim
- Department of Pharmaceutical Science, University of Piemonte Orientale Novara, Italy
| | - Andrew F Hill
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University Melbourne, VIC, Australia
| | - M Catia Sorgato
- Department of Biomedical Science, University of Padova Padova, Italy ; CNR Neuroscience Institute, University of Padova Padova, Italy
| | | |
Collapse
|
31
|
Zhao Y, Xu P, Hu S, Du L, Xu Z, Zhang H, Cui W, Mak S, Xu D, Shen J, Han Y, Liu Y, Xue M. Tanshinone II A, a multiple target neuroprotectant, promotes caveolae-dependent neuronal differentiation. Eur J Pharmacol 2015; 765:437-46. [DOI: 10.1016/j.ejphar.2015.09.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Revised: 09/02/2015] [Accepted: 09/03/2015] [Indexed: 01/19/2023]
|
32
|
Schmitz M, Zafar S, Silva CJ, Zerr I. Behavioral abnormalities in prion protein knockout mice and the potential relevance of PrP(C) for the cytoskeleton. Prion 2015; 8:381-6. [PMID: 25517431 DOI: 10.4161/19336896.2014.983746] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The cellular prion protein (PrP(C)) is a highly conserved protein, which is anchored to the outer surface of the plasma membrane. Even though its physiological function has already been investigated in different cell or mouse models where PrP(C) expression is either upregulated or depleted, its exact physiological role in a mammalian organism remains elusive. Recent studies indicate that PrP(C) has multiple functions and is involved in cognition, learning, anxiety, locomotion, depression, offensive aggression and nest building behavior. While young animals (3 months of age) show only marginal abnormalities, most of the deficits become apparent as the animals age, which might indicate its role in neurodegeneration or neuroprotection. However, the exact biochemical mechanism and signal transduction pathways involving PrP(C) are only gradually becoming clearer. We report the observations made in different studies using different Prnp0/0 mouse models and propose that PrP(C) plays an important role in the regulation of the cytoskeleton and associated proteins. In particular, we showed a nocodazole treatment influenced colocalization of PrP(C) and α tubulin 1. In addition, we confirmed the observed deficits in nest building using a different backcrossed Prnp0/0 mouse line.
Collapse
Affiliation(s)
- Matthias Schmitz
- a Department of Neurology ; University Medical Center Göttingen, and German Center for Neurodegenerative Diseases (DZNE) ; Göttingen , Germany
| | | | | | | |
Collapse
|
33
|
Slingshot-Cofilin activation mediates mitochondrial and synaptic dysfunction via Aβ ligation to β1-integrin conformers. Cell Death Differ 2015; 22:921-34. [PMID: 25698445 PMCID: PMC4423195 DOI: 10.1038/cdd.2015.5] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 11/19/2014] [Accepted: 12/16/2014] [Indexed: 02/08/2023] Open
Abstract
The accumulation of amyloid-β protein (Aβ) is an early event associated with synaptic and mitochondrial damage in Alzheimer's disease (AD). Recent studies have implicated the filamentous actin (F-actin) severing protein, Cofilin, in synaptic remodeling, mitochondrial dysfunction, and AD pathogenesis. However, whether Cofilin is an essential component of the AD pathogenic process and how Aβ impinges its signals to Cofilin from the neuronal surface are unknown. In this study, we found that Aβ42 oligomers (Aβ42O, amyloid-β protein 1–42 oligomers) bind with high affinity to low or intermediate activation conformers of β1-integrin, resulting in the loss of surface β1-integrin and activation of Cofilin via Slingshot homology-1 (SSH1) activation. Specifically, conditional loss of β1-integrin prevented Aβ42O-induced Cofilin activation, and allosteric modulation or activation of β1-integrin significantly reduced Aβ42O binding to neurons while blocking Aβ42O-induced reactive oxygen species (ROS) production, mitochondrial dysfunction, depletion of F-actin/focal Vinculin, and apoptosis. Cofilin, in turn, was required for Aβ42O-induced loss of cell surface β1-integrin, disruption of F-actin/focal Talin–Vinculin, and depletion of F-actin-associated postsynaptic proteins. SSH1 reduction, which mitigated Cofilin activation, prevented Aβ42O-induced mitochondrial Cofilin translocation and apoptosis, while AD brain mitochondria contained significantly increased activated/oxidized Cofilin. In mechanistic support in vivo, AD mouse model (APP (amyloid precursor protein)/PS1) brains contained increased SSH1/Cofilin and decreased SSH1/14-3-3 complexes, indicative of SSH1–Cofilin activation via release of SSH1 from 14-3-3. Finally, genetic reduction in Cofilin rescued APP/Aβ-induced synaptic protein loss and gliosis in vivo as well as deficits in long-term potentiation (LTP) and contextual memory in APP/PS1 mice. These novel findings therefore implicate the essential involvement of the β1-integrin–SSH1–Cofilin pathway in mitochondrial and synaptic dysfunction in AD.
Collapse
|
34
|
Xu L, Guo R, Xie Y, Ma M, Ye R, Liu X. Caveolae: molecular insights and therapeutic targets for stroke. Expert Opin Ther Targets 2015; 19:633-50. [PMID: 25639269 DOI: 10.1517/14728222.2015.1009446] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Caveolae are specialized plasma membrane micro-invaginations of most mammalian cell types. The organization and function of caveolae are carried out by their coat proteins, caveolins and adaptor proteins, cavins. Caveolae/caveolins physically interact with membrane-associated signaling molecules and function in cholesterol incorporation, signaling transduction and macromolecular transport/permeability. AREAS COVERED Recent investigations have implicated a check-and-balance role of caveolae in the pathophysiology of cerebral ischemia. Caveolin knockout mice displayed exacerbated ischemic injury, whereas caveolin peptide exerted remarkable protection against ischemia/reperfusion injury. This review attempts to provide a comprehensive synopsis of how caveolae/caveolins modulate blood-brain barrier permeability, pro-survival signaling, angiogenesis and neuroinflammation, and how this may contribute to a better understanding of the participation of caveolae in ischemic cascade. The role of caveolin in the preconditioning-induced tolerance against ischemia is also discussed. EXPERT OPINION Caveolae represent a novel target for cerebral ischemia. It remains open how to manipulate caveolin expression in a practical way to recapitulate the beneficial therapeutic outcomes. Caveolin peptides and associated antagomirs may be efficacious and deserve further investigations for their potential benefits for stroke.
Collapse
Affiliation(s)
- Lili Xu
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University , Nanjing 210002 , China
| | | | | | | | | | | |
Collapse
|
35
|
Sakudo A, Onodera T. Prion protein (PrP) gene-knockout cell lines: insight into functions of the PrP. Front Cell Dev Biol 2015; 2:75. [PMID: 25642423 PMCID: PMC4295555 DOI: 10.3389/fcell.2014.00075] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 12/22/2014] [Indexed: 11/13/2022] Open
Abstract
Elucidation of prion protein (PrP) functions is crucial to fully understand prion diseases. A major approach to studying PrP functions is the use of PrP gene-knockout (Prnp (-/-)) mice. So far, six types of Prnp (-/-) mice have been generated, demonstrating the promiscuous functions of PrP. Recently, other PrP family members, such as Doppel and Shadoo, have been found. However, information obtained from comparative studies of structural and functional analyses of these PrP family proteins do not fully reveal PrP functions. Recently, varieties of Prnp (-/-) cell lines established from Prnp (-/-) mice have contributed to the analysis of PrP functions. In this mini-review, we focus on Prnp (-/-) cell lines and summarize currently available Prnp (-/-) cell lines and their characterizations. In addition, we introduce the recent advances in the methodology of cell line generation with knockout or knockdown of the PrP gene. We also discuss how these cell lines have provided valuable insights into PrP functions and show future perspectives.
Collapse
Affiliation(s)
- Akikazu Sakudo
- Laboratory of Biometabolic Chemistry, Faculty of Medicine, School of Health Sciences, University of the Ryukyus Nishihara, Japan
| | - Takashi Onodera
- Research Center for Food Safety, School of Agricultural and Life Sciences, University of Tokyo Tokyo, Japan
| |
Collapse
|
36
|
Liebert A, Bicknell B, Adams R. Prion Protein Signaling in the Nervous System—A Review and Perspective. ACTA ACUST UNITED AC 2014. [DOI: 10.4137/sti.s12319] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Prion protein (PrPC) was originally known as the causative agent of transmissible spongiform encephalopathy (TSE) but with recent research, its true function in cells is becoming clearer. It is known to act as a scaffolding protein, binding multiple ligands at the cell membrane and to be involved in signal transduction, passing information from the extracellular matrix (ECM) to the cytoplasm. Its role in the coordination of transmitters at the synapse, glyapse, and gap junction and in short- and long-range neurotrophic signaling gives PrPC a major part in neural transmission and nervous system signaling. It acts to regulate cellular function in multiple targets through its role as a controller of redox status and calcium ion flux. Given the importance of PrPC in cell physiology, this review considers its potential role in disease apart from TSE. The putative functions of PrPC point to involvement in neurodegenerative disease, neuropathic pain, chronic headache, and inflammatory disease including neuroinflammatory disease of the nervous system. Potential targets for the treatment of disease influenced by PrPC are discussed.
Collapse
Affiliation(s)
- Ann Liebert
- Faculty of Health Science, University of Sydney, Australia
| | - Brian Bicknell
- Faculty of Health Science, Australian Catholic University, Australia
| | | |
Collapse
|
37
|
Zebrafish prion protein PrP2 controls collective migration process during lateral line sensory system development. PLoS One 2014; 9:e113331. [PMID: 25436888 PMCID: PMC4249873 DOI: 10.1371/journal.pone.0113331] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 10/27/2014] [Indexed: 12/05/2022] Open
Abstract
Prion protein is involved in severe neurodegenerative disorders but its physiological role is still in debate due to an absence of major developmental defects in knockout mice. Previous reports in zebrafish indicate that the two prion genes, PrP1 and PrP2, are both involved in several steps of embryonic development thus providing a unique route to discover prion protein function. Here we investigate the role of PrP2 during development of a mechano-sensory system, the posterior lateral line, using morpholino knockdown and PrP2 targeted inactivation. We confirm the efficiency of the translation blocking morpholino at the protein level. Development of the posterior lateral line is altered in PrP2 morphants, including nerve axonal outgrowth and primordium migration defects. Reduced neuromast deposition was observed in PrP2 morphants as well as in PrP2−/− mutants. Rosette formation defects were observed in PrP2 morphants, strongly suggesting an abnormal primordium organization and reflecting loss of cell cohesion during migration of the primordium. In addition, the adherens junction proteins, E-cadherin and ß-catenin, were mis-localized after reduction of PrP2 expression and thus contribute to the primordium disorganization. Consequently, hair cell differentiation and number were affected and this resulted in reduced functional neuromasts. At later developmental stages, myelination of the posterior lateral line nerve was altered. Altogether, our study reports an essential role of PrP2 in collective migration process of the primordium and in neuromast formation, further implicating a role for prion protein in cell adhesion.
Collapse
|
38
|
Ochs K, Málaga-Trillo E. Common themes in PrP signaling: the Src remains the same. Front Cell Dev Biol 2014; 2:63. [PMID: 25364767 PMCID: PMC4211543 DOI: 10.3389/fcell.2014.00063] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2014] [Accepted: 10/02/2014] [Indexed: 01/06/2023] Open
Abstract
The ability of the cellular prion protein (PrPC) to trigger intracellular signals appears central to neurodegeneration pathways, yet the physiological significance of such signals is rather puzzling. For instance, PrPC deregulation disrupts phenomena as diverse as synaptic transmission in mammals and cell adhesion in zebrafish. Although unrelated, the key proteins in these events -the NMDA receptor (NMDAR) and E-cadherin, respectively- are similarly modulated by the Src family kinase (SFK) Fyn. These observations highlight the importance of PrPC-mediated Fyn activation, a finding reported nearly two decades ago. Given their complex functions and regulation, SFKs may hold the key to intriguing aspects of PrP biology such as its seemingly promiscuous functions and the lack of strong phenotypes in knockout mice. Here we provide a mechanistic perspective on how SFKs might contribute to the uncertain molecular basis of neuronal PrP phenotypes affecting ion channel activity, axon myelination and olfactory function. In particular, we discuss SFK target proteins involved in these processes and the role of tyrosine phosphorylation in the regulation of their activity and cell surface expression.
Collapse
Affiliation(s)
- Katharina Ochs
- Department of Biology, University of Konstanz Konstanz, Germany
| | | |
Collapse
|
39
|
Mehrabian M, Ehsani S, Schmitt-Ulms G. An emerging role of the cellular prion protein as a modulator of a morphogenetic program underlying epithelial-to-mesenchymal transition. Front Cell Dev Biol 2014; 2:53. [PMID: 25453033 PMCID: PMC4233941 DOI: 10.3389/fcell.2014.00053] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 09/03/2014] [Indexed: 12/31/2022] Open
Abstract
Knowledge of phenotypic changes the cellular prion protein (PrP(C)) contributes to may provide novel avenues for understanding its function. Here we consider data from functional knockout/down studies and protein-protein interaction analyses from the perspective of PrP's relationship to its ancestral ZIP metal ion transporting proteins. When approached in this manner, a role of PrP(C) as a modulator of a complex morphogenetic program that underlies epithelial-to-mesenchymal transition (EMT) emerges. To execute EMT, cells have to master the challenge to shift from cell-cell to cell-substrate modes of adherence. During this process, cell-cell junctions stabilized by E-cadherins are replaced by focal adhesions that mediate cell-substrate contacts. A similar reprogramming occurs during distinct organogenesis events that have been shown to rely on ZIP transporters. A model is presented that sees ZIP transporters, and possibly also PrP(C), affect this balance of adherence modes at both the transcriptional and post-translational levels.
Collapse
Affiliation(s)
- Mohadeseh Mehrabian
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto Toronto, ON, Canada ; Department of Laboratory Medicine and Pathobiology, University of Toronto Toronto, ON, Canada
| | - Sepehr Ehsani
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto Toronto, ON, Canada ; Department of Laboratory Medicine and Pathobiology, University of Toronto Toronto, ON, Canada
| | - Gerold Schmitt-Ulms
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto Toronto, ON, Canada ; Department of Laboratory Medicine and Pathobiology, University of Toronto Toronto, ON, Canada
| |
Collapse
|
40
|
Petit CSV, Besnier L, Morel E, Rousset M, Thenet S. Roles of the cellular prion protein in the regulation of cell-cell junctions and barrier function. Tissue Barriers 2014; 1:e24377. [PMID: 24665391 PMCID: PMC3887058 DOI: 10.4161/tisb.24377] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 03/18/2013] [Accepted: 03/19/2013] [Indexed: 01/12/2023] Open
Abstract
The cellular prion protein was historically characterized owing to its misfolding in prion disease. Although its physiological role remains incompletely understood, PrP(C) has emerged as an evolutionary conserved, multifaceted protein involved in a wide-range of biological processes. PrP(C) is a GPI-anchored protein targeted to the plasma membrane, in raft microdomains, where its interaction with a repertoire of binding partners, which differ depending on cell models, mediates its functions. Among identified PrP(C) partners are cell adhesion molecules. This review will focus on the multiple implications of PrP(C) in cell adhesion processes, mainly the regulation of cell-cell junctions in epithelial and endothelial cells and the consequences on barrier properties. We will show how recent findings argue for a role of PrP(C) in the recruitment of signaling molecules, which in turn control the targeting or the stability of adhesion complexes at the plasma membrane.
Collapse
Affiliation(s)
- Constance S V Petit
- Centre de Recherche des Cordeliers; Université Pierre et Marie Curie; Paris, France ; INSERM; Paris, France ; Université Paris Descartes; Paris, France
| | - Laura Besnier
- Centre de Recherche des Cordeliers; Université Pierre et Marie Curie; Paris, France ; INSERM; Paris, France ; Université Paris Descartes; Paris, France
| | - Etienne Morel
- Centre de Recherche des Cordeliers; Université Pierre et Marie Curie; Paris, France ; INSERM; Paris, France ; Université Paris Descartes; Paris, France
| | - Monique Rousset
- Centre de Recherche des Cordeliers; Université Pierre et Marie Curie; Paris, France ; INSERM; Paris, France ; Université Paris Descartes; Paris, France
| | - Sophie Thenet
- Centre de Recherche des Cordeliers; Université Pierre et Marie Curie; Paris, France ; INSERM; Paris, France ; Université Paris Descartes; Paris, France ; Ecole Pratique des Hautes Etudes; Laboratoire de Pharmacologie Cellulaire et Moléculaire ; Paris, France
| |
Collapse
|
41
|
Hirsch TZ, Hernandez-Rapp J, Martin-Lannerée S, Launay JM, Mouillet-Richard S. PrP(C) signalling in neurons: from basics to clinical challenges. Biochimie 2014; 104:2-11. [PMID: 24952348 DOI: 10.1016/j.biochi.2014.06.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 06/10/2014] [Indexed: 01/05/2023]
Abstract
The cellular prion protein PrP(C) was identified over twenty-five years ago as the normal counterpart of the scrapie prion protein PrP(Sc), itself the main if not the sole component of the infectious agent at the root of Transmissible Spongiform Encephalopathies (TSEs). PrP(C) is a ubiquitous cell surface protein, abundantly expressed in neurons, which constitute the targets of PrP(Sc)-mediated toxicity. Converging evidence have highlighted that neuronal, GPI-anchored PrP(C) is absolutely required for prion-induced neuropathogenesis, which warrants investigating into the normal function exerted by PrP(C) in a neuronal context. It is now well-established that PrP(C) can serve as a cell signalling molecule, able to mobilize transduction cascades in response to interactions with partners. This function endows PrP(C) with the capacity to participate in multiple neuronal processes, ranging from survival to synaptic plasticity. A diverse array of data have allowed to shed light on how this function is corrupted by PrP(Sc). Recently, amyloid Aβ oligomers, whose accumulation is associated with Alzheimer's disease (AD), were shown to similarly instigate toxic events by deviating PrP(C)-mediated signalling. Here, we provide an overview of the various signal transduction cascades ascribed to PrP(C) in neurons, summarize how their subversion by PrP(Sc) or Aβ oligomers contributes to TSE or AD neuropathogenesis and discuss the ensuing clinical implications.
Collapse
Affiliation(s)
- Théo Z Hirsch
- INSERM UMR-S1124, 75006 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, UMR-S1124, 75006 Paris, France
| | - Julia Hernandez-Rapp
- INSERM UMR-S1124, 75006 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, UMR-S1124, 75006 Paris, France; Université Paris Sud 11, ED419 Biosigne, 91400 Orsay, France
| | - Séverine Martin-Lannerée
- INSERM UMR-S1124, 75006 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, UMR-S1124, 75006 Paris, France
| | - Jean-Marie Launay
- AP-HP Service de Biochimie, Fondation FondaMental, INSERM U942 Hôpital Lariboisière, 75010 Paris, France; Pharma Research Department, F. Hoffmann-La-Roche Ltd., CH-4070 Basel, Switzerland
| | - Sophie Mouillet-Richard
- INSERM UMR-S1124, 75006 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, UMR-S1124, 75006 Paris, France.
| |
Collapse
|
42
|
Yang X, Zhang Y, Zhang L, He T, Zhang J, Li C. Prion protein and cancers. Acta Biochim Biophys Sin (Shanghai) 2014; 46:431-40. [PMID: 24681883 DOI: 10.1093/abbs/gmu019] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The normal cellular prion protein, PrP(C) is a highly conserved and widely expressed cell surface glycoprotein in all mammals. The expression of PrP is pivotal in the pathogenesis of prion diseases; however, the normal physiological functions of PrP(C) remain incompletely understood. Based on the studies in cell models, a plethora of functions have been attributed to PrP(C). In this paper, we reviewed the potential roles that PrP(C) plays in cell physiology and focused on its contribution to tumorigenesis.
Collapse
Affiliation(s)
- Xiaowen Yang
- Department of the First Abdominal Surgery, Jiangxi Tumor Hospital, Nanchang 330029, China
| | - Yan Zhang
- Department of Molecular Endocrinology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Lihua Zhang
- Department of Pathology, Zhongda Hospital, Southeast University, Nanjing 210009, China
| | - Tianlin He
- Department of General Surgery, Changhai Hospital of Second Military Medical University, Shanghai 200433, China
| | - Jie Zhang
- Department of Stomatology, The First Affiliated Hospital of Shihezi University Medical College, Shihezi 832000, China
| | - Chaoyang Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| |
Collapse
|
43
|
Hernandez-Rapp J, Martin-Lannerée S, Hirsch TZ, Pradines E, Alleaume-Butaux A, Schneider B, Baudry A, Launay JM, Mouillet-Richard S. A PrP(C)-caveolin-Lyn complex negatively controls neuronal GSK3β and serotonin 1B receptor. Sci Rep 2014; 4:4881. [PMID: 24810941 PMCID: PMC4013941 DOI: 10.1038/srep04881] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 04/08/2014] [Indexed: 12/25/2022] Open
Abstract
The cellular prion protein, PrPC, is a glycosylphosphatidylinositol-anchored protein, abundant in lipid rafts and highly expressed in the brain. While PrPC is much studied for its involvement under its abnormal PrPSc isoform in Transmissible Spongiform Encephalopathies, its physiological role remains unclear. Here, we report that GSK3β, a multifunctional kinase whose inhibition is neuroprotective, is a downstream target of PrPC signalling in serotonergic neuronal cells. We show that the PrPC-dependent inactivation of GSK3β is relayed by a caveolin-Lyn platform located on neuronal cell bodies. Furthermore, the coupling of PrPC to GSK3β potentiates serotonergic signalling by altering the distribution and activity of the serotonin 1B receptor (5-HT1BR), a receptor that limits neurotransmitter release. In vivo, our data reveal an increased GSK3β kinase activity in PrP-deficient mouse brain, as well as sustained 5-HT1BR activity, whose inhibition promotes an anxiogenic behavioural response. Collectively, our data unveil a new facet of PrPC signalling that strengthens neurotransmission.
Collapse
Affiliation(s)
- Julia Hernandez-Rapp
- 1] INSERM UMR-S1124, 75006 Paris France [2] Université Paris Descartes, Sorbonne Paris Cité, UMR-S1124, 75006 Paris France [3] Université Paris Sud 11, ED419 Biosigne, 91400 Orsay, France [4]
| | - Séverine Martin-Lannerée
- 1] INSERM UMR-S1124, 75006 Paris France [2] Université Paris Descartes, Sorbonne Paris Cité, UMR-S1124, 75006 Paris France [3]
| | - Théo Z Hirsch
- 1] INSERM UMR-S1124, 75006 Paris France [2] Université Paris Descartes, Sorbonne Paris Cité, UMR-S1124, 75006 Paris France [3]
| | - Elodie Pradines
- 1] INSERM UMR-S1124, 75006 Paris France [2] Université Paris Descartes, Sorbonne Paris Cité, UMR-S1124, 75006 Paris France
| | - Aurélie Alleaume-Butaux
- 1] INSERM UMR-S1124, 75006 Paris France [2] Université Paris Descartes, Sorbonne Paris Cité, UMR-S1124, 75006 Paris France
| | - Benoît Schneider
- 1] INSERM UMR-S1124, 75006 Paris France [2] Université Paris Descartes, Sorbonne Paris Cité, UMR-S1124, 75006 Paris France
| | - Anne Baudry
- 1] INSERM UMR-S1124, 75006 Paris France [2] Université Paris Descartes, Sorbonne Paris Cité, UMR-S1124, 75006 Paris France
| | - Jean-Marie Launay
- 1] AP-HP Service de Biochimie, Fondation FondaMental, INSERM U942 Hôpital Lariboisière, 75010 Paris, France [2] Pharma Research Department, F. Hoffmann-La-Roche Ltd., CH-4070 Basel, Switzerland
| | - Sophie Mouillet-Richard
- 1] INSERM UMR-S1124, 75006 Paris France [2] Université Paris Descartes, Sorbonne Paris Cité, UMR-S1124, 75006 Paris France
| |
Collapse
|
44
|
Loss of prion protein leads to age-dependent behavioral abnormalities and changes in cytoskeletal protein expression. Mol Neurobiol 2014; 50:923-36. [PMID: 24604355 DOI: 10.1007/s12035-014-8655-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 01/29/2014] [Indexed: 12/13/2022]
Abstract
The cellular prion protein (PrPC) is a highly conserved protein whose exact physiological role remains elusive. In the present study, we investigated age-dependent behavioral abnormalities in PrPC-knockout (Prnp0/0) mice and wild-type (WT) controls. Prnp0/0 mice showed age-dependent behavioral deficits in memory performance, associative learning, basal anxiety, and nest building behavior. Using a hypothesis-free quantitative proteomic investigation, we found that loss of PrPC affected the levels of neurofilament proteins in an age-dependent manner. In order to understand the biochemical basis of these observations, we analyzed the phosphorylation status of neurofilament heavy chain (NF-H). We found a reduction in NF-H phosphorylation in both Prnp0/0 mice and in PrPC-deficient cells. The expression of Fyn and phospho-Fyn, a potential regulator for NF phosphorylation, was associated with PrPC ablation. The number of β-tubulin III-positive neurons in the hippocampus was diminished in Prnp0/0 mice relative to WT mice. These data indicate that PrPC plays an important role in cytoskeletal organization, brain function, and age-related neuroprotection. Our work represents the first direct biochemical link between these proteins and the observed behavioral phenotypes.
Collapse
|
45
|
Chen RJ, Chang WW, Lin YC, Cheng PL, Chen YR. Alzheimer's amyloid-β oligomers rescue cellular prion protein induced tau reduction via the Fyn pathway. ACS Chem Neurosci 2013; 4:1287-96. [PMID: 23805846 DOI: 10.1021/cn400085q] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Amyloid-β (Aβ) and tau are the pathogenic hallmarks in Alzheimer's disease (AD). Aβ oligomers are considered the actual toxic entities, and the toxicity relies on the presence of tau. Recently, Aβ oligomers have been shown to specifically interact with cellular prion protein (PrP(C)) where the role of PrP(C) in AD is still not fully understood. To investigate the downstream mechanism of PrP(C) and Aβ oligomer interaction and their possible relationships to tau, we examined tau expression in human neuroblastoma BE(2)-C cells transfected with murine PrP(C) and studied the effect under Aβ oligomer treatment. By Western blotting, we found that PrP(C) overexpression down-regulated tau protein and Aβ oligomer binding alleviated the tau reduction induced by wild type but not M128V PrP(C), the high AD risk polymorphic allele in human prion gene. PrP(C) lacking the Aβ oligomer binding site was incapable of rescuing the level of tau reduction. Quantitative RT-PCR showed the PrP(C) effect was attributed to tau reduction at the transcription level. Treatment with Fyn pathway inhibitors, Fyn kinase inhibitor PP2 and MEK inhibitor U0126, reversed the PrP(C)-induced tau reduction and Aβ oligomer treatment modulated Fyn kinase activity. The results suggested Fyn pathway regulated Aβ-PrP(C)-tau signaling. Overall, our results demonstrated that PrP(C) down-regulated tau via the Fyn pathway and the effect can be regulated by Aβ oligomers. Our study facilitated the understanding of molecular mechanisms among PrP(C), tau, and Aβ oligomers.
Collapse
Affiliation(s)
- Rong-Jie Chen
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Wei-Wei Chang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Yu-Chun Lin
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Pei-Lin Cheng
- Institute
of Molecular Biology, Academia Sinica,
Taipei, Taiwan
| | - Yun-Ru Chen
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
46
|
Zhao CF, Liu Y, Ni YL, Yang JW, Hui HD, Sun ZB, Liu SJ. SCIRR39 promotes neurite extension via RhoA in NGF-induced PC12 cells. Dev Neurosci 2013; 35:373-83. [PMID: 24021527 DOI: 10.1159/000350715] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 03/17/2013] [Indexed: 11/19/2022] Open
Abstract
SCIRR39 is an identified upregulated gene in rat primary neuron injury and/or regeneration process with roles largely unexplored. Using real-time quantitative PCR, Western blotting and immunofluorescence, SCIRR39 expression was detected in normal PC12 cells and upregulated in differentiated cells. The results of cell proliferation by Cell Counting Kit and cell cycle by flow cytometry indicated that SCIRR39 inhibited cell proliferation and induced the decrease in S phase. Importantly, immunofluorescent and RhoA pull-down assays showed that SCIRR39 strongly affected the neurite extension of NGF-treated PC12 cells through a RhoA-dependent mechanism, but the truncated mutants of SCIRR39 containing a truncation from 141AA to 211AA or from 397AA to 424AA failed to mock the SCIRR39 effect on neurite extension. Moreover, change of SCIRR39 expression in NGF-treated PC12 cells regulated the expression and phosphorylation of Fyn, a regulator of RhoA activity, but not the expression of ROCK II protein. Finally, immunofluorescence and RhoA pull-down assays revealed that obvious inhibition of neurite extension by SCIRR39 shRNA was reversed by RhoA inhibitor C3-transferase. Our results indicated that SCIRR39 increased the neurite extension in NGF-treated PC12 cells via RhoA, suggesting that SCIRR39 contributes to the regeneration of neuron injury by specifically altering the differentiation program.
Collapse
Affiliation(s)
- C F Zhao
- State Key Laboratory of Proteomics, Department of Neurobiology, Institute of Basic Medical Sciences, The Academy of Military Medical Sciences, Beijing, PR China
| | | | | | | | | | | | | |
Collapse
|
47
|
Wang H, Ren CH, Gunawardana CG, Schmitt-Ulms G. Overcoming barriers and thresholds - signaling of oligomeric Aβ through the prion protein to Fyn. Mol Neurodegener 2013; 8:24. [PMID: 23856335 PMCID: PMC3722066 DOI: 10.1186/1750-1326-8-24] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 07/09/2013] [Indexed: 12/20/2022] Open
Abstract
Evidence has been mounting for an involvement of the prion protein (PrP) in a molecular pathway assumed to play a critical role in the etiology of Alzheimer disease. A currently popular model sees oligomeric amyloid β (oAβ) peptides bind directly to PrP to emanate a signal that causes activation of the cytoplasmic tyrosine kinase Fyn, an essential player in a cascade of events that ultimately leads to NMDA receptor-mediated excitotoxicity and hyper-phosphorylation of tau. The model does not reveal, however, how extracellular binding of oAβ to PrP is communicated across the plasma membrane barrier to affect activation of Fyn. A scenario whereby PrP may adapt a transmembrane topology to affect Fyn activation in the absence of additional partners is currently not supported by evidence. A survey of known candidate PrP interactors leads to a small number of molecules that are known to acquire a transmembrane topology and understood to contribute to Fyn activation. Because multiple signaling pathways converge onto Fyn, a realistic model needs to take into account a reality of Fyn acting as a hub that integrates signals from multiple inhibitory and activating effectors. To clarify the role of PrP in oAβ-dependent excitotoxicity, future studies may need to incorporate experimental designs that can probe the contributions of Fyn modulator pathways and rely on analogous readouts, rather than threshold effects, known to underlie excitotoxic signaling.
Collapse
Affiliation(s)
- Hansen Wang
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Tanz Neuroscience Building, 6 Queen's Park Crescent West, Toronto, Ontario M5S 3H2, Canada
| | | | | | | |
Collapse
|
48
|
Atorvastatin stimulates neuroblastoma cells to induce neurite outgrowth by increasing cellular prion protein expression. Neurosci Lett 2012; 531:114-9. [PMID: 23127852 DOI: 10.1016/j.neulet.2012.10.032] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Revised: 10/03/2012] [Accepted: 10/05/2012] [Indexed: 12/26/2022]
Abstract
Recently, 3-hydroxy-3-methyl glutaryl coenzyme A (HMG-CoA) reductase inhibitors were reported to induce neurite outgrowth in vitro. However, the mechanism underlying this effect remains unclear. Cellular prion protein (PrP(C)) is a ubiquitous glycoprotein present on the surfaces of various cells, including neurons, and is suggested to be involved in neurite outgrowth. Therefore, the present study aimed to determine whether PrP(C) mediates neurite outgrowth induced by HMG-CoA reductase inhibitors. Atorvastatin, a strong HMG-CoA reductase inhibitor, induced neurite outgrowth and increased PrP(C) levels in Neuro2a cells in a time- and dose-dependent manner. PrP(C) mRNA expression was also increased by atorvastatin. Farnesol, a non-sterol mevalonate derivative, attenuated the atorvastatin-induced neurite outgrowth and increase in PrP(C). Neuro2a cells overexpressing PrP(C) showed a remarkable enhancement of atorvastatin-induced neurite outgrowth compared with mock cells transfected with empty pCI-neo vector. These findings suggest that PrP(C) contributes, at least in part, to atorvastatin-induced neurite outgrowth. This phenomenon may be included among the mechanisms underlying decreased risk of Alzheimer's disease in patients treated with HMG-CoA reductase inhibitors.
Collapse
|
49
|
Chen S, Charness ME. Ethanol disrupts axon outgrowth stimulated by netrin-1, GDNF, and L1 by blocking their convergent activation of Src family kinase signaling. J Neurochem 2012; 123:602-12. [PMID: 22924694 DOI: 10.1111/j.1471-4159.2012.07954.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Revised: 08/23/2012] [Accepted: 08/23/2012] [Indexed: 01/17/2023]
Abstract
Pre-natal alcohol exposure causes fetal alcohol spectrum disorders (FASD), the most common, preventable cause of developmental disability. The developing cerebellum is particularly vulnerable to the effects of ethanol. We reported that ethanol inhibits the stimulation of axon outgrowth in cerebellar granule neurons (CGN) by NAP, an active motif of activity-dependent neuroprotective protein (ADNP), by blocking NAP activation of Fyn kinase and its downstream signaling molecule, the scaffolding protein Cas. Here, we asked whether ethanol inhibits the stimulation of axon outgrowth by diverse axon guidance molecules through a common action on the Src family kinases (SFK). We first demonstrated that netrin-1, glial cell line-derived neurotrophic factor (GDNF), and neural cell adhesion molecule L1 stimulate axon outgrowth in CGNs by activating SFK, Cas, and extracellular signal-regulated kinase 1 and 2 (ERK1/2). The specific SFK inhibitor, PP2, blocked the stimulation of axon outgrowth and the activation of the SFK-Cas-ERK1/2 signaling pathway by each of these axon-guidance molecules. In contrast, brain-derived neurotrophic factor (BDNF) stimulated axon outgrowth and activated ERK1/2 without first activating SFK or Cas. Clinically relevant concentrations of ethanol inhibited axon outgrowth and the activation of the SFK-Cas-ERK1/2 pathway by netrin-1, GDNF, and L1, but did not disrupt BDNF-induced axon outgrowth or ERK1/2 activation. These results indicate that SFK, but not ERK1/2, is a primary target for ethanol inhibition of axon outgrowth. The ability of ethanol to block the convergent activation of the SFK-Cas-ERK1/2 pathway by netrin-1, GDNF, L1, and ADNP could contribute significantly to the pathogenesis of FASD.
Collapse
Affiliation(s)
- Suzhen Chen
- VA Boston Healthcare System and Department of Neurology, Harvard Medical School, West Roxbury, MA, USA
| | | |
Collapse
|
50
|
Um JW, Strittmatter SM. Amyloid-β induced signaling by cellular prion protein and Fyn kinase in Alzheimer disease. Prion 2012; 7:37-41. [PMID: 22987042 DOI: 10.4161/pri.22212] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Alzheimer disease (AD) is the most prevalent cause of dementia. Amyloid-β (Aβ) oligomers are potent synaptotoxins thought to mediate AD-related phenotypes. Cellular prion protein (PrP(C)) has been identified as a high-affinity receptor for Aβ oligomers. Herein, we review the functional consequences of Aβ oligomer binding to PrP(C) on the neuronal surface. We highlight recent evidence that Fyn kinase mediates signal transduction downstream of the PrP(C)-Aβ oligomer complex. These studies suggest that PrP(C) has a central role in AD pathogenesis and may provide a target for therapeutic intervention in AD.
Collapse
Affiliation(s)
- Ji Won Um
- Cellular Neuroscience, Neurodegeneration and Repair Program, Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | | |
Collapse
|