1
|
Bader J, Brigger F, Leroux JC. Extracellular vesicles versus lipid nanoparticles for the delivery of nucleic acids. Adv Drug Deliv Rev 2024:115461. [PMID: 39490384 DOI: 10.1016/j.addr.2024.115461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
Extracellular vesicles (EVs) are increasingly investigated for delivering nucleic acid (NA) therapeutics, leveraging their natural role in transporting NA and protein-based cargo in cell-to-cell signaling. Their synthetic counterparts, lipid nanoparticles (LNPs), have been developed over the past decades as NA carriers, culminating in the approval of several marketed formulations such as patisiran/Onpattro® and the mRNA-1273/BNT162 COVID-19 vaccines. The success of LNPs has sparked efforts to develop innovative technologies to target extrahepatic organs, and to deliver novel therapeutic modalities, such as tools for in vivo gene editing. Fueled by the recent advancements in both fields, this review aims to provide a comprehensive overview of the basic characteristics of EV and LNP-based NA delivery systems, from EV biogenesis to structural properties of LNPs. It addresses the primary challenges encountered in utilizing these nanocarriers from a drug formulation and delivery perspective. Additionally, biodistribution profiles, in vitro and in vivo transfection outcomes, as well as their status in clinical trials are compared. Overall, this review provides insights into promising research avenues and potential dead ends for EV and LNP-based NA delivery systems.
Collapse
Affiliation(s)
- Johannes Bader
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Finn Brigger
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Jean-Christophe Leroux
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland.
| |
Collapse
|
2
|
Korde DS, Humpel C. A Combination of Heavy Metals and Intracellular Pathway Modulators Induces Alzheimer Disease-like Pathologies in Organotypic Brain Slices. Biomolecules 2024; 14:165. [PMID: 38397402 PMCID: PMC10887098 DOI: 10.3390/biom14020165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/17/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that is characterized by amyloid-beta (Aβ) plaques and tau neurofibrillary tangles (NFT). Modelling aspects of AD is challenging due to its complex multifactorial etiology and pathology. The present study aims to establish a cost-effective and rapid method to model the two primary pathologies in organotypic brain slices. Coronal hippocampal brain slices (150 µm) were generated from postnatal (day 8-10) C57BL6 wild-type mice and cultured for 9 weeks. Collagen hydrogels containing either an empty load or a mixture of human Aβ42 and P301S aggregated tau were applied to the slices. The media was further supplemented with various intracellular pathway modulators or heavy metals to augment the appearance of Aβ plaques and tau NFTs, as assessed by immunohistochemistry. Immunoreactivity for Aβ and tau was significantly increased in the ventral areas in slices with a mixture of human Aβ42 and P301S aggregated tau compared to slices with empty hydrogels. Aβ plaque- and tau NFT-like pathologies could be induced independently in slices. Heavy metals (aluminum, lead, cadmium) potently augmented Aβ plaque-like pathology, which developed intracellularly prior to cell death. Intracellular pathway modulators (scopolamine, wortmannin, MHY1485) significantly boosted tau NFT-like pathologies. A combination of nanomolar concentrations of scopolamine, wortmannin, MHY1485, lead, and cadmium in the media strongly increased Aβ plaque- and tau NFT-like immunoreactivity in ventral areas compared to the slices with non-supplemented media. The results highlight that we could harness the potential of the collagen hydrogel-based spreading of human Aβ42 and P301S aggregated tau, along with pharmacological manipulation, to produce pathologies relevant to AD. The results offer a novel ex vivo organotypic slice model to investigate AD pathologies with potential applications for screening drugs or therapies in the future.
Collapse
Affiliation(s)
| | - Christian Humpel
- Laboratory of Psychiatry and Experimental Alzheimer’s Research, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| |
Collapse
|
3
|
Bodart-Santos V, Pinheiro LS, da Silva-Junior AJ, Froza RL, Ahrens R, Gonçalves RA, Andrade MM, Chen Y, Alcantara CDL, Grinberg LT, Leite REP, Ferreira ST, Fraser PE, De Felice FG. Alzheimer's disease brain-derived extracellular vesicles reveal altered synapse-related proteome and induce cognitive impairment in mice. Alzheimers Dement 2023; 19:5418-5436. [PMID: 37204850 DOI: 10.1002/alz.13134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 03/15/2023] [Accepted: 04/17/2023] [Indexed: 05/20/2023]
Abstract
INTRODUCTION Extracellular vesicles (EVs) have been implicated in the spread of neuropathology in Alzheimer's disease (AD), but their involvement in behavioral outcomes linked to AD remains to be determined. METHODS EVs isolated from post mortem brain tissue from control, AD, or frontotemporal dementia (FTD) donors, as well as from APP/PS1 mice, were injected into the hippocampi of wild-type (WT) or a humanized Tau mouse model (hTau/mTauKO). Memory tests were carried out. Differentially expressed proteins in EVs were assessed by proteomics. RESULTS Both AD-EVs and APP/PS1-EVs trigger memory impairment in WT mice. We further demonstrate that AD-EVs and FTD-EVs carry Tau protein, present altered protein composition associated with synapse regulation and transmission, and trigger memory impairment in hTau/mTauKO mice. DISCUSSION Results demonstrate that AD-EVs and FTD-EVs have negative impacts on memory in mice and suggest that, in addition to spreading pathology, EVs may contribute to memory impairment in AD and FTD. HIGHLIGHTS Aβ was detected in EVs from post mortem AD brain tissue and APP/PS1 mice. Tau was enriched in EVs from post mortem AD, PSP and FTD brain tissue. AD-derived EVs and APP/PS1-EVs induce cognitive impairment in wild-type (WT) mice. AD- and FTD-derived EVs induce cognitive impairment in humanized Tau mice. Proteomics findings associate EVs with synapse dysregulation in tauopathies.
Collapse
Affiliation(s)
- Victor Bodart-Santos
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
| | - Lisandra S Pinheiro
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Almir J da Silva-Junior
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rudimar L Froza
- Oswaldo Cruz Institute, Oswaldo Cruz Foundation, FIOCRUZ, Rio de Janeiro, Brazil
| | - Rosemary Ahrens
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
| | - Rafaella A Gonçalves
- Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences and Department of Psychiatry, Queen's University, Kingston, Canada
| | - Mayara M Andrade
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Yan Chen
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
| | - Carolina de Lima Alcantara
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lea T Grinberg
- Department of Pathology, University of São Paulo Medical School, Sao Paulo, Brazil
- Memory and Aging Center, Department of Neurology and Pathology, University of California San Francisco, San Francisco, California, USA
| | - Renata E P Leite
- Department of Pathology, University of São Paulo Medical School, Sao Paulo, Brazil
| | - Sergio T Ferreira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paul E Fraser
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Fernanda G De Felice
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences and Department of Psychiatry, Queen's University, Kingston, Canada
- D'OR Institute for Research and Education, Rio de Janeiro, Brazil
| |
Collapse
|
4
|
Braun JEA. Extracellular chaperone networks and the export of J-domain proteins. J Biol Chem 2023; 299:102840. [PMID: 36581212 PMCID: PMC9867986 DOI: 10.1016/j.jbc.2022.102840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/18/2022] [Accepted: 12/20/2022] [Indexed: 12/27/2022] Open
Abstract
An extracellular network of molecular chaperones protects a diverse array of proteins that reside in or pass through extracellular spaces. Proteins in the extracellular milieu face numerous challenges that can lead to protein misfolding and aggregation. As a checkpoint for proteins that move between cells, extracellular chaperone networks are of growing clinical relevance. J-domain proteins (JDPs) are ubiquitous molecular chaperones that are known for their essential roles in a wide array of fundamental cellular processes through their regulation of heat shock protein 70s. As the largest molecular chaperone family, JDPs have long been recognized for their diverse functions within cells. Some JDPs are elegantly selective for their "client proteins," some do not discriminate among substrates and others act cooperatively on the same target. The realization that JDPs are exported through both classical and unconventional secretory pathways has fueled investigation into the roles that JDPs play in protein quality control and intercellular communication. The proposed functions of exported JDPs are diverse. Studies suggest that export of DnaJB11 enhances extracellular proteostasis, that intercellular movement of DnaJB1 or DnaJB6 enhances the proteostasis capacity in recipient cells, whereas the import of DnaJB8 increases resistance to chemotherapy in recipient cancer cells. In addition, the export of DnaJC5 and concurrent DnaJC5-dependent ejection of dysfunctional and aggregation-prone proteins are implicated in the prevention of neurodegeneration. This review provides a brief overview of the current understanding of the extracellular chaperone networks and outlines the first wave of studies describing the cellular export of JDPs.
Collapse
Affiliation(s)
- Janice E A Braun
- Department of Biochemistry and Molecular Biology, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
5
|
Whitehead M, Yusoff S, Ahmad S, Schmidt L, Mayr M, Madine J, Middleton D, Shanahan CM. Vascular smooth muscle cell senescence accelerates medin aggregation via small extracellular vesicle secretion and extracellular matrix reorganization. Aging Cell 2023; 22:e13746. [PMID: 36433666 PMCID: PMC9924949 DOI: 10.1111/acel.13746] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 11/07/2022] [Accepted: 11/13/2022] [Indexed: 11/27/2022] Open
Abstract
Vascular amyloidosis, caused when peptide monomers aggregate into insoluble amyloid, is a prevalent age-associated pathology. Aortic medial amyloid (AMA) is the most common human amyloid and is composed of medin, a 50-amino acid peptide. Emerging evidence has implicated extracellular vesicles (EVs) as mediators of pathological amyloid accumulation in the extracellular matrix (ECM). To determine the mechanisms of AMA formation with age, we explored the impact of vascular smooth muscle cell (VSMC) senescence, EV secretion, and ECM remodeling on medin accumulation. Medin was detected in EVs secreted from primary VSMCs. Small, round medin aggregates colocalized with EV markers in decellularized ECM in vitro and medin was shown on the surface of EVs deposited in the ECM. Decreasing EV secretion with an inhibitor attenuated aggregation and deposition of medin in the ECM. Medin accumulation in the aortic wall of human subjects was strongly correlated with age and VSMC senescence increased EV secretion, increased EV medin loading and triggered deposition of fibril-like medin. Proteomic analysis showed VSMC senescence induced changes in EV cargo and ECM composition, which led to enhanced EV-ECM binding and accelerated medin aggregation. Abundance of the proteoglycan, HSPG2, was increased in the senescent ECM and colocalized with EVs and medin. Isolated EVs selectively bound to HSPG2 in the ECM and its knock-down decreased formation of fibril-like medin structures. These data identify VSMC-derived EVs and HSPG2 in the ECM as key mediators of medin accumulation, contributing to age-associated AMA development.
Collapse
Affiliation(s)
- Meredith Whitehead
- School of Cardiovascular and Metabolic Medicine & SciencesKing's College LondonLondonUK
| | - Syabira Yusoff
- School of Cardiovascular and Metabolic Medicine & SciencesKing's College LondonLondonUK
| | - Sadia Ahmad
- School of Cardiovascular and Metabolic Medicine & SciencesKing's College LondonLondonUK
| | - Lukas Schmidt
- School of Cardiovascular and Metabolic Medicine & SciencesKing's College LondonLondonUK
| | - Manuel Mayr
- School of Cardiovascular and Metabolic Medicine & SciencesKing's College LondonLondonUK
| | - Jillian Madine
- Institute of Systems, Molecular and Integrative BiologyUniversity of LiverpoolLondonUK
| | | | - Catherine M. Shanahan
- School of Cardiovascular and Metabolic Medicine & SciencesKing's College LondonLondonUK
| |
Collapse
|
6
|
Takasugi N, Komai M, Kaneshiro N, Ikeda A, Kamikubo Y, Uehara T. The Pursuit of the "Inside" of the Amyloid Hypothesis-Is C99 a Promising Therapeutic Target for Alzheimer's Disease? Cells 2023; 12:454. [PMID: 36766796 PMCID: PMC9914381 DOI: 10.3390/cells12030454] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/27/2023] [Accepted: 01/28/2023] [Indexed: 02/04/2023] Open
Abstract
Aducanumab, co-developed by Eisai (Japan) and Biogen (U.S.), has received Food and Drug Administration approval for treating Alzheimer's disease (AD). In addition, its successor antibody, lecanemab, has been approved. These antibodies target the aggregated form of the small peptide, amyloid-β (Aβ), which accumulates in the patient brain. The "amyloid hypothesis" based therapy that places the aggregation and toxicity of Aβ at the center of the etiology is about to be realized. However, the effects of immunotherapy are still limited, suggesting the need to reconsider this hypothesis. Aβ is produced from a type-I transmembrane protein, Aβ precursor protein (APP). One of the APP metabolites, the 99-amino acids C-terminal fragment (C99, also called βCTF), is a direct precursor of Aβ and accumulates in the AD patient's brain to demonstrate toxicity independent of Aβ. Conventional drug discovery strategies have focused on Aβ toxicity on the "outside" of the neuron, but C99 accumulation might explain the toxicity on the "inside" of the neuron, which was overlooked in the hypothesis. Furthermore, the common region of C99 and Aβ is a promising target for multifunctional AD drugs. This review aimed to outline the nature, metabolism, and impact of C99 on AD pathogenesis and discuss whether it could be a therapeutic target complementing the amyloid hypothesis.
Collapse
Affiliation(s)
- Nobumasa Takasugi
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan
- Department of Cellular and Molecular Pharmacology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo Bunkyo-ku, Tokyo 113-8421, Japan
| | - Masato Komai
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan
| | - Nanaka Kaneshiro
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA
- Center for RNA Biology and Medicine, University of California, Riverside, CA 92521, USA
| | - Atsuya Ikeda
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan
| | - Yuji Kamikubo
- Department of Cellular and Molecular Pharmacology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo Bunkyo-ku, Tokyo 113-8421, Japan
| | - Takashi Uehara
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan
| |
Collapse
|
7
|
Cheng L, Hill AF. Therapeutically harnessing extracellular vesicles. Nat Rev Drug Discov 2022; 21:379-399. [PMID: 35236964 DOI: 10.1038/s41573-022-00410-w] [Citation(s) in RCA: 310] [Impact Index Per Article: 155.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2022] [Indexed: 02/06/2023]
Abstract
The field of extracellular vesicle (EV) research has developed rapidly over the last decade from the study of fundamental biology to a subject of significant clinical relevance. The potential of harnessing EVs in the diagnosis and treatment of diseases - including cancer and neurological and cardiovascular disorders - is now being recognized. Accordingly, the applications of EVs as therapeutic targets, biomarkers, novel drug delivery agents and standalone therapeutics are being actively explored. This Review provides a brief overview of the characteristics and physiological functions of the various classes of EV, focusing on their association with disease and emerging strategies for their therapeutic exploitation.
Collapse
Affiliation(s)
- Lesley Cheng
- La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia
| | - Andrew F Hill
- La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia. .,Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia.
| |
Collapse
|
8
|
Kaur S, Verma H, Dhiman M, Tell G, Gigli GL, Janes F, Mantha AK. Brain Exosomes: Friend or Foe in Alzheimer's Disease? Mol Neurobiol 2021; 58:6610-6624. [PMID: 34595669 DOI: 10.1007/s12035-021-02547-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 08/23/2021] [Indexed: 01/18/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease. It is known to be a multifactorial disease and several causes are associated with its occurrence as well as progression. However, the accumulation of amyloid beta (Aβ) is widely considered its major pathogenic hallmark. Additionally, neurofibrillary tangles (NFT), mitochondrial dysfunction, oxidative stress, and aging (cellular senescence) are considered as additional hits affecting the disease pathology. Several studies are now suggesting important role of inflammation in AD, which shifts our thought towards the brain's resident immune cells, microglia, and astrocytes; how they interact with neurons; and how these interactions are affected by intra and extracellular stressful factors. These interactions can be modulated by different mechanisms and pathways, in which exosomes could play an important role. Exosomes are multivesicular bodies secreted by nearly all types of cells. The exosomes secreted by glial cells or neurons affect the interactions and thus the physiology of these cells by transmitting miRNAs, proteins, and lipids. Exosomes can serve as a friend or foe to the neuron function, depending upon the carried signals. Exosomes, from the healthy microenvironment, may assist neuron function and health, whereas, from the stressed microenvironment, they carry oxidative and inflammatory signals to the neurons and thus prove detrimental to the neuronal function. Furthermore, exosomes can cross the blood-brain barrier (BBB), and from the blood plasma they can enter the brain cells and activate microglia and astrocytes. Exosomes can transport Aβ or Tau, cytokines, miRNAs between the cells, and alter the physiology of recipient cells. They can also assist in Aβ clearance and regulation of synaptic activity. The exosomes derived from different cells play different roles, and this field is still in its infancy stage. This review advocates exosomes' role as a friend or foe in neurodegenerative diseases, especially in the case of Alzheimer's disease.
Collapse
Affiliation(s)
- Sharanjot Kaur
- Department of Microbiology, School of Biological Sciences , Central University of Punjab, Bathinda, Punjab, India
| | - Harkomal Verma
- Department of Zoology, School of Biological Sciences, Central University of Punjab, Village Ghudda151 401, Punjab, Bathinda, India
| | - Monisha Dhiman
- Department of Microbiology, School of Biological Sciences , Central University of Punjab, Bathinda, Punjab, India
| | - Gianluca Tell
- Department of Medicine, University of Udine, Udine, Italy
| | - Gian Luigi Gigli
- Department of Medicine, University of Udine, Udine, Italy
- Clinical Neurology, Udine University Hospital, Udine, Italy
| | | | - Anil K Mantha
- Department of Zoology, School of Biological Sciences, Central University of Punjab, Village Ghudda151 401, Punjab, Bathinda, India.
| |
Collapse
|
9
|
Torres VI, Barrera DP, Varas-Godoy M, Arancibia D, Inestrosa NC. Selective Surface and Intraluminal Localization of Wnt Ligands on Small Extracellular Vesicles Released by HT-22 Hippocampal Neurons. Front Cell Dev Biol 2021; 9:735888. [PMID: 34722516 PMCID: PMC8548728 DOI: 10.3389/fcell.2021.735888] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 09/23/2021] [Indexed: 12/12/2022] Open
Abstract
The Wnt signaling pathway induces various responses underlying the development and maturation of the nervous system. Wnt ligands are highly hydrophobic proteins that limit their diffusion through an aqueous extracellular medium to a target cell. Nevertheless, their attachment to small extracellular vesicles-like exosomes is one of the described mechanisms that allow their transport under this condition. Some Wnt ligands in these vehicles are expected to be dependent on post-translational modifications such as acylation. The mechanisms determining Wnt loading in exosomes and delivery to the target cells are largely unknown. Here, we took advantage of a cell model that secret a highly enriched population of small extracellular vesicles (sEVs), hippocampal HT-22 neurons. First, to establish the cell model, we characterized the morphological and biochemical properties of an enriched fraction of sEVs obtained from hippocampal HT-22 neurons that express NCAM-L1, a specific exosomal neuronal marker. Transmission electron microscopy showed a highly enriched fraction of exosome-like vesicles. Next, the exosomal presence of Wnt3a, Wnt5a, and Wnt7a was confirmed by western blot analysis and electron microscopy combined with immunogold. Also, we studied whether palmitoylation is a necessary post-translational modification for the transport Wnt in these vesicles. We found that proteinase-K treatment of exosomes selectively decreased their Wnt5a and Wnt7a content, suggesting that their expression is delimited to the exterior membrane surface. In contrast, Wnt3a remained attached, suggesting that it is localized within the exosome lumen. On the other hand, Wnt-C59, a specific inhibitor of porcupine O-acyltransferase (PORCN), decreased the association of Wnt with exosomes, suggesting that Wnt ligand acylation is necessary for them to be secreted by exosomes. These findings may help to understand the action of the Wnt ligands in the target cell, which could be defined during the packaging of the ligands in the secretory cell sEVs.
Collapse
Affiliation(s)
- Viviana I Torres
- Departamento Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Daniela P Barrera
- Centro de Envejecimiento y Regeneración (CARE UC), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Manuel Varas-Godoy
- Cancer Cell Biology Laboratory, Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Duxan Arancibia
- Departamento Biomédico, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, Chile
| | - Nibaldo C Inestrosa
- Centro de Envejecimiento y Regeneración (CARE UC), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| |
Collapse
|
10
|
Vandendriessche C, Bruggeman A, Van Cauwenberghe C, Vandenbroucke RE. Extracellular Vesicles in Alzheimer's and Parkinson's Disease: Small Entities with Large Consequences. Cells 2020; 9:cells9112485. [PMID: 33203181 PMCID: PMC7696752 DOI: 10.3390/cells9112485] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 02/07/2023] Open
Abstract
Alzheimer’s disease (AD) and Parkinson’s disease (PD) are incurable, devastating neurodegenerative disorders characterized by the formation and spreading of protein aggregates throughout the brain. Although the exact spreading mechanism is not completely understood, extracellular vesicles (EVs) have been proposed as potential contributors. Indeed, EVs have emerged as potential carriers of disease-associated proteins and are therefore thought to play an important role in disease progression, although some beneficial functions have also been attributed to them. EVs can be isolated from a variety of sources, including biofluids, and the analysis of their content can provide a snapshot of ongoing pathological changes in the brain. This underlines their potential as biomarker candidates which is of specific relevance in AD and PD where symptoms only arise after considerable and irreversible neuronal damage has already occurred. In this review, we discuss the known beneficial and detrimental functions of EVs in AD and PD and we highlight their promising potential to be used as biomarkers in both diseases.
Collapse
Affiliation(s)
- Charysse Vandendriessche
- VIB Center for Inflammation Research, VIB, 9052 Ghent, Belgium; (C.V.); (A.B.); (C.V.C.)
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Arnout Bruggeman
- VIB Center for Inflammation Research, VIB, 9052 Ghent, Belgium; (C.V.); (A.B.); (C.V.C.)
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
- Department of Neurology, Ghent University Hospital, 9000 Ghent, Belgium
| | - Caroline Van Cauwenberghe
- VIB Center for Inflammation Research, VIB, 9052 Ghent, Belgium; (C.V.); (A.B.); (C.V.C.)
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Roosmarijn E. Vandenbroucke
- VIB Center for Inflammation Research, VIB, 9052 Ghent, Belgium; (C.V.); (A.B.); (C.V.C.)
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
- Correspondence: ; Tel.: +32-9-3313730
| |
Collapse
|
11
|
Astoricchio E, Alfano C, Rajendran L, Temussi PA, Pastore A. The Wide World of Coacervates: From the Sea to Neurodegeneration. Trends Biochem Sci 2020; 45:706-717. [PMID: 32417131 DOI: 10.1016/j.tibs.2020.04.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/30/2020] [Accepted: 04/16/2020] [Indexed: 12/12/2022]
Abstract
The formation of immiscible liquid phases or coacervates is a phenomenon widely observed in biology. Marine organisms, for instance, use liquid-liquid phase separation (LLPS) as the precursor phase to form various fibrillar or crustaceous materials that are essential for surface adhesion. More recently, the importance of LLPS has been realized in the compartmentalization of living cells and in obtaining ordered but dynamic partitions that can be reversed according to necessity. Here, we compare the properties, features, and peculiarities of intracellular and extracellular coacervates, drawing parallels and learning from the differences. A more general view of the phenomenon may in the future inform new studies to allow a better comprehension of its laws.
Collapse
Affiliation(s)
- Emanuele Astoricchio
- UK Dementia Research Institute at the Wohl Institute of the King's College London, London SE5 9RT, UK
| | | | - Lawrence Rajendran
- UK Dementia Research Institute at the Wohl Institute of the King's College London, London SE5 9RT, UK
| | - Piero Andrea Temussi
- UK Dementia Research Institute at the Wohl Institute of the King's College London, London SE5 9RT, UK.
| | - Annalisa Pastore
- UK Dementia Research Institute at the Wohl Institute of the King's College London, London SE5 9RT, UK.
| |
Collapse
|
12
|
Vanherle S, Haidar M, Irobi J, Bogie JF, Hendriks JJ. Extracellular vesicle-associated lipids in central nervous system disorders. Adv Drug Deliv Rev 2020; 159:322-331. [PMID: 32360577 DOI: 10.1016/j.addr.2020.04.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/03/2020] [Accepted: 04/26/2020] [Indexed: 12/19/2022]
Abstract
Increasing evidence indicates that lipid metabolism is disturbed in central nervous system (CNS) disorders, such as multiple sclerosis, Alzheimer's, and Parkinson's disease. Extracellular vesicles (EVs), including exosomes and microvesicles, are nanosized particles that play an essential role in intercellular communication and tissue homeostasis by transporting diverse biologically active molecules, including a large variety of lipid species. In the last decade, studies defined that changes in the EV lipidome closely correlate with disease-progression and -remission in CNS disorders. In this review, we summarize and discuss these changes in the EV lipidome and elaborate on the impact of different EV-associated lipids on pathological processes in CNS disorders. We conclude that EV-associated lipids are closely associated with neuroinflammation, CNS repair, and pathological protein aggregation in CNS disorders, and that modulation of the EV lipidome represents a promising therapeutic strategy to halt disease progression in multiple sclerosis, Alzheimer's, and Parkinson's disease. Moreover, we predict that disease-stage specific EV-associated lipid signatures can be invaluable markers for the diagnosis and early detection of CNS disorders in the future.
Collapse
|
13
|
Angelopoulou E, Paudel YN, Shaikh MF, Piperi C. Flotillin: A Promising Biomarker for Alzheimer's Disease. J Pers Med 2020; 10:jpm10020020. [PMID: 32225073 PMCID: PMC7354424 DOI: 10.3390/jpm10020020] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 03/19/2020] [Accepted: 03/20/2020] [Indexed: 12/18/2022] Open
Abstract
Alzheimer’s disease (AD) is characterized by the accumulation of beta amyloid (Aβ) in extracellular senile plaques and intracellular neurofibrillary tangles (NFTs) mainly consisting of tau protein. Although the exact etiology of the disease remains elusive, accumulating evidence highlights the key role of lipid rafts, as well as the endocytic pathways in amyloidogenic amyloid precursor protein (APP) processing and AD pathogenesis. The combination of reduced Aβ42 levels and increased phosphorylated tau protein levels in the cerebrospinal fluid (CSF) is the most well established biomarker, along with Pittsburgh compound B and positron emission tomography (PiB-PET) for amyloid imaging. However, their invasive nature, the cost, and their availability often limit their use. In this context, an easily detectable marker for AD diagnosis even at preclinical stages is highly needed. Flotillins, being hydrophobic proteins located in lipid rafts of intra- and extracellular vesicles, are mainly involved in signal transduction and membrane–protein interactions. Accumulating evidence highlights the emerging implication of flotillins in AD pathogenesis, by affecting APP endocytosis and processing, Ca2+ homeostasis, mitochondrial dysfunction, neuronal apoptosis, Aβ-induced neurotoxicity, and prion-like spreading of Aβ. Importantly, there is also clinical evidence supporting their potential use as biomarker candidates for AD, due to reduced serum and CSF levels that correlate with amyloid burden in AD patients compared with controls. This review focuses on the emerging preclinical and clinical evidence on the role of flotillins in AD pathogenesis, further addressing their potential usage as disease biomarkers.
Collapse
Affiliation(s)
- Efthalia Angelopoulou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Yam Nath Paudel
- Neuropharmacology Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor, Malaysia;
| | - Mohd. Farooq Shaikh
- Neuropharmacology Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor, Malaysia;
- Correspondence: (M.F.S.); (C.P.)
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
- Correspondence: (M.F.S.); (C.P.)
| |
Collapse
|
14
|
Gill J, Mustapic M, Diaz-Arrastia R, Lange R, Gulyani S, Diehl T, Motamedi V, Osier N, Stern RA, Kapogiannis D. Higher exosomal tau, amyloid-beta 42 and IL-10 are associated with mild TBIs and chronic symptoms in military personnel. Brain Inj 2018; 32:1277-1284. [PMID: 29913077 DOI: 10.1080/02699052.2018.1471738] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Identify biomarkers in peripheral blood that relate to chronic post-concussive and behavioural symptoms following traumatic brain injuries (TBIs) to ultimately improve clinical management. RESEARCH DESIGN We compared military personnel with mild TBIs (mTBIs) (n = 42) to those without TBIs (n = 22) in concentrations of tau, amyloid-beta (Aβ42) and cytokines (tumour necrosis factor alpha (TNFα, interleukin (IL)-6 and -10) in neuronal-derived exosomes from the peripheral blood. We utilized nanosight technology coupled with ultra-sensitivity immunoassay methods. We also examined the impact of post-concussive and behavioural symptoms including depression and post-traumatic stress disorder (PTSD) on these neuronal-derived markers. RESULTS We report that concentrations of exosomal tau (F1, 62 = 10.50), Aβ42 (F1, 61 = 5.32) and IL-10 (F1, 59 = 4.32) were elevated in the mTBI group compared to the controls. Within the mTBI group, regression models show that post-concussive symptoms were most related to exosomal tau elevations, whereas exosomal IL-10 levels were related to PTSD symptoms. CONCLUSIONS These findings suggest that chronic post-concussive symptoms following an mTBI relate to altered exosomal activity, and that greater tau pathology may underlie chronic post-concussive symptoms that develop following mTBIs. It also suggests that central inflammatory activity contributes to PTSD symptoms following an mTBI, providing necessary insights into the role of inflammation in chronic PTSD symptoms.
Collapse
Affiliation(s)
- Jessica Gill
- a Tissue Injury Branch, National Institutes of Health, National Institute of Nursing Research , Bethesda , MD , United States
| | - Maja Mustapic
- b Aging, National Institutes of Health, National Institute of Aging , Baltimore , MD , United States
| | - Ramon Diaz-Arrastia
- c Department of Neurology, School of Medicine , University of Pennsylvania , Philadelphia , PA , United States
| | - Rael Lange
- d Defense and Veterans Brain Injury Center , Walter Reed National Military Medical Center , Bethesda , MD , United States
| | - Seema Gulyani
- b Aging, National Institutes of Health, National Institute of Aging , Baltimore , MD , United States
| | - Tom Diehl
- b Aging, National Institutes of Health, National Institute of Aging , Baltimore , MD , United States
| | - Vida Motamedi
- a Tissue Injury Branch, National Institutes of Health, National Institute of Nursing Research , Bethesda , MD , United States
| | - Nicole Osier
- a Tissue Injury Branch, National Institutes of Health, National Institute of Nursing Research , Bethesda , MD , United States
| | - Robert A Stern
- e Neurosurgery, and Anatomy & Neurobiology , Boston University, Boston University Alzheimer's Disease and CTE Center , Boston , MA , United States
| | - Dimitrios Kapogiannis
- b Aging, National Institutes of Health, National Institute of Aging , Baltimore , MD , United States
| |
Collapse
|
15
|
Paolicelli RC, Bergamini G, Rajendran L. Cell-to-cell Communication by Extracellular Vesicles: Focus on Microglia. Neuroscience 2018; 405:148-157. [PMID: 29660443 DOI: 10.1016/j.neuroscience.2018.04.003] [Citation(s) in RCA: 239] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 03/30/2018] [Accepted: 04/04/2018] [Indexed: 12/20/2022]
Abstract
Extracellular vesicles, including exosomes and microvesicles, are small, nano-to-micrometer vesicles that are released from cells. While initially observed in immune cells and reticulocytes as vesicles meant to remove archaic proteins, now they have been observed in almost all cell types of multicellular organisms. Growing evidence indicates that extracellular vesicles, containing lipids, proteins and RNAs, represent an efficient way to transfer functional cargoes from one cell to another. In the central nervous system, the extensive cross-talk ongoing between neurons and glia, including microglia, the immune cells of the brain, takes advantage of secreted vesicles, which mediate intercellular communication over long range distance. Recent literature supports a critical role for extracellular vesicles in mediating complex and coordinated communication among neurons, astrocytes and microglia, both in the healthy and in the diseased brain. In this review, we focus on the biogenesis and function of microglia-related extracellular vesicles and focus on their putative role in Alzheimer's disease pathology.
Collapse
Affiliation(s)
- Rosa C Paolicelli
- Systems and Cell Biology of Neurodegeneration, IREM - Institute for Regenerative Medicine, University of Zurich, Switzerland.
| | - Giorgio Bergamini
- Preclinical Laboratory for Translational Research into Affective Disorders (PLaTRAD), Department of Psychiatry, Psychotherapy and Psychosomatics, University of Zurich, Switzerland
| | - Lawrence Rajendran
- Systems and Cell Biology of Neurodegeneration, IREM - Institute for Regenerative Medicine, University of Zurich, Switzerland
| |
Collapse
|
16
|
Del Prete D, Suski JM, Oulès B, Debayle D, Gay AS, Lacas-Gervais S, Bussiere R, Bauer C, Pinton P, Paterlini-Bréchot P, Wieckowski MR, Checler F, Chami M. Localization and Processing of the Amyloid-β Protein Precursor in Mitochondria-Associated Membranes. J Alzheimers Dis 2018; 55:1549-1570. [PMID: 27911326 PMCID: PMC5181669 DOI: 10.3233/jad-160953] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Alteration of mitochondria-associated membranes (MAMs) has been proposed to contribute to the pathogenesis of Alzheimer’s disease (AD). We studied herein the subcellular distribution, the processing, and the protein interactome of the amyloid-β protein precursor (AβPP) and its proteolytic products in MAMs. We reveal that AβPP and its catabolites are present in MAMs in cellular models overexpressing wild type AβPP or AβPP harboring the double Swedish or London familial AD mutations, and in brains of transgenic mice model of AD. Furthermore, we evidenced that both β- and γ-secretases are present and harbor AβPP processing activities in MAMs. Interestingly, cells overexpressing APPswe show increased ER-mitochondria contact sites. We also document increased neutral lipid accumulation linked to Aβ production and reversed by inhibiting β- or γ-secretases. Using a proteomic approach, we show that AβPP and its catabolites interact with key proteins of MAMs controlling mitochondria and ER functions. These data highlight the role of AβPP processing and proteomic interactome in MAMs deregulation taking place in AD.
Collapse
Affiliation(s)
- Dolores Del Prete
- Université Côte d'Azur, INSERM, CNRS, IPMC, France, Laboratory of Excellence DistALZ, Sophia-Antipolis, Valbonne, France.,Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jan M Suski
- Department of Biochemistry, Nencki Institute of Experimental Biology, Warsaw, Poland.,CCMA-Université de Nice-Sophia-Antipolis, Nice, France
| | - Bénédicte Oulès
- Centre for Stem Cells and Regenerative Medicine, King's College London, London, UK
| | - Delphine Debayle
- Université Côte d'Azur, INSERM, CNRS, IPMC, France, Laboratory of Excellence DistALZ, Sophia-Antipolis, Valbonne, France
| | - Anne Sophie Gay
- Université Côte d'Azur, INSERM, CNRS, IPMC, France, Laboratory of Excellence DistALZ, Sophia-Antipolis, Valbonne, France
| | | | - Renaud Bussiere
- Université Côte d'Azur, INSERM, CNRS, IPMC, France, Laboratory of Excellence DistALZ, Sophia-Antipolis, Valbonne, France
| | - Charlotte Bauer
- Université Côte d'Azur, INSERM, CNRS, IPMC, France, Laboratory of Excellence DistALZ, Sophia-Antipolis, Valbonne, France
| | - Paolo Pinton
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, University of Ferrara, Ferrara, Italy
| | | | - Mariusz R Wieckowski
- Department of Biochemistry, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Frédéric Checler
- Université Côte d'Azur, INSERM, CNRS, IPMC, France, Laboratory of Excellence DistALZ, Sophia-Antipolis, Valbonne, France
| | - Mounia Chami
- Université Côte d'Azur, INSERM, CNRS, IPMC, France, Laboratory of Excellence DistALZ, Sophia-Antipolis, Valbonne, France
| |
Collapse
|
17
|
Williamson RL, Laulagnier K, Miranda AM, Fernandez MA, Wolfe MS, Sadoul R, Di Paolo G. Disruption of amyloid precursor protein ubiquitination selectively increases amyloid β (Aβ) 40 levels via presenilin 2-mediated cleavage. J Biol Chem 2017; 292:19873-19889. [PMID: 29021256 PMCID: PMC5712626 DOI: 10.1074/jbc.m117.818138] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Revised: 09/30/2017] [Indexed: 11/06/2022] Open
Abstract
Amyloid plaques, a neuropathological hallmark of Alzheimer's disease, are largely composed of amyloid β (Aβ) peptide, derived from cleavage of amyloid precursor protein (APP) by β- and γ-secretases. The endosome is increasingly recognized as an important crossroad for APP and these secretases, with major implications for APP processing and amyloidogenesis. Among various post-translational modifications affecting APP accumulation, ubiquitination of cytodomain lysines may represent a key signal controlling APP endosomal sorting. Here, we show that substitution of APP C-terminal lysines with arginine disrupts APP ubiquitination and that an increase in the number of substituted lysines tends to increase APP metabolism. An APP mutant lacking all C-terminal lysines underwent the most pronounced increase in processing, leading to accumulation of both secreted and intracellular Aβ40. Artificial APP ubiquitination with rapalog-mediated proximity inducers reduced Aβ40 generation. A lack of APP C-terminal lysines caused APP redistribution from endosomal intraluminal vesicles (ILVs) to the endosomal limiting membrane, with a subsequent decrease in APP C-terminal fragment (CTF) content in secreted exosomes, but had minimal effects on APP lysosomal degradation. Both the increases in secreted and intracellular Aβ40 were abolished by depletion of presenilin 2 (PSEN2), recently shown to be enriched on the endosomal limiting membrane compared with PSEN1. Our findings demonstrate that ubiquitin can act as a signal at five cytodomain-located lysines for endosomal sorting of APP. They further suggest that disruption of APP endosomal sorting reduces its sequestration in ILVs and results in PSEN2-mediated processing of a larger pool of APP-CTF on the endosomal membrane.
Collapse
Affiliation(s)
| | - Karine Laulagnier
- the Grenoble Institut des Neurosciences, Inserm, Grenoble 38042, France
| | - André M Miranda
- From the Department of Pathology and Cell Biology and
- the Life and Health Sciences Research Institute, School of Medicine, University of Minho and
- Life and Health Sciences Research Institute/3B's Research Group-Biomaterials, Biodegradables, and Biomimetics Associate Laboratory, 4710-057 Braga/Guimarães, Portugal, and
| | - Marty A Fernandez
- the Center for Neurologic Diseases, Harvard University, Boston, Massachusetts 02115
| | - Michael S Wolfe
- the Center for Neurologic Diseases, Harvard University, Boston, Massachusetts 02115
| | - Rémy Sadoul
- the Grenoble Institut des Neurosciences, Inserm, Grenoble 38042, France
| | - Gilbert Di Paolo
- From the Department of Pathology and Cell Biology and
- the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, New York 10032
| |
Collapse
|
18
|
Eitan E, Hutchison ER, Marosi K, Comotto J, Mustapic M, Nigam SM, Suire C, Maharana C, Jicha GA, Liu D, Machairaki V, Witwer KW, Kapogiannis D, Mattson MP. Extracellular Vesicle-Associated Aβ Mediates Trans-Neuronal Bioenergetic and Ca 2+-Handling Deficits in Alzheimer's Disease Models. NPJ Aging Mech Dis 2016; 2:16019. [PMID: 27928512 PMCID: PMC5137253 DOI: 10.1038/npjamd.2016.19] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 04/21/2016] [Accepted: 06/05/2016] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's Disease (AD) is an age-related neurodegenerative disorder in which aggregation-prone neurotoxic amyloid β-peptide (Aβ) accumulates in the brain. Extracellular vesicles (EVs) are small 50-150 nanometer membrane vesicles that have recently been implicated in the prion-like spread of self-aggregating proteins. Here we report that EVs isolated from AD patient CSF and plasma, from the plasma of two AD mouse models, and from the medium of neural cells expressing familial AD presenilin 1 mutations, destabilize neuronal Ca2+ homeostasis, impair mitochondrial function, and sensitize neurons to excitotoxicity. EVs contain a relatively low amount of Aβ but have an increased Aβ42/ Aβ40 ratio; the majority of Aβ is located on the surface of the EVs. Impairment of lysosome function results in increased generation EVs with elevated Aβ42 levels. EVs may mediate transcellular spread of pathogenic Aβ species and that impair neuronal Ca2+ handling and mitochondrial function, and may thereby render neurons vulnerable to excitotoxicity.
Collapse
Affiliation(s)
- Erez Eitan
- Laboratory of Neurosciences, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Emmette R Hutchison
- Laboratory of Neurosciences, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Krisztina Marosi
- Laboratory of Neurosciences, National Institute on Aging, NIH, Baltimore, MD, USA
| | - James Comotto
- Laboratory of Neurosciences, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Maja Mustapic
- Laboratory of Neurosciences, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Saket M Nigam
- Laboratory of Neurosciences, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Caitlin Suire
- Laboratory of Neurosciences, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Chinmoyee Maharana
- Laboratory of Neurosciences, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Gregory A Jicha
- Sanders-Brown Center on Aging, and Department of Neurology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Dong Liu
- Laboratory of Neurosciences, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Vasiliki Machairaki
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kenneth W Witwer
- Department of Molecular and Comparative Pathobiology and Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Mark P Mattson
- Laboratory of Neurosciences, National Institute on Aging, NIH, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
19
|
van Niel G. Study of Exosomes Shed New Light on Physiology of Amyloidogenesis. Cell Mol Neurobiol 2016; 36:327-42. [PMID: 26983829 DOI: 10.1007/s10571-016-0357-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 02/27/2016] [Indexed: 12/18/2022]
Abstract
Accumulation of toxic amyloid oligomers, a key feature in the pathogenesis of amyloid-related diseases, results from an imbalance between generation and clearance of amyloidogenic proteins. Cell biology has brought to light the key roles of multivesicular endosomes (MVEs) and their intraluminal vesicles (ILVs), which can be secreted as exosomes, in amyloid generation and clearance. To better understand these roles, we have investigated a relevant physiological model of amyloid formation in pigment cells. These cells have tuned their endosomes to optimize the formation of functional amyloid fibrils from the premelanosome protein (PMEL) and to avoid potential accumulation of toxic species. The functional amyloids derived from PMEL reveal striking analogies with the generation of Aβ peptides. We have recently strengthened these analogies using extracellular vesicles as reporters of the endosomal processes that regulate PMEL melanogenesis. We have shown that in pigmented cells, apolipoprotein E (ApoE) is associated with ILVs and exosomes, and regulates the formation of PMEL amyloid fibrils in endosomes. This process secures the generation of amyloid fibrils by exploiting ILVs as amyloid-nucleating platforms. This physiological model of amyloidogenesis could shed new light on the roles of MVEs and exosomes in conditions with pathological amyloid metabolism, such as Alzheimer's disease.
Collapse
Affiliation(s)
- Guillaume van Niel
- Institut Curie, PSL Research University, UMR144, Centre de Recherche, 26 rue d'ULM, 75231, Paris, France.
- Centre National de la Recherche Scientifique, UMR144, 75248, Paris, France.
| |
Collapse
|
20
|
Bourdenx M, Koulakiotis NS, Sanoudou D, Bezard E, Dehay B, Tsarbopoulos A. Protein aggregation and neurodegeneration in prototypical neurodegenerative diseases: Examples of amyloidopathies, tauopathies and synucleinopathies. Prog Neurobiol 2015. [PMID: 26209472 DOI: 10.1016/j.pneurobio.2015.07.003] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Alzheimer's and Parkinson's diseases are the most prevalent neurodegenerative diseases that generate important health-related direct and indirect socio-economic costs. They are characterized by severe neuronal losses in several disease-specific brain regions associated with deposits of aggregated proteins. In Alzheimer's disease, β-amyloid peptide-containing plaques and intraneuronal neurofibrillary tangles composed of hyperphosphorylated microtubule-associated protein tau are the two main neuropathological lesions, while Parkinson's disease is defined by the presence of Lewy Bodies that are intraneuronal proteinaceous cytoplasmic inclusions. α-Synuclein has been identified as a major protein component of Lewy Bodies and heavily implicated in the pathogenesis of Parkinson's disease. In the past few years, evidence has emerged to explain how these aggregate-prone proteins can undergo spontaneous self-aggregation, propagate from cell to cell, and mediate neurotoxicity. Current research now indicates that oligomeric forms are probably the toxic species. This article discusses recent progress in the understanding of the pathogenesis of these diseases, with a focus on the underlying mechanisms of protein aggregation, and emphasizes the pathophysiological molecular mechanisms leading to cellular toxicity. Finally, we present the putative direct link between β-amyloid peptide and tau in causing toxicity in Alzheimer's disease as well as α-synuclein in Parkinson's disease, along with some of the most promising therapeutic strategies currently in development for those incurable neurodegenerative disorders.
Collapse
Affiliation(s)
- Mathieu Bourdenx
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| | | | - Despina Sanoudou
- National and Kapodistrian University of Athens Medical School, Department of Internal Medicine, 75 Mikras Asias Street, Athens 11527, Greece
| | - Erwan Bezard
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| | - Benjamin Dehay
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France.
| | - Anthony Tsarbopoulos
- GAIA Research Center, Bioanalytical Department, The Goulandris Natural History Museum, Kifissia 14562, Greece; National and Kapodistrian University of Athens Medical School, Department of Pharmacology, 75 Mikras Asias Street, Athens 11527, Greece.
| |
Collapse
|
21
|
John BA, Meister M, Banning A, Tikkanen R. Flotillins bind to the dileucine sorting motif of β-site amyloid precursor protein-cleaving enzyme 1 and influence its endosomal sorting. FEBS J 2014; 281:2074-87. [DOI: 10.1111/febs.12763] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 02/12/2014] [Accepted: 02/19/2014] [Indexed: 11/27/2022]
Affiliation(s)
- Bincy A. John
- Institute of Biochemistry; Medical Faculty; University of Giessen; Germany
| | - Melanie Meister
- Institute of Biochemistry; Medical Faculty; University of Giessen; Germany
| | - Antje Banning
- Institute of Biochemistry; Medical Faculty; University of Giessen; Germany
| | - Ritva Tikkanen
- Institute of Biochemistry; Medical Faculty; University of Giessen; Germany
| |
Collapse
|
22
|
Peggion C, Sorgato MC, Bertoli A. Prions and prion-like pathogens in neurodegenerative disorders. Pathogens 2014; 3:149-63. [PMID: 25437612 PMCID: PMC4235734 DOI: 10.3390/pathogens3010149] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 01/24/2014] [Accepted: 02/01/2014] [Indexed: 12/24/2022] Open
Abstract
Prions are unique elements in biology, being able to transmit biological information from one organism to another in the absence of nucleic acids. They have been identified as self-replicating proteinaceous agents responsible for the onset of rare and fatal neurodegenerative disorders—known as transmissible spongiform encephalopathies, or prion diseases—which affect humans and other animal species. More recently, it has been proposed that other proteins associated with common neurodegenerative disorders, such as Alzheimer’s and Parkinson’s disease, can self-replicate like prions, thus sustaining the spread of neurotoxic entities throughout the nervous system. Here, we review findings that have contributed to expand the prion concept, and discuss if the involved toxic species can be considered bona fide prions, including the capacity to infect other organisms, or whether these pathogenic aggregates share with prions only the capability to self-replicate.
Collapse
Affiliation(s)
- Caterina Peggion
- Department of Biomedical Sciences, University of Padova, Viale G. Colombo 3, Padova 35131, Italy.
| | - Maria Catia Sorgato
- Department of Biomedical Sciences, University of Padova, Viale G. Colombo 3, Padova 35131, Italy.
| | - Alessandro Bertoli
- Department of Biomedical Sciences, University of Padova, Viale G. Colombo 3, Padova 35131, Italy.
| |
Collapse
|
23
|
Bitsikas V, Riento K, Howe JD, Barry NP, Nichols BJ. The role of flotillins in regulating aβ production, investigated using flotillin 1-/-, flotillin 2-/- double knockout mice. PLoS One 2014; 9:e85217. [PMID: 24465508 PMCID: PMC3897416 DOI: 10.1371/journal.pone.0085217] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 11/28/2013] [Indexed: 11/18/2022] Open
Abstract
Flotillin 1 and flotillin 2 associate in the plasma membrane to form microdomains that have roles in cell signaling, regulation of cell-cell contacts, membrane-cytoskeletal interactions, and endocytosis. They are thought to be involved in the trafficking and hence processing of the Amyloid Precursor Protein, APP. In this study we set out to obtain in vivo confirmation of a link between flotillins and cleavage of APP to release amyloidogenic Aβ peptide, and to generate tools that would allow us to ask whether flotillins are functionally redundant. We used a mouse model for Aβ-dependent cerebral amyloidosis, APPPS1 mice, combined with deletion of either flotillin 1 singly, or flotillin 1 and flotillin 2 together. There was a small but significant reduction in Aβ levels, and the abundance of congo-red stained plaques, in brains of 12 week old mice lacking flotillin 1. A similar reduction in Aβ levels was observed in the flotillin 1-/-, flotillin 2-/- double knockouts. We did not observe large effects on the clustering or endocytosis of APP in flotillin 1-/- mouse embryonic fibroblasts. We conclude that flotillins are likely to play some role in APP trafficking or processing, but the relevant cellular mechanisms require more investigation. The availability of flotillin 1-/-, flotillin 2-/- mice, which have no overt phenotypes, will facilitate research into flotillin function in vivo.
Collapse
Affiliation(s)
- Vassilis Bitsikas
- Laboratory of Molecular Biology, Medical Research Council, Cambridge, Cambridgeshire, United Kingdom
| | - Kirsi Riento
- Laboratory of Molecular Biology, Medical Research Council, Cambridge, Cambridgeshire, United Kingdom
| | - Jonathan D. Howe
- Laboratory of Molecular Biology, Medical Research Council, Cambridge, Cambridgeshire, United Kingdom
| | - Nicholas P. Barry
- Laboratory of Molecular Biology, Medical Research Council, Cambridge, Cambridgeshire, United Kingdom
| | - Benjamin J. Nichols
- Laboratory of Molecular Biology, Medical Research Council, Cambridge, Cambridgeshire, United Kingdom
- * E-mail:
| |
Collapse
|
24
|
Armato U, Chiarini A, Chakravarthy B, Chioffi F, Pacchiana R, Colarusso E, Whitfield JF, Dal Prà I. Calcium-sensing receptor antagonist (calcilytic) NPS 2143 specifically blocks the increased secretion of endogenous Aβ42 prompted by exogenous fibrillary or soluble Aβ25-35 in human cortical astrocytes and neurons-therapeutic relevance to Alzheimer's disease. BIOCHIMICA ET BIOPHYSICA ACTA 2013; 1832:1634-52. [PMID: 23628734 DOI: 10.1016/j.bbadis.2013.04.020] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 03/28/2013] [Accepted: 04/18/2013] [Indexed: 11/17/2022]
Abstract
The "amyloid-β (Aβ) hypothesis" posits that accumulating Aβ peptides (Aβs) produced by neurons cause Alzheimer's disease (AD). However, the Aβs contribution by the more numerous astrocytes remains undetermined. Previously we showed that fibrillar (f)Aβ25-35, an Aβ42 proxy, evokes a surplus endogenous Aβ42 production/accumulation in cortical adult human astrocytes. Here, by using immunocytochemistry, immunoblotting, enzymatic assays, and highly sensitive sandwich ELISA kits, we investigated the effects of fAβ25-35 and soluble (s)Aβ25-35 on Aβ42 and Aβ40 accumulation/secretion by human cortical astrocytes and HCN-1A neurons and, since the calcium-sensing receptor (CaSR) binds Aβs, their modulation by NPS 2143, a CaSR allosteric antagonist (calcilytic). The fAβ25-35-exposed astrocytes and surviving neurons produced, accumulated, and secreted increased amounts of Aβ42, while Aβ40 also accrued but its secretion was unchanged. Accordingly, secreted Aβ42/Aβ40 ratio values rose for astrocytes and neurons. While slightly enhancing Aβ40 secretion by fAβ25-35-treated astrocytes, NPS 2143 specifically suppressed the fAβ25-35-elicited surges of endogenous Aβ42 secretion by astrocytes and neurons. Therefore, NPS 2143 addition always kept Aβ42/Aβ40 values to baseline or lower levels. Mechanistically, NPS 2143 decreased total CaSR protein complement, transiently raised proteasomal chymotrypsin activity, and blocked excess NO production without affecting the ongoing increases in BACE1/β-secretase and γ-secretase activity in fAβ25-35-treated astrocytes. Compared to fAβ25-35, sAβ25-35 also stimulated Aβ42 secretion by astrocytes and neurons and NPS 2143 specifically and wholly suppressed this effect. Therefore, since NPS 2143 thwarts any Aβ/CaSR-induced surplus secretion of endogenous Aβ42 and hence further vicious cycles of Aβ self-induction/secretion/spreading, calcilytics might effectively prevent/stop the progression to full-blown AD.
Collapse
Affiliation(s)
- Ubaldo Armato
- Department of Life and Reproduction Sciences, University of Verona Medical School, Verona, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Membrane trafficking in neuronal maintenance and degeneration. Cell Mol Life Sci 2012; 70:2919-34. [PMID: 23132096 PMCID: PMC3722462 DOI: 10.1007/s00018-012-1201-4] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Revised: 10/13/2012] [Accepted: 10/15/2012] [Indexed: 10/28/2022]
Abstract
Defects in membrane trafficking and degradation are hallmarks of most, and maybe all, neurodegenerative disorders. Such defects typically result in the accumulation of undegraded proteins due to aberrant endosomal sorting, lysosomal degradation, or autophagy. The genetic or environmental cause of a specific disease may directly affect these membrane trafficking processes. Alternatively, changes in intracellular sorting and degradation can occur as cellular responses of degenerating neurons to unrelated primary defects such as insoluble protein aggregates or other neurotoxic insults. Importantly, altered membrane trafficking may contribute to the pathogenesis or indeed protect the neuron. The observation of dramatic changes to membrane trafficking thus comes with the challenging need to distinguish pathological from protective alterations. Here, we will review our current knowledge about the protective and destructive roles of membrane trafficking in neuronal maintenance and degeneration. In particular, we will first focus on the question of what type of membrane trafficking keeps healthy neurons alive in the first place. Next, we will discuss what alterations of membrane trafficking are known to occur in Alzheimer's disease and other tauopathies, Parkinson's disease, polyQ diseases, peripheral neuropathies, and lysosomal storage disorders. Combining the maintenance and degeneration viewpoints may yield insight into how to distinguish when membrane trafficking functions protectively or contributes to degeneration.
Collapse
|
26
|
Otto GP, Nichols BJ. The roles of flotillin microdomains--endocytosis and beyond. J Cell Sci 2012; 124:3933-40. [PMID: 22194304 DOI: 10.1242/jcs.092015] [Citation(s) in RCA: 187] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Flotillins are membrane proteins that form microdomains in the plasma membrane of all mammalian cell types studied to date. They span the evolutionary spectrum, with proteins related to flotillins present in bacteria, fungi, plants and metazoans, which suggests that they perform important, and probably conserved, functions. Flotillins have been implicated in myriad processes that include endocytosis, signal transduction and regulation of the cortical cytoskeleton, yet the molecular mechanisms that underlie flotillin function in these different cases are still poorly understood. In this Commentary, we will provide an introduction to these intriguing proteins, summarise their proposed functions and discuss in greater detail some recent insights into the role of flotillin microdomains in endocytosis that have been provided by several independent studies. Finally, we will focus on the questions that are raised by these new experiments and their implications for future studies.
Collapse
Affiliation(s)
- Grant P Otto
- Medical Research Council Laboratory of Molecular Biology, Hills Road, Cambridge CB2 0QH, UK
| | | |
Collapse
|
27
|
Siegenthaler BM, Rajendran L. Retromers in Alzheimer's disease. NEURODEGENER DIS 2012; 10:116-21. [PMID: 22398391 DOI: 10.1159/000335910] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Accepted: 12/18/2011] [Indexed: 11/19/2022] Open
Abstract
Amyloid-β peptide (Aβ), the key pathogenic agent in Alzheimer's disease (AD), is released after sequential proteolytic cleavage of the transmembrane amyloid precursor protein (APP). β-Site APP-cleaving enzyme 1 (BACE1) cleaves APP in early endosomes, and the cause of increased BACE cleavage of APP in AD is not fully resolved yet. It has been proposed that perturbed intracellular trafficking of APP, which leads to prolonged residence time in early endosomes, influences Aβ production and hence the risk for AD. Retromers are a family of proteins that mediate the retrieval of transmembrane proteins from the endosomes to the trans-Golgi network. Misregulation of retromers or retromer-associated proteins influences endosomal localization of APP/BACE1. Here we review the role of retromers in the amyloidogenic processing of APP and their pathogenic role in AD.
Collapse
Affiliation(s)
- Barbara M Siegenthaler
- Systems and Cell Biology of Neurodegeneration, Division of Psychiatry Research, University of Zurich, Zurich, Switzerland
| | | |
Collapse
|
28
|
Rajendran L, Annaert W. Membrane Trafficking Pathways in Alzheimer's Disease. Traffic 2012; 13:759-70. [DOI: 10.1111/j.1600-0854.2012.01332.x] [Citation(s) in RCA: 146] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Revised: 01/20/2012] [Accepted: 01/23/2012] [Indexed: 11/28/2022]
Affiliation(s)
- Lawrence Rajendran
- Systems and Cell Biology of Neurodegeneration; Division of Psychiatry Research; University of Zurich; August-Forel Str. 1; Zurich; 8008; Switzerland
| | - Wim Annaert
- Laboratory for Membrane Trafficking; Center for Human Genetics (KULeuven) & VIB-Center for the Biology of Disease; Gasthuisberg O&N4, Herestraat 49; Leuven; B-3000; Belgium
| |
Collapse
|
29
|
Salgado IK, Serrano M, García JO, Martínez NA, Maldonado HM, Báez-Pagán CA, Lasalde-Dominicci JA, Silva WI. SorLA in glia: shared subcellular distribution patterns with caveolin-1. Cell Mol Neurobiol 2011; 32:409-21. [PMID: 22127416 DOI: 10.1007/s10571-011-9771-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Accepted: 11/08/2011] [Indexed: 01/25/2023]
Abstract
SorLA is an established sorting and trafficking protein in neurons with demonstrated relevance to Alzheimer's disease (AD). It shares these roles with the caveolins, markers of membrane rafts microdomains. To further our knowledge on sorLA's expression and traffic, we studied sorLA expression in various cultured glia and its relation to caveolin-1 (cav-1), a caveolar microdomain marker. RT-PCR and immunoblots demonstrated sorLA expression in rat C6 glioma, primary cultures of rat astrocytes (PCRA), and human astrocytoma 1321N1 cells. PCRA were determined to express the highest levels of sorLA's message. Induction of differentiation of C6 cells into an astrocyte-like phenotype led to a significant decrease in sorLA's mRNA and protein expression. A set of complementary experimental approaches establish that sorLA and cav-1 directly or indirectly interact in glia: (1) co-fractionation in light-density membrane raft fractions of rat C6 glioma, PCRA, and human 1321N1 astrocytoma cells; (2) a subcellular co-localization distribution pattern in vesicular perinuclear compartments seen via confocal imaging in C6 and PCRA; (3) additional confocal analysis in C6 cells suggesting that the perinuclear compartments correspond to their co-localization in early endosomes and the trans-Golgi; and; (4) co-immunoprecipitation data strongly supporting their direct or indirect physical interaction. These findings further establish that sorLA is expressed in glia and that it shares its subcellular distribution pattern with cav-1. A direct or indirect cav-1/sorLA interaction could modify the trafficking and sorting functions of sorLA in glia and its proposed neuroprotective role in AD.
Collapse
Affiliation(s)
- Iris K Salgado
- Department of Physiology, UPR-School of Medicine, University of Puerto Rico, San Juan, PR 00936-5067, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Zhao F, Zhang J, Liu YS, Li L, He YL. Research advances on flotillins. Virol J 2011; 8:479. [PMID: 22023811 PMCID: PMC3215287 DOI: 10.1186/1743-422x-8-479] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Accepted: 10/25/2011] [Indexed: 08/15/2023] Open
Abstract
The proteins of flotillin-1 and flotillin-2 were originally discovered in axon regeneration of goldfish retinal ganglion cells. They are generally used as marker proteins of lipid rafts and considered to be scaffolding proteins of lipid microdomains. Although they are ubiquitously expressed and well-conserved from fly to man, their exact functions remain controversial. In this review, we summarize the structure of flotillins and some functions of them, such as regulating axon regeneration, endocytosis, T cell activation, insulin signaling, membrane protein recruitment, roles in the progression of some diseases and so on.
Collapse
Affiliation(s)
- Feng Zhao
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046 Gansu, P.R. China
| | | | | | | | | |
Collapse
|
31
|
Bali J, Halima SB, Felmy B, Goodger Z, Zurbriggen S, Rajendran L. Cellular basis of Alzheimer's disease. Ann Indian Acad Neurol 2011; 13:S89-93. [PMID: 21369424 PMCID: PMC3039159 DOI: 10.4103/0972-2327.74251] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2010] [Revised: 07/25/2010] [Accepted: 09/27/2010] [Indexed: 12/20/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common form of neurodegenerative disease. A characteristic feature of the disease is the presence of amyloid-β (Aβ) which either in its soluble oligomeric form or in the plaque-associated form is causally linked to neurodegeneration. Aβ peptide is liberated from the membrane-spanning -amyloid precursor protein by sequential proteolytic processing employing β- and γ-secretases. All these proteins involved in the production of Aβ peptide are membrane associated and hence, membrane trafficking and cellular compartmentalization play important roles. In this review, we summarize the key cellular events that lead to the progression of AD.
Collapse
Affiliation(s)
- Jitin Bali
- Systems and Cell Biology of Neurodegeneration, Psychiatry Research, University of Zürich, 8008 Zurich, Switzerland
| | | | | | | | | | | |
Collapse
|
32
|
Shi C, Wu F, Xu J. H2O2 and PAF mediate Abeta1-42-induced Ca2+ dyshomeostasis that is blocked by EGb761. Neurochem Int 2010; 56:893-905. [PMID: 20362023 DOI: 10.1016/j.neuint.2010.03.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Revised: 03/05/2010] [Accepted: 03/23/2010] [Indexed: 11/27/2022]
Abstract
Calcium (Ca2+) dyshomeostasis may be of pivotal importance in mediating the neurotoxic action of amyloid beta peptide (Abeta), but the mechanism whereby Abeta disrupts Ca2+ homeostasis remains unclear. Using hippocampal neuronal cultures, the present study investigated possible mechanisms underlying Ca2+ dyshomeostasis induced by the oligomeric form of Abeta1-42 and two possible mediators of its toxicity, hydrogen peroxide (H2O2) and platelet-activating factor (PAF). It was found that, both H2O2 and PAF were able to reproduce each of the events induced by oligomeric Abeta1-42, including (a) Ca2+ influx via N-methyl-D-aspartic acid (NMDA) receptors, (b) enhancement of Ca2+ response to NMDA via activation of protein kinase C (PKC), (c) the increase of extracellular concentrations of glutamate and (d) the increase in cytosolic free Ca2+ ([Ca2+]i). Moreover, each of these events could be blocked by Ginkgo biloba extract EGb761, a free radical scavenger with PAF antagonism, and by quercetin, a constituent with well-established free radical scavenging property. In contrast, ginkgolide B, another constituent of EGb761 with well-established PAF-antagonizing activity protected the neurons against Ca2+ dyshomeostasis induced by Abeta1-42 and PAF, but not by H2O2. These results suggested the possibility that Abeta1-42-induced Ca2+ dyshomeostasis might be mediated by formation of toxic mediators such as H2O2 and PAF. Therefore, increased production of toxic mediators such as H2O2 and PAF in the brain may be critical in the pathological mechanism of neurodegenerative diseases, particularly Alzheimer's disease (AD), and may serve as major therapeutic targets for these diseases.
Collapse
Affiliation(s)
- Chun Shi
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-Sen University Guangzhou, Guangdong 510080, China.
| | | | | |
Collapse
|
33
|
Abstract
Recent reports indicate that a growing number of intracellular proteins are not only prone to pathological aggregation but can also be released and "infect" neighboring cells. Therefore, many complex diseases may obey a simple model of propagation where the penetration of seeds into hosts determines spatial spread and disease progression. We term these proteins prionoids, as they appear to infect their neighbors just like prions--but how can bulky protein aggregates be released from cells and how do they access other cells? The widespread existence of such prionoids raises unexpected issues that question our understanding of basic cell biology.
Collapse
Affiliation(s)
- Adriano Aguzzi
- Institute of Neuropathology, University Hospital of Zürich, Schmelzbergstrasse 12, CH-8091 Zürich, Switzerland.
| | | |
Collapse
|
34
|
Mechanism of amyloid plaque formation suggests an intracellular basis of Abeta pathogenicity. Proc Natl Acad Sci U S A 2010; 107:1942-7. [PMID: 20133839 DOI: 10.1073/pnas.0904532106] [Citation(s) in RCA: 225] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The formation of extracellular amyloid plaques is a common patho-biochemical event underlying several debilitating human conditions, including Alzheimer's disease (AD). Considerable evidence implies that AD damage arises primarily from small oligomeric amyloid forms of Abeta peptide, but the precise mechanism of pathogenicity remains to be established. Using a cell culture system that reproducibly leads to the formation of Alzheimer's Abeta amyloid plaques, we show here that the formation of a single amyloid plaque represents a template-dependent process that critically involves the presence of endocytosis- or phagocytosis-competent cells. Internalized Abeta peptide becomes sorted to multivesicular bodies where fibrils grow out, thus penetrating the vesicular membrane. Upon plaque formation, cells undergo cell death and intracellular amyloid structures become released into the extracellular space. These data imply a mechanism where the pathogenic activity of Abeta is attributed, at least in part, to intracellular aggregates.
Collapse
|
35
|
Rajendran L, Beckmann J, Magenau A, Boneberg EM, Gaus K, Viola A, Giebel B, Illges H. Flotillins are involved in the polarization of primitive and mature hematopoietic cells. PLoS One 2009; 4:e8290. [PMID: 20027317 PMCID: PMC2794375 DOI: 10.1371/journal.pone.0008290] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2009] [Accepted: 11/10/2009] [Indexed: 11/30/2022] Open
Abstract
Background Migration of mature and immature leukocytes in response to chemokines is not only essential during inflammation and host defense, but also during development of the hematopoietic system. Many molecules implicated in migratory polarity show uniform cellular distribution under non-activated conditions, but acquire a polarized localization upon exposure to migratory cues. Methodology/Principal Findings Here, we present evidence that raft-associated endocytic proteins (flotillins) are pre-assembled in lymphoid, myeloid and primitive hematopoietic cells and accumulate in the uropod during migration. Furthermore, flotillins display a polarized distribution during immunological synapse formation. Employing the membrane lipid-order sensitive probe Laurdan, we show that flotillin accumulation in the immunological synapse is concomittant with membrane ordering in these regions. Conclusions Together with the observation that flotillin polarization does not occur in other polarized cell types such as polarized epithelial cells, our results suggest a specific role for flotillins in hematopoietic cell polarization. Based on our results, we propose that in hematopoietic cells, flotillins provide intrinsic cues that govern segregation of certain microdomain-associated molecules during immune cell polarization.
Collapse
Affiliation(s)
- Lawrence Rajendran
- Systems and Cell Biology of Neurodegeneration, Department of Psychiatry Research, University of Zurich, Zurich, Switzerland
- Immunology, University of Konstanz, Konstanz, Germany
- Max-Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- * E-mail: (LR); (BG); (HI)
| | - Julia Beckmann
- Institute for Transplantation Diagnostics and Cellular Therapeutics, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
- Institute of Transfusionsmedicine, Transplantations Diagnostics, Universitätsklinikum Essen, Essen, Germany
| | - Astrid Magenau
- Centre for Vascular Research, University of New South Wales, Sydney, Australia
- Department of Haematology, Prince of Wales Hospital, Sydney, Australia
| | | | - Katharina Gaus
- Centre for Vascular Research, University of New South Wales, Sydney, Australia
- Department of Haematology, Prince of Wales Hospital, Sydney, Australia
| | - Antonella Viola
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Bernd Giebel
- Institute for Transplantation Diagnostics and Cellular Therapeutics, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
- Institute of Transfusionsmedicine, Transplantations Diagnostics, Universitätsklinikum Essen, Essen, Germany
- * E-mail: (LR); (BG); (HI)
| | - Harald Illges
- Immunology, University of Konstanz, Konstanz, Germany
- Biotechnologie Institut Thurgau, Taegerwilen, Switzerland
- Immunology and Cell Biology, University of Applied Sciences, Rheinbach, Germany
- * E-mail: (LR); (BG); (HI)
| |
Collapse
|
36
|
Abstract
PURPOSE OF REVIEW Lipid membrane microdomains are involved in major types of disease, ranging from vascular and metabolic diseases to neurodegeneration, autoimmunity, infectious and inflammatory diseases, and cancer. This review provides an update of membrane microdomain abnormalities. RECENT FINDINGS Lipid membrane microdomains are dynamic assemblies of sphingolipids, cholesterol and proteins that dissociate and associate rapidly and form functional clusters. Membrane microdomain clustering is the key to how membrane microdomains can form lipid-protein platforms in cell membranes, functioning in membrane trafficking, cell polarization and signalling. Clustering of membrane microdomains can be modified, for example by dietary lipids and pharmacological agents. SUMMARY Metabolic overload through a cholesterol-rich and fat-rich diet can trigger metabolic learning, which is associated with membrane microdomain persistence, persistent signalling and disturbed vesicular traffic. Detailed characterization of lipid membrane microdomains and dynamics at the molecular level is necessary and will help to identify new dietary and pharmacological therapeutic targets for the treatment and prevention of lipid membrane microdomain related diseases.
Collapse
Affiliation(s)
- Gerd Schmitz
- Institute for Clinical Chemistry and Laboratory Medicine, University of Regensburg, Regensburg, Germany.
| | | |
Collapse
|